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:

IFNγ and GM-CSF control complementary differentiation programs in the monocyte-to-phagocyte transition during neuroinflammation

Abstract

During inflammation, Ly6Chi monocytes are rapidly mobilized from the bone marrow (BM) and are recruited into inflamed tissues, where they undergo monocyte-to-phagocyte transition (MTPT). The in vivo developmental trajectories of the MTPT and the contribution of individual cytokines to this process remain unclear. Here, we used a murine model of neuroinflammation to investigate how granulocyte–macrophage colony-stimulating factor (GM-CSF) and interferon-γ (IFNγ), two type 1 cytokines, controlled MTPT. Using genetic fate mapping, gene targeting and high-dimensional single-cell multiomics analyses, we found that IFNγ was essential for the gradual acquisition of a mature inflammatory phagocyte phenotype in Ly6Chi monocytes, while GM-CSF was required to license interleukin-1β (IL-1β) production, phagocytosis and oxidative burst. These results suggest that the proinflammatory cytokine environment guided MTPT trajectories in the inflamed central nervous system (CNS) and indicated that GM-CSF was the most prominent target for the disarming of monocyte progenies during neuroinflammation.

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: Mapping tissue-invading phagocytes in the lymph node and in the CNS during EAE progression.
Fig. 2: CCR2+Ly6Chi progeny terminally differentiate into MerTK+ macrophages.
Fig. 3: Ly6Chi monocytes arise from BM and are the precursors of tissue-invading Mdcs.
Fig. 4: Ly6Chi monocytes develop into monocyte-derived macrophages in the CNS, while Cxcl10+ monocytes do not contribute to the MTPT.
Fig. 5: IFNγ, but not GM-CSF, is required for expression of key MTPT markers on Mdcs in the inflamed CNS.
Fig. 6: GM-CSF controls the transcriptional pathogenic program of monocytes and moMACs during CNS inflammation.
Fig. 7: GM-CSF modulates the effector functions in monocyte and Mdc subsets.
Fig. 8: MHCII expression is required on CCR2+ APCs during EAE induction but not in established CNS inflammation.

Similar content being viewed by others

Data availability

The scRNA-seq datasets generated in this study are available at https://doi.org/10.5281/zenodo.5722108, https://doi.org/10.5281/zenodo.5722170, https://doi.org/10.5281/zenodo.5722172 and https://doi.org/10.5281/zenodo.5711708. Source data are provided with this paper.

Code availability

Scripts to reproduce the data will be made available upon request.

References

  1. Hoeffel, G. & Ginhoux, F. Fetal monocytes and the origins of tissue-resident macrophages. Cell. Immunol. 330, 5–15 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Ginhoux, F. & Guilliams, M. Tissue-resident macrophage ontogeny and homeostasis. Immunity 44, 439–449 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Geissmann, F., Jung, S. & Littman, D. R. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 19, 71–82 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Ginhoux, F. & Jung, S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat. Rev. Immunol. 14, 392–404 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. King, I. L., Dickendesher, T. L. & Segal, B. M. Circulating Ly-6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease. Blood 113, 3190–3197 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Menezes, S. et al. The heterogeneity of Ly6Chi monocytes controls their differentiation into iNOS+ macrophages or monocyte-derived dendritic cells. Immunity 45, 1205–1218 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bain, C. C. et al. Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors. Mucosal Immunol. 6, 498–510 (2013).

  8. Zigmond, E. et al. Ly6Chi monocytes in the inflamed colon give rise to proinflammatory effector cells and migratory antigen-presenting cells. Immunity 37, 1076–1090 (2012).

  9. Mrdjen, D., Hartmann, F. J. & Becher, B. High dimensional cytometry of central nervous system leukocytes during neuroinflammation. Methods Mol. Biol. 1559, 321–332 (2017).

    Article  CAS  PubMed  Google Scholar 

  10. See, P., Lum, J., Chen, J. & Ginhoux, F. A single-cell sequencing guide for immunologists. Front. Immunol. 9, 2425 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Schaum, N. et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

    Article  PubMed Central  Google Scholar 

  12. Giladi, A. et al. Cxcl10+ monocytes define a pathogenic subset in the central nervous system during autoimmune neuroinflammation. Nat. Immunol. 21, 525–534 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Ajami, B. et al. Single-cell mass cytometry reveals distinct populations of brain myeloid cells in mouse neuroinflammation and neurodegeneration models. Nat. Neurosci. 21, 541–551 (2018).

  14. Yáñez, A. et al. Granulocyte–monocyte progenitors and monocyte-dendritic cell progenitors independently produce functionally distinct monocytes. Immunity 47, 890–902 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Weinreb, C., Rodriguez-Fraticelli, A., Camargo, F. D. & Klein, A. M. Lineage tracing on transcriptional landscapes links state to fate during differentiation. Science 367, eaaw3381 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Liu, Z. et al. Fate mapping via Ms4a3-expression history traces monocyte-derived cells. Cell 178, 1509–1525 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Croxford, A. L., Spath, S. & Becher, B. GM-CSF in neuroinflammation: licensing myeloid cells for tissue damage. Trends Immunol. 36, 651–662 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Tuzlak, S. et al. Repositioning TH cell polarization from single cytokines to complex help. Nat. Immunol. 22, 1210–1217 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Askenase, M. H. et al. Bone-marrow-resident NK cells prime monocytes for regulatory function during infection. Immunity 42, 1130–1142 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sallusto, F. & Lanzavecchia, A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor α. J. Exp. Med. 179, 1109–1118 (1994).

    Article  CAS  PubMed  Google Scholar 

  21. Abdi, K. et al. Bone marrow–derived dendritic cell cultures from RAG−/− mice include IFN-γ-producing NK cells. Immunohorizons 4, 415–419 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Croxford, A. L. et al. The cytokine GM-CSF drives the inflammatory signature of CCR2+ monocytes and licenses autoimmunity. Immunity 43, 502–514 (2015).

  23. Codarri, L., Greter, M. & Becher, B. Communication between pathogenic T cells and myeloid cells in neuroinflammatory disease. Trends Immunol. 34, 114–119 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Becher, B., Tugues, S. & Greter, M. GM-CSF: from growth factor to central mediator of tissue inflammation. Immunity 45, 963–973 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Piskurich, J. F. et al. Expression of the MHC class II transactivator (CIITA) type IV promoter in B lymphocytes and regulation by IFN-γ. Mol. Immunol. 43, 519–528 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Kageyama, Y. et al. Reduced susceptibility to collagen-induced arthritis in mice deficient in IFN-γ receptor. J. Immunol. 161, 1542–1548 (1998).

    CAS  PubMed  Google Scholar 

  27. Barin, J. G. et al. Fatal eosinophilic myocarditis develops in the absence of IFN-γ and IL-17A. J. Immunol. 191, 4038–4047 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Willenborg, D. O., Fordham, S., Bernard, C. C., Cowden, W. B. & Ramshaw, I. A. IFN-γ plays a critical down-regulatory role in the induction and effector phase of myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. J. Immunol. 157, 3223–3227 (1996).

    CAS  PubMed  Google Scholar 

  29. Ferber, I. A. et al. Mice with a disrupted IFN-γ gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). J. Immunol. 156, 5–7 (1996).

  30. Chu, C. Q., Wittmer, S. & Dalton, D. K. Failure to suppress the expansion of the activated CD4 T cell population in interferon γ-deficient mice leads to exacerbation of experimental autoimnaune encephalomyelitis. J. Exp. Med. 192, 123–128 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Komuczki, J. et al. Fate-mapping of GM-CSF expression identifies a discrete subset of inflammation-driving T helper cells regulated by cytokines IL-23 and IL-1β. Immunity 50, 1289–1304 (2019).

    Article  CAS  PubMed  Google Scholar 

  32. Mildner, A., Giladi, A., Leutz, A., Amit, I. & Jung, S. Genomic characterization of murine monocytes reveals C/EBPβ transcription factor dependence of Ly6C cells. Immunity 46, 849–862 (2017).

  33. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Greter, M. et al. GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells. Immunity 36, 1031–1046 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Croxford, A. L. & Buch, T. Cytokine reporter mice in immunological research: perspectives and lessons learned. Immunology 132, 1–8 (2010).

  36. Mendiola, A. S. et al. Transcriptional profiling and therapeutic targeting of oxidative stress in neuroinflammation. Nat. Immunol. 21, 513–524 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Mundt, S. et al. Conventional DCs sample and present myelin antigens in the healthy CNS and allow parenchymal T cell entry to initiate neuroinflammation. Sci. Immunol. 4, eaau8380 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. McQualter, J. L. et al. Granulocyte macrophage colony-stimulating factor: a new putative therapeutic target in multiple sclerosis. J. Exp. Med. 194, 873–881 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Spath, S. et al. Dysregulation of the cytokine GM-CSF induces spontaneous phagocyte invasion and immunopathology in the central nervous system. Immunity 46, 245–260 (2017).

    Article  CAS  PubMed  Google Scholar 

  40. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Locatelli, G. et al. Mononuclear phagocytes locally specify and adapt their phenotype in a multiple sclerosis model. Nat. Neurosci. 21, 1196–1208 (2018).

    Article  CAS  PubMed  Google Scholar 

  42. Helft, J. et al. GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c+MHCII+ macrophages and dendritic cells. Immunity 42, 1197–1211 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Robb, L. et al. Hematopoietic and lung abnormalities in mice with a null mutation of the common β subunit of the receptors for granulocyte–macrophage colony-stimulating factor and interleukins 3 and 5. Proc. Natl Acad. Sci. USA 92, 9565–9569 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  45. Bettelli, E. et al. Myelin oligodendrocyte glycoprotein-specific T cell receptor transgenic mice develop spontaneous autoimmune optic neuritis. J. Exp. Med. 197, 1073–1081 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Codarri, L. et al. RORγ3t drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat. Immunol. 12, 560–567 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Mair, F. et al. The end of gating? An introduction to automated analysis of high dimensional cytometry data. Eur. J. Immunol. 46, 34–43 (2016).

    Article  CAS  PubMed  Google Scholar 

  48. Brummelman, J. et al. Development, application and computational analysis of high-dimensional fluorescent antibody panels for single-cell flow cytometry. Nat. Protoc. 14, 1946–1969 (2019).

    Article  CAS  PubMed  Google Scholar 

  49. Becht, E et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat Biotechnol. https://doi.org/10.1038/nbt.4314 (2018).

  50. Spitzer, M. H. et al. IMMUNOLOGY. An interactive reference framework for modeling a dynamic immune system. Science 349, 1259425 (2015).

  51. Bastian, M., Heymann, S. & Jacomy, M. Gephi: an open source software for exploring and manipulating networks. Proceedings of the International AAAI Conference on Weblogs and Social Media 3, 361–362 (2009).

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

    Article  PubMed  PubMed Central  Google Scholar 

  53. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 38, 1408–1414 (2020).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Lutz and P. Zwicky for technical assistance. We wish to thank L. Robinson of Insight Editing London for critical review of the manuscript. This work was supported by the Swiss National Science Foundation (733 310030_170320, 310030_188450 and CRSII5_183478 to B.B. and BSSGI0_155832, 310030_184915 to M.G.), the Clinical Research Priority Program ImmunoCure (B.B.), the University of Zurich postdoctoral fellowship (D.D.F.), the Swiss Multiple Sclerosis Society (B.B. and D.D.F.) and the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme grant agreement No 882424 (B.B.).

Author information

Authors and Affiliations

Authors

Contributions

A.A., D.D.F. and E.F. designed and performed experiments, evaluated and interpreted data and wrote the manuscript. F.I. and C.D.A. performed bioinformatic analyses. S.K. and M.A. supported flow cytometry experiments and data analysis. C.A.W. performed histological analyses. Z.L. and F.G. shared the Ms4a3 fate mapping system. F.G. and M.G. interpreted data and edited the manuscript. B.B. supervised and financed the study and wrote the manuscript.

Corresponding author

Correspondence to Burkhard Becher.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Ioana Visan was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial 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 Experiment setup and pre-gating strategy for downstream analysis of the CD11b + myeloid compartment in sdLNs and CNS during EAE.

a, Schematic representation of the experimental approach and the EAE time course used for the experiment. C57BL/6 mice were actively immunized with MOG35-55 emulsified in CFA (day 0, s.c.) and pertussis toxin (days 0 and 2) to induce EAE. Cells were isolated from the sdLN and the CNS from EAE mice at the indicated time points from day 0 to day 20 p.i. and flow cytometry analysis was carried out, n = 2 day 0 and n = 5 day 220. b, Gating strategy for myeloid cells in sdLN and CNS of MOG35-55 immunized mice before dimensionality reduction and clustering. c, Representative flow cytometry plots showing the strategy to gate on Ly6Chi monocytes, moDCs and moMACs in sdLN and in the CNS at different stages of EAE development. d, Representative contour plots showing NOS2, pro-IL1β and Arginase-1 producing CD11b + and CD88 + cells in the CNS. e, Frequencies of pro-IL-1β, Arginase-1 and NOS2 among Ly6Chi monocytes, moMACs and moDCs in the CNS across the indicated time points (n = 2 for day 0 and n = 5 day 2 to 20 p.i.). f, Continuous time course of relative frequencies of pro-IL-1β + , Arginase-1+ and NOS2 + cells among CD11b + cells from sdLN and CNS, the mean value (centerline) ± s.d. (colored area) (n = 2, day 0 and n = 5, day 2-20 p.i.).

Extended Data Fig. 2 Characterization of Ccr2Ai14 and Ms4a3Ai14 targeting in the BM and blood at steady-state and during EAE.

a, Gating strategy of BM myeloid precursors and monocytes in nonimmunized and EAE from Ms4a3Ai14 and Ccr2Ai14 mice. Ccr2Ai14 mice received tamoxifen (5 mg) via oral gavage (nonimmunized: 48 h before analysis; EAE onset: at 2 and 5 d.p.i.; EAE peak: at 2, 5 and 10 d.p.i.). b, Bubble plot summarizing tdTomato+ cell frequencies per subset in BM and blood at day 0, 7 (EAE onset) and 12 p.i. (EAE peak) Ms4a3Ai14 mice (n = 4 per group). c, Bubble plots summarizing tdTomato+ cell frequencies per subset in the BM and blood at day 0, day 7 (EAE onset) and day 12 p.i. (EAE peak) Ccr2Ai14 mice (n = 4 per group). The size of the bubble indicates a mean value of relative frequencies; the bubble color shows negative log-transformed p-value. p-values are calculated using one-way ANOVA with Benjamini-Hochberg method. Data are representative of two experiments.

Extended Data Fig. 3 Characterization of Ccr2Ai14 and Ms4a3Ai14 targeting in the sdLNs and CNS at steady-state and during EAE.

a, Gating strategy of sdLN mononuclear phagocytes from N.I and EAE Ms4a3Ai14 and Ccr2Ai14 mice. sdLNs and CNS were obtained from the same Ms4a3Ai14 and Ccr2Ai14 mice and have been equally treated as in the Extended Data Fig. 2. Bubble plot summarizing tdTomato+ cell frequencies per subset and stage of disease in the sdLNs at day 0, 7 (EAE onset) and 12 p.i. (EAE peak). b, Gating strategy of CNS HSC-derived mononuclear phagocytes in Ms4a3Ai14 and Ccr2Ai14 EAE mice. Bubble plot summarizing tdTomato+ cell frequencies per subset and stages of disease, of the CNS inflammatory infiltrate at day 7 (EAE onset) and 12 p.i. (EAE peak) of Ms4a3Ai14 and Ccr2Ai14 mice (n = 4). The bubble size indicates a mean value of relative frequencies, the bubble color shows negative log-transformed p-value. p-values are calculated using one-way ANOVA with Benjamini-Hochberg method. Data are representative of two experiments.

Extended Data Fig. 4 HSC-derived phagocyte composition during steady state and EAE.

a-c, scRNA-seq analysis of HSC-derived phagocytes sorted from the BM, peripheral blood, sdLN, and the CNS of Ms4a3Ai14 mice at the day 0, 7 and 12 after EAE induction, n = 1 mice per time point. a, Gating strategy used to sort HSC-derived phagocytes. b, Relative frequencies among HSC-derived phagocytes from BM, blood, sdLN, and CNS. c, GSVA score displayed in red overlayered to the UMAP map of merged CNS at onset and peak of EAE, also shown in Fig. 4. The GSVA score was calculated using the list of genes expressed per cluster published in Giladi et. al. Each red dot corresponds to a cell expressing the same genes on Giladi et. al cluster list.

Extended Data Fig. 5 Chimerism assessment, sorting strategy, post-sequencing quality and antigen presentation score of Mdcs from the CNS of EAE BM chimeras.

a, Mixed BM chimeric mice (CD45.2 Csf2rb−/−: CD45.1 Csf2rb + /+ and CD45.2 Ifngr1−/−: CD45.1 Ifngr1 + /+) n = 6 per group were immunized with MOG35-55, 6 weeks after chimerism induction. At the peak of EAE (14 d.p.i.), CNS-infiltrating phagocytes were analyzed by high dimensional flow cytometry. The mean values ± s.d. of clinical severity are plotted against time. b, Gating strategy for CD45.1+ and CD45.2 + Ly6Chi monocytes in the CNS of Csf2rb + /+ (CD45.1+): Csf2rb−/− (CD45.2+) and Ifngr + /+(CD45.1+): Ifngr−/−(CD45.2+) BM chimeras. Remaining host cells (CD45.1 + CD45.2+) were gated out. c, The mean value ± s.d. normalized to the ratio of CD45.1+ and CD45.2+ from BM, blood, sdLN and CNS Ly6Chi monocytes of Csf2rb + /+:Csf2rb−/− and Ifngr + /+:Ifngr−/−, respectively. n = 10/group, from 4 independent experiments. d, Representative flow cytometry plots depict the gating strategy to sort CD11b + NK1.1-Ly6G- CD44hiCX3CR1int cells from the CNS of cytokine-receptor mixed BM chimeras at the peak of EAE (7 d.p.i). e, Violin plot showing number of genes, number of UMIs, mitochondria count percentage, and UMI per gene of all QC-passed cells in different genotype for all cells (top) and Mdcs (bottom). f, UMAP map of all CNS-infiltrating leukocytes, colored according to results of Seurat-guided clustering (left) or genotype (right). g, Violin plots displaying expression of selected genes across Mdcs clusters. h, Violin plot showing the scores for the antigen processing and presentation calculated as the average normalized expression of antigen processing and presentation-related genes for M1-10 clusters. For the box plot within each violin plot, middle lines indicate median values, box range from the 25th to 75th percentiles, and upper/lower whiskers extend from the hinge to the largest/smallest value no further than 1.5 times the interquartile range (IQR) from the hinge. Colored areas indicate density distribution of data. Significance was determined by one way ANOVA and Benjamini–Hochberg-corrected p-values are summarized in Supplementary Table 24. Antigen processing and presentation score projected on UMAP displaying M1-10 clusters. Antigen processing and presentation score is computed on individual genes listed in Supplementary Table 6.

Extended Data Fig. 6 GM-CSF and IFNγ dependent genes and pathways in CNS MdC subsets.

a, Correlation between integrated and non-integrated scRNA-seq dataset from WT, Csf2rb−/− and Ifngr1−/− CNS-invading mononuclear phagocyte dataset (see also Fig. 6 and Extended Data Fig. 5). Shown are the fraction of indicated scRNA-seq defined integrated clusters in each previously annotated non-integrated clusters. Each row is normalized by row sums. b, Volcano plots displaying genes that are up- (blue) or downregulated (red) in WTvsCsf2rb−/− for the indicated CNS MdC clusters. Dashed lines denote fold change thresholds used when identifying DEGs. c, Volcano plots displaying genes that are up- (blue) or downregulated (red) in WT vs Ifngr1−/− for the indicated CNS MdC clusters. Dashed lines denote fold change thresholds used when identifying DEGs. d, Gene ontology (GO) analysis of DEGs from b; e, Gene ontology (GO) analysis of DEGs from c. (See also Supplementary Table 2). Top5 GO BP terms with Benjamini-Hochberg-corrected p-values < 0.05 are shown.

Extended Data Fig. 7 Gating strategy and analysis of the CD11b + myeloid cells in CNS and sdLN of mixed BM chimeras during EAE; marker expression of Ccr2Ai14 CD11b + cells at peak EAE and targeting of Ccr2CreERT2xIabfl in sdLNs during EAE induction.

a, Gating strategy for the myeloid cells in sdLN and CNS of MOG35-55 immunized mice before dimensionality reduction and clustering. from the CNS of Csf2rb + /+ (CD45.1+): Csf2rb−/− (CD45.2+) and Ifngr1 + /+(CD45.1+): Ifngr1−/− (CD45.2+) BM chimeras. b, Median expression of the markers selected to define MdC populations, overlaid to the UMAP of CNS Mdcs from Ccr2Ai14 mice at peak EAE (14 d.p.i.). c, Representative plots are shown for: CD45 + Ly6G-CD11b + Ly6C+moDCs +and CD45 + Ly6G-Ly6C-CD11b + CD11c + CD26 + XCR1 + cDCs. Data represents 2 independent experiments (n = 5 per group), p values were calculated using an unpaired two-tailed t test with Welch’s correction. **p < 0.0079. Data are shown as means ± SEM. n.s., not significant.

Supplementary information

Source data

Source Data Fig. 1

Individual data points, statistical source data.

Source Data Fig. 2

Individual data points, statistical source data.

Source Data Fig. 6

Individual data points, statistical source data.

Source Data Fig. 7

Individual data points, statistical source data.

Source Data Fig. 8

Individual data points, statistical source data.

Source Data Extended Fig. 1

Individual data points, statistical source data.

Source Data Extended Fig. 2

Individual data points, statistical source data.

Source Data Extended Fig. 3

Individual data points, statistical source data.

Source Data Extended Fig. 5

Individual data points, statistical source data.

Source Data Extended Fig. 7

Individual data points, statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Amorim, A., De Feo, D., Friebel, E. et al. IFNγ and GM-CSF control complementary differentiation programs in the monocyte-to-phagocyte transition during neuroinflammation. Nat Immunol 23, 217–228 (2022). https://doi.org/10.1038/s41590-021-01117-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01117-7

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