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:

Origin, fate and dynamics of macrophages at central nervous system interfaces

Abstract

Perivascular, subdural meningeal and choroid plexus macrophages are non-parenchymal macrophages that mediate immune responses at brain boundaries. Although the origin of parenchymal microglia has recently been elucidated, much less is known about the precursors, the underlying transcriptional program and the dynamics of the other macrophages in the central nervous system (CNS). It was assumed that they have a high turnover from blood-borne monocytes. However, using parabiosis and fate-mapping approaches in mice, we found that CNS macrophages arose from hematopoietic precursors during embryonic development and established stable populations, with the notable exception of choroid plexus macrophages, which had dual origins and a shorter life span. The generation of CNS macrophages relied on the transcription factor PU.1, whereas the MYB, BATF3 and NR4A1 transcription factors were not required.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Molecular census of non-parenchymal macrophages, microglia and monocytes.
Figure 2: Development of CNS macrophages.
Figure 3: Ontogeny of brain macrophages at brain interfaces.
Figure 4: Maintenance of non-parenchymal macrophages in adulthood.
Figure 5: In vivo dynamics of CNS macrophages.
Figure 6: Turnover of CNS macrophages.

Similar content being viewed by others

References

  1. Ransohoff, R.M. & Cardona, A.E. The myeloid cells of the central nervous system parenchyma. Nature 468, 253–262 (2010).

    CAS  PubMed  Google Scholar 

  2. Prinz, M., Priller, J., Sisodia, S.S. & Ransohoff, R.M. Heterogeneity of CNS myeloid cells and their roles in neurodegeneration. Nat. Neurosci. 14, 1227–1235 (2011).

    CAS  PubMed  Google Scholar 

  3. Prinz, M. & Priller, J. Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease. Nat. Rev. Neurosci. 15, 300–312 (2014).

    CAS  PubMed  Google Scholar 

  4. Alliot, F., Godin, I. & Pessac, B. Microglia derive from progenitors, originating from the yolk sac, and which proliferate in the brain. Brain Res. Dev. Brain Res. 117, 145–152 (1999).

    CAS  PubMed  Google Scholar 

  5. Ginhoux, F. et al. Fate-mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330, 841–845 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336, 86–90 (2012).

    CAS  PubMed  Google Scholar 

  7. Kierdorf, K. et al. Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nat. Neurosci. 16, 273–280 (2013).

    CAS  PubMed  Google Scholar 

  8. Aguzzi, A., Barres, B.A. & Bennett, M.L. Microglia: scapegoat, saboteur, or something else? Science 339, 156–161 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Hickey, W.F. & Kimura, H. Perivascular microglial cells of the CNS are bone marrow-derived and present antigen in vivo. Science 239, 290–292 (1988).

    CAS  PubMed  Google Scholar 

  10. Hickey, W.F., Vass, K. & Lassmann, H. Bone marrow–derived elements in the central nervous system: an immunohistochemical and ultrastructural survey of rat chimeras. J. Neuropathol. Exp. Neurol. 51, 246–256 (1992).

    CAS  PubMed  Google Scholar 

  11. Yang, Y. et al. Perivascular, but not parenchymal, cerebral engraftment of donor cells after non-myeloablative bone marrow transplantation. Exp. Mol. Pathol. 95, 7–17 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Barr, C.M., Manning, J., Lewis, C.A., Rossi, F.M. & Krieger, C. Submyeloablative conditioning with busulfan permits bone marrow–derived cell accumulation in a murine model of Alzheimer's disease. Neurosci. Lett. 588, 196–201 (2015).

    CAS  PubMed  Google Scholar 

  13. Mildner, A. et al. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat. Neurosci. 10, 1544–1553 (2007).

    CAS  PubMed  Google Scholar 

  14. Mildner, A. et al. Distinct and non-redundant roles of microglia and myeloid subsets in mouse models of Alzheimer's disease. J. Neurosci. 31, 11159–11171 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Kierdorf, K., Katzmarski, N., Haas, C.A. & Prinz, M. Bone marrow cell recruitment to the brain in the absence of irradiation or parabiosis bias. PLoS One 8, e58544 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Galea, I. et al. Mannose receptor expression specifically reveals perivascular macrophages in normal, injured and diseased mouse brain. Glia 49, 375–384 (2005).

    PubMed  Google Scholar 

  17. Kim, J.V., Kang, S.S., Dustin, M.L. & McGavern, D.B. Myelomonocytic cell recruitment causes fatal CNS vascular injury during acute viral meningitis. Nature 457, 191–195 (2009).

    CAS  PubMed  Google Scholar 

  18. Zeisel, A. et al. Brain structure. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science 347, 1138–1142 (2015).

    CAS  PubMed  Google Scholar 

  19. Jamieson, A.R. et al. Exploring nonlinear feature space dimension reduction and data representation in breast Cadx with Laplacian eigenmaps and t-SNE. Med. Phys. 37, 339–351 (2010).

    PubMed  Google Scholar 

  20. Bechmann, I. et al. Turnover of rat brain perivascular cells. Exp. Neurol. 168, 242–249 (2001).

    CAS  PubMed  Google Scholar 

  21. Priller, J. et al. Targeting gene-modified hematopoietic cells to the central nervous system: use of green fluorescent protein uncovers microglial engraftment. Nat. Med. 7, 1356–1361 (2001).

    CAS  PubMed  Google Scholar 

  22. Gomez Perdiguero, E. et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518, 547–551 (2015).

    PubMed  Google Scholar 

  23. Molawi, K. et al. Progressive replacement of embryo-derived cardiac macrophages with age. J. Exp. Med. 211, 2151–2158 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Ensan, S. et al. Self-renewing resident arterial macrophages arise from embryonic CX3CR1 precursors and circulating monocytes immediately after birth. Nat. Immunol. 17, 159–168 (2015).

    PubMed  Google Scholar 

  25. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

    CAS  PubMed  Google Scholar 

  26. Goldmann, T. et al. A new type of microglia gene targeting shows TAK1 to be pivotal in CNS autoimmune inflammation. Nat. Neurosci. 16, 1618–1626 (2013).

    CAS  PubMed  Google Scholar 

  27. Rosenbauer, F. & Tenen, D.G. Transcription factors in myeloid development: balancing differentiation with transformation. Nat. Rev. Immunol. 7, 105–117 (2007).

    CAS  PubMed  Google Scholar 

  28. Geissmann, F. et al. Development of monocytes, macrophages, and dendritic cells. Science 327, 656–661 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Ajami, B., Bennett, J.L., Krieger, C., Tetzlaff, W. & Rossi, F.M. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat. Neurosci. 10, 1538–1543 (2007).

    CAS  PubMed  Google Scholar 

  30. Wieghofer, P., Knobeloch, K.P. & Prinz, M. Genetic targeting of microglia. Glia 63, 1–22 (2015).

    PubMed  Google Scholar 

  31. Nimmerjahn, A., Kirchhoff, F. & Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 308, 1314–1318 (2005).

    CAS  PubMed  Google Scholar 

  32. Davalos, D. et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 8, 752–758 (2005).

    CAS  PubMed  Google Scholar 

  33. Ajami, B., Bennett, J.L., Krieger, C., McNagny, K.M. & Rossi, F.M. Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat. Neurosci. 14, 1142–1149 (2011).

    CAS  PubMed  Google Scholar 

  34. Butovsky, O. et al. Identification of a unique TGF-β–dependent molecular and functional signature in microglia. Nat. Neurosci. 17, 131–143 (2014).

    CAS  PubMed  Google Scholar 

  35. Hickman, S.E. et al. The microglial sensome revealed by direct RNA sequencing. Nat. Neurosci. 16, 1896–1905 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  37. Chorro, L. et al. Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network. J. Exp. Med. 206, 3089–3100 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Guilliams, M. et al. Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J. Exp. Med. 210, 1977–1992 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Epelman, S. et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity 40, 91–104 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Sheng, J., Ruedl, C. & Karjalainen, K. Most tissue-resident macrophages except microglia are derived from fetal hematopoietic stem cells. Immunity 43, 382–393 (2015).

    CAS  PubMed  Google Scholar 

  41. Adolfsson, J. et al. Upregulation of Flt3 expression within the bone marrow Lin(−)Sca1(+)c-kit(+) stem cell compartment is accompanied by loss of self-renewal capacity. Immunity 15, 659–669 (2001).

    CAS  PubMed  Google Scholar 

  42. Epelman, S., Lavine, K.J. & Randolph, G.J. Origin and functions of tissue macrophages. Immunity 41, 21–35 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Kunis, G. et al. IFN-γ-dependent activation of the brain's choroid plexus for CNS immune surveillance and repair. Brain 136, 3427–3440 (2013).

    PubMed  Google Scholar 

  44. Engelhardt, B., Wolburg-Buchholz, K. & Wolburg, H. Involvement of the choroid plexus in central nervous system inflammation. Microsc. Res. Tech. 52, 112–129 (2001).

    CAS  PubMed  Google Scholar 

  45. Axtell, R.C. & Steinman, L. Gaining entry to an uninflamed brain. Nat. Immunol. 10, 453–455 (2009).

    CAS  PubMed  Google Scholar 

  46. Kivisäkk, P. et al. Localizing central nervous system immune surveillance: meningeal antigen-presenting cells activate T cells during experimental autoimmune encephalomyelitis. Ann. Neurol. 65, 457–469 (2009).

    PubMed  PubMed Central  Google Scholar 

  47. Anandasabapathy, N. et al. Flt3L controls the development of radiosensitive dendritic cells in the meninges and choroid plexus of the steady-state mouse brain. J. Exp. Med. 208, 1695–1705 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Brendecke, S.M. & Prinz, M. Do not judge a cell by its cover—diversity of CNS resident, adjoining and infiltrating myeloid cells in inflammation. Semin. Immunopathol. 37, 591–605 (2015).

    CAS  PubMed  Google Scholar 

  49. Misgeld, T., Nikic, I. & Kerschensteiner, M. In vivo imaging of single axons in the mouse spinal cord. Nat. Protoc. 2, 263–268 (2007).

    CAS  PubMed  Google Scholar 

  50. Romanelli, E. et al. Cellular, subcellular and functional in vivo labeling of the spinal cord using vital dyes. Nat. Protoc. 8, 481–490 (2013).

    CAS  PubMed  Google Scholar 

  51. Goldmann, T. et al. USP18 lack in microglia causes destructive interferonopathy of the mouse brain. EMBO J. 34, 1612–1629 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Oberle, M. Ditter and T. el Gaz for excellent technical assistance and T. Leng Tay for critical reading of the manuscript. Supported by the DFG (SFB 992, SFB 1160, PR 577/8-1, Reinhart Koselleck Grant for M.P.; SFB 1160, ZE872/3-1 for R.Z.; and FOR1336 for J.P., I.B., M.P. and S.J.), the Fritz-Thyssen Foundation (M.P.), the European Union's Seventh Framework Program FP7 under Grant agreement 607962 (nEUROinflammation for M.P.), the Gemeinnützige Hertie Foundation (GHST for M.P.), the Sobek Foundation (M.P.) and the BMBF-funded Competence Network on Multiple Sclerosis (KKNMS for M.P. and M. Kerschensteiner).

Author information

Authors and Affiliations

Authors

Contributions

T.G., P.W., M.J.C.J., F.P., N.H., K.F., O.S., K.K., L.A., M. Krueger, G.L. and H.H. conducted the experiments and analyzed the data. R.Z., S.E., F.G., J.P., F.M.V.R., I.B., S.L., M. Kerschensteiner and S.J. analyzed the data, contributed to the in vivo studies and provided mice or reagents. T.G. and M.P. supervised the project and wrote the manuscript.

Corresponding author

Correspondence to Marco Prinz.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Single-cell RNA sequencing of pvMΦ, microglia and monocytes.

(a) Work flow for obtaining and analyzing single-cell RNA-seq from mouse cortical cells and monocytes from dissection to single-cell RNA-seq and unbiased biclustering.

(b) Representative image of single cells captured in the chip. All cells were checked individually by light microscopy and chambers with no or damaged cells (red squares) were omitted from subsequent analysis.

(c-f) Bar graphs for commonly expressed genes (c), markers selectively expressed by pvMΦ (d), selectively expressed by cortical microglia (e) or just by monocytes (f) evaluated by single cell RNA-seq.

Picture from cell capture for RNAseq is representative of one independent experiment (b). Single cell RNAseq data (c-e) is representative of two independent experiments with 167 ptMΦ, 246 monocytes, 33 microglia and 65 pvMΦ. Data is represented as mean ± s.e.m.

Supplementary Figure 2 Flow cytometry of cortical pvMΦ and microglia.

(a) Representative gating strategy for the flow cytometry-based isolation of bone marrow monocytes for subsequent single-cell RNA-sequencing as shown in Fig. 1d-g.

(b) Representative gating strategy for the isolation of cortical CD11b+ CD45lo microglia and CD11b+ CD45hi cells from the cortex (meninges and choroid plexus were removed) as shown in Fig. 1j-k.

(c) CD11b+ CD45hi pvMΦ can be further separated from CD11b+CD45lo microglia by gating them as CD45hi CD11b+ Ly6C Ly6G CD206+ F4/80+ cells. Representative histograms for CD206, CD36 and F4/80 staining on pvMΦ and microglia (black line) are shown over isotype controls (filled gray).

d) Representative gating strategy for the flow cytometry-based characterization of Ly6C cells as shown in Fig. 6a-b. Peripheral blood leukocytes were gated according to physical parameters and further subdivided in myeloid cells by the expression of CD45 and CD11b. CD45+ CD11b+ SSClo CD115+ monocytes are then further discriminated into Ly6Chi inflammatory monocytes and Ly6Clo patrolling monocytes.

Gating strategies are representative of six mice from two independent experiments (a, d) or from three biological replicates from two independent experiments (b, c).

Supplementary Figure 3 Irradiation induces engraftment of bone-marrow-derived CNS macrophages and microglia.

a) Scheme for the induction of recombination (injection of tamoxifen [TAM]) and subsequent analysis in Cx3cr1CreERRosa26-YFP animals.

b) Scheme and timeline for labelling and analyses of pvMΦ, mMΦ and cpMΦ in adulthood using TAM injection in adult Cx3cr1CreERRosa26-YFP animals.

c) Direct fluorescence microscopic visualization revealed numerous GFP+ donor-derived Iba-1+ pvMΦ, mMΦ, cpMΦ and few microglia 20 weeks after transfer of bone marrow from Acta1-GFP mice into lethally irradiated wild-type mice. Arrows indicate double positive, asterisks single Iba-1 (red) positive cells. Scale bar = 25 μm.

d) Quantification of donor-derived GFP+ Iba-1+ cells.

Immunofluorescence pictures (c) are representative of four mice from one independent experiment. Data were obtained from five mice per group from one independent experiment (d). Each symbol represents one mouse and three tissue sections per mouse were quantified. (means ± s.e.m.)

Supplementary Figure 4 CNS macrophages do not require Batf3.

a) Localization and presence of pvMΦ, mMΦ and cpMΦ in adult wild-type (WT) and Batf3−/− mice evaluated using Iba-1 immunohistochemistry. Representative figure are presented (upper images) and quantification thereof.

b) Confocal pictures showing Tomato+ cells in the choroid plexus of Cx3cr1CreERRosa26-Tomato mice (tomato, red) at 8 weeks after TAM expressing the macrophage marker F4/80 and to a much lesser extent CD206. Scale bar: 25 μm.

Immunofluorescence pictures are representative of of five mice from two independent experiments (b). Immunohistochemistry pictures (a) are representative of three mice per genotype from one independent experiment. Macrophage density (a) is representative of three mice per genotype from one independent experiment (Meninges: P= 0.7; perivascular space: P=0.7; choroid plexus P= 1). Mann Whitney test was applied. Each symbol represents one mouse with quantification of a minimum of three tissue sections. (error bars, s.e.m.). N.S. = not significant.

Supplementary Figure 5 Model for experimental findings on the origin, fate and dynamics of microglia and macrophages at CNS boundaries.

Traditional and proposed view on the origin, fate and turnover of non-parenchymal CNS macrophages and microglia.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 (PDF 1295 kb)

Location and morphology of pvMΦ, mMΦ and microglial cells.

3D-rendering (Imaris, Bitplane) of the confocal image stack represented in Fig. 5b that illustrates the distinct pvMΦ, mMΦ and microglial cells in the intact spinal cord of a Cx3cr1CreER:Rosa26-Tomato mice (tomato, red) at 8 weeks after TAM and injected with dextran-AF647 (blood vessel, blue). (MP4 8147 kb)

Dynamics of the myeloid cells.

Confocal projection of the dorsal spinal cord as shown in Fig. 5c and 5d followed by in vivo 2-photon time-lapse imaging of the different myeloid cells in Cx3cr1CreER:Rosa26-Tomato mice at 8 weeks after TAM. (MP4 979 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Goldmann, T., Wieghofer, P., Jordão, M. et al. Origin, fate and dynamics of macrophages at central nervous system interfaces. Nat Immunol 17, 797–805 (2016). https://doi.org/10.1038/ni.3423

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3423

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