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:

A new fate mapping system reveals context-dependent random or clonal expansion of microglia

Abstract

Microglia constitute a highly specialized network of tissue-resident immune cells that is important for the control of tissue homeostasis and the resolution of diseases of the CNS. Little is known about how their spatial distribution is established and maintained in vivo. Here we establish a new multicolor fluorescence fate mapping system to monitor microglial dynamics during steady state and disease. Our findings suggest that microglia establish a dense network with regional differences, and the high regional turnover rates found challenge the universal concept of microglial longevity. Microglial self-renewal under steady state conditions constitutes a stochastic process. During pathology this randomness shifts to selected clonal microglial expansion. In the resolution phase, excess disease-associated microglia are removed by a dual mechanism of cell egress and apoptosis to re-establish the stable microglial network. This study unravels the dynamic yet discrete self-organization of mature microglia in the healthy and diseased CNS.

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

Figure 1: Characterization of the Cx3cr1creERR26RConfetti ('Microfetti') mouse model for fate mapping studies in microglia.
Figure 2: The microglial network is largely stable during homeostasis.
Figure 3: Regional difference of random microglial self-renewal.
Figure 4: Detection of microglial proliferation in steady state and disease is independent of Confetti label.
Figure 5: Clonal expansion of microglia upon injury revealed by spatiotemporal cluster analysis.
Figure 6: Reestablishment of the microglial network is achieved by egress and cell death during resolution of neurodegeneration.
Figure 7: Resolution of excess microglia is accompanied by increased regulation of cell migration, interferon response and lipid mediation.

Similar content being viewed by others

Accession codes

Primary accessions

European Nucleotide Archive

Gene Expression Omnibus

References

  1. Hanisch, U.K. & Kettenmann, H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 10, 1387–1394 (2007).

    CAS  PubMed  Google Scholar 

  2. 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 

  3. Ransohoff, R.M. & Perry, V.H. Microglial physiology: unique stimuli, specialized responses. Annu. Rev. Immunol. 27, 119–145 (2009).

    CAS  PubMed  Google Scholar 

  4. 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 

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

    PubMed  Google Scholar 

  6. Hashimoto, D. et al. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38, 792–804 (2013).

    CAS  PubMed  Google Scholar 

  7. Hagemeyer, N. et al. Transcriptome-based profiling of yolk sac-derived macrophages reveals a role for Irf8 in macrophage maturation. EMBO J. 35, 1730–1744 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. 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 

  9. 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 

  10. 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 

  11. Goldmann, T. et al. Origin, fate and dynamics of macrophages at central nervous system interfaces. Nat. Immunol. 17, 797–805 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Grabert, K. et al. Microglial brain region-dependent diversity and selective regional sensitivities to aging. Nat. Neurosci. 19, 504–516 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Shemer, A., Erny, D., Jung, S. & Prinz, M. Microglia plasticity during health and disease: an immunological perspective. Trends Immunol. 36, 614–624 (2015).

    CAS  PubMed  Google Scholar 

  14. Bruttger, J. et al. Genetic cell ablation reveals clusters of local self-renewing microglia in the mammalian central nervous system. Immunity 43, 92–106 (2015).

    CAS  PubMed  Google Scholar 

  15. Elmore, M.R.P. et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 82, 380–397 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Sieweke, M.H. & Allen, J.E. Beyond stem cells: self-renewal of differentiated macrophages. Science 342, 1242974 (2013).

    PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  18. 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 

  19. 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 

  20. 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 

  21. 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 

  22. Lawson, L.J., Perry, V.H., Dri, P. & Gordon, S. Heterogeneity in the distribution and morphology of microglia in the normal adult mouse brain. Neuroscience 39, 151–170 (1990).

    CAS  PubMed  Google Scholar 

  23. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Parkhurst, C.N. et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. 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 

  26. 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 

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

    PubMed  Google Scholar 

  28. Lawson, L.J., Perry, V.H. & Gordon, S. Turnover of resident microglia in the normal adult mouse brain. Neuroscience 48, 405–415 (1992).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  30. Gómez-Nicola, D., Fransen, N.L., Suzzi, S. & Perry, V.H. Regulation of microglial proliferation during chronic neurodegeneration. J. Neurosci. 33, 2481–2493 (2013).

    PubMed  PubMed Central  Google Scholar 

  31. Olmos-Alonso, A. et al. Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer's-like pathology. Brain 139, 891–907 (2016).

    PubMed  PubMed Central  Google Scholar 

  32. Snippert, H.J. et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144 (2010).

    CAS  PubMed  Google Scholar 

  33. Wuidart, A. et al. Quantitative lineage tracing strategies to resolve multipotency in tissue-specific stem cells. Genes Dev. 30, 1261–1277 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Ghigo, C. et al. Multicolor fate mapping of Langerhans cell homeostasis. J. Exp. Med. 210, 1657–1664 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Gosselin, D. et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 159, 1327–1340 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Xu, J. et al. Temporal-spatial resolution fate mapping reveals distinct origins for embryonic and adult microglia in zebrafish. Dev. Cell 34, 632–641 (2015).

    CAS  PubMed  Google Scholar 

  39. Gallego, B.I. et al. IOP induces upregulation of GFAP and MHC-II and microglia reactivity in mice retina contralateral to experimental glaucoma. J. Neuroinflammation 9, 92 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Cen, L.P. et al. Bilateral retinal microglial response to unilateral optic nerve transection in rats. Neuroscience 311, 56–66 (2015).

    CAS  PubMed  Google Scholar 

  41. Bellingan, G.J., Caldwell, H., Howie, S.E., Dransfield, I. & Haslett, C. In vivo fate of the inflammatory macrophage during the resolution of inflammation: inflammatory macrophages do not die locally, but emigrate to the draining lymph nodes. J. Immunol. 157, 2577–2585 (1996).

    CAS  PubMed  Google Scholar 

  42. Gautier, E.L., Ivanov, S., Lesnik, P. & Randolph, G.J. Local apoptosis mediates clearance of macrophages from resolving inflammation in mice. Blood 122, 2714–2722 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Schwab, J.M., Chiang, N., Arita, M. & Serhan, C.N. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature 447, 869–874 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Drew, P.J. et al. Chronic optical access through a polished and reinforced thinned skull. Nat. Methods 7, 981–984 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Preibisch, S., Saalfeld, S. & Tomancak, P. Globally optimal stitching of tiled 3D microscopic image acquisitions. Bioinformatics 25, 1463–1465 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Shankaran, M. et al. Measurement of brain microglial proliferation rates in vivo in response to neuroinflammatory stimuli: application to drug discovery. J. Neurosci. Res. 85, 2374–2384 (2007).

    CAS  PubMed  Google Scholar 

  47. Salic, A. & Mitchison, T.J. A chemical method for fast and sensitive detection of DNA synthesis in vivo. Proc. Natl. Acad. Sci. USA 105, 2415–2420 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Emmenlauer, M. et al. XuvTools: free, fast and reliable stitching of large 3D datasets. J. Microsc. 233, 42–60 (2009).

    CAS  PubMed  Google Scholar 

  49. Kim, D., Langmead, B. & Salzberg, S.L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Liao, Y., Smyth, G.K. & Shi, W. The Subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 41, e108 (2013).

    PubMed  PubMed Central  Google Scholar 

  51. Love, M.I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  52. Alexa, A. & Rahnenfuhrer, J. TopGO: enrichment analysis for gene ontology. R package version 2.26.0 (2016).

Download references

Acknowledgements

The authors thank H. Snippert (University Medical Center Utrecht) and H. Clevers (Hubrecht Institute) for the R26RConfetti mouse line, M. Follo and team at Lighthouse Fluorescence Technologies Core Facility, T. Blank for critical feedback, J. Bührer for technical support, and CEMT, University of Freiburg for excellent animal care. T.L.T. is supported by the German Research Foundation (DFG, TA1029/1-1) and Ministry of Science, Research and the Arts, Baden-Wuerttemberg (Research Seed Capital). M.P. is supported by the BMBF Competence Network of Multiple Sclerosis (KKNMS), the Sobek-Stiftung and the DFG (SFB992, SFB1140, Reinhart-Koselleck grant). F.F.-K., J.A.P., S.J., J.P. and M.P. are supported by the SFB/TRR167 “NeuroMac”.

Author information

Authors and Affiliations

Authors

Contributions

T.L.T. and M.P. conceived the study. T.L.T. established the methods. D.M., O.R. and T.L.T. designed the software and validated imaging data. T.L.T., J.D., F.F.-K., G.L., S., D.G., M.D., A.D., T.S., A.A.-F., O.S., A.M., A.S. and S.J. performed experiments and/or analyses. T.L.T., L.M.S., J.A.P., D.G., J.P., O.R. and M.P. provided supervision. T.L.T., D.M. and M.P. wrote the manuscript. All authors contributed to the editing of the paper.

Corresponding authors

Correspondence to Tuan Leng Tay or Marco Prinz.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterization of the Microfetti mouse model.

(a) Gating strategy for 8-color flow analysis of peripheral Ly-6C+ monocytes from Microfetti mice 5-7 days after TAM treatment. Living cells in the forward and side scatter were gated on CD45+CD11b+, followed by CD115+ to separate monocytes from granulocytes. Monocytes were distinguished by Ly-6C expression levels. Representative histograms of Confetti (GFP, YFP, RFP and CFP) expression in Microfetti Ly-6Clo monocytes (left). Confetti signals were gated on Confetti-negative WT counterparts. Expression of Confetti reporter in Ly-6Chi (open circle) and Ly-6Clo (colored) monocytes (right). Each symbol represents one mouse. Animals with Confetti-labeled Ly-6Clo monocytes were used for tracking long-term microglial self-renewal during homeostasis (n = 21 mice; data shown as total Confetti expression in Fig. 2b, 1 dead animal excluded).

(b) Detection of microglial division in vivo. Three Microfetti mice were examined by two-photon laser scanning microscopy via a cortical window six weeks after TAM treatment (see Supplemental Experimental Procedures). A representative image shows RFP+ microglial cells (arrowheads) emerged two days after targeted laser lesion (asterisk). One RFP+ microglial cell (arrow) was no longer detected in the same 200 μm depth volume on Day 2. Scale bar, 50 μm (same scale in both panels).

(c) Quantification of change in area of coverage by GFP+ and RFP+ microglia per minute of in vivo imaging over 30 minutes from Cx3cr1GFP/+ and Microfetti mice, respectively. Each symbol represents one microglial cell. Mann Whitney U-test, P = 0.64. ns, not significant. Data are represented as mean ± SEM. n = 3 mice per genotype.

(d) Representative maximum intensity projection images for c showing two consecutive time points, t1 (red) and t2 (green), with 1 minute interval. Static structures are revealed in the overlay (yellow). Scale bar, 30 μm (same scale in both panels). See also Supplementary Videos 1 and 2.

Supplementary Figure 2 Differential regional stability of the microglial network during steady state.

(a) Representative 2D renderings of Confetti+ (green, yellow, red, and cyan colored balls) and Iba-1 immunolabeled (magenta balls) microglia from 636 μm x 636 μm x 50 μm confocal volumes of the cortex, hippocampus and cerebellum over 1-36 weeks after TAM treatment of 8 weeks old female Microfetti mice.

(b) Confetti+ microglial densities in different brain regions over 36 weeks. Microglial density (y-axis) was obtained from dividing same color cell counts by ring volume of each central ring size (x-axis). Cell densities from recorded data (red line) were plotted against densities derived from 10,000 Monte Carlo (MC) simulation of random distribution of Confetti+ microglia within each dataset (blue box plots represent simulated density distributions: central mark is the median, box represents the 25th to 75th percentiles, whiskers extend up to the 98th percentile and data points above were regarded as outliers). Measured values (red line) that appear above the 98th percentile of MC simulations (blue whiskers), for example at 8 weeks after TAM treatment in the hippocampus and cerebellum where r = 40 μm, indicate the non-random appearance of same color Confetti+ microglial cells and imply that proliferation occurred. This peak represents a single cell replication event in an arbitrary volume of 0.001 mm3. Mean data are shown. n = 5 mice per time point.

(c) Quantitative analysis of randomness of microglial cell proliferation events detected in b. A pair of same color Confetti+ microglial cells with ≤50 μm spatial distance was considered a single cell replication event. Distribution of standard deviation (SD) of a MC simulation (10,000 iterations) of locations of proliferation events derived from a uniform distribution are represented as box plots (blue box plots: central mark is the median, box represents the 25th to 75th percentiles, whiskers represent the 2nd and 98th percentiles, and outliers are plotted in red + signs). Average SD of the distribution of observed replication events to a perfect uniform distribution (black cross) was compared to MC simulation. Where the observed SD falls within the MC range between the whiskers, the locations of proliferation events can be considered randomly distributed.

Supplementary Figure 3 Spatiotemporal analysis of microglial expansion and redistribution in the injured facial nucleus.

(a) Coronal brain section of a Cx3cr1GFP/+ mouse 7d post facial nerve lesion depicts areas analyzed for kinetics of microglial proliferation in the facial nucleus degeneration model. Microglial cells were marked by GFP (green). Neurons were immunolabeled with NeuN (blue) to locate the facial nucleus in the pons. The facial nucleus, a 200 μm broad perinuclear zone, and the parvicellular reticular nucleus, alpha part (PCRtA) were isolated in equal dimensions on lesion and contralateral sides for quantification of GFP+ microglia using the Imaris software.

(b) Quantification of GFP+ microglial cells in contralateral (black) and injured (red) facial nuclei from onset (1 to 2d) to peak (7 and 14d) to resolution of injury (60d) in 8-14 weeks old female Cx3cr1GFP/+ mice.

(c) Representative confocal images of microglia (GFP, green) with DAPI nuclear counterstain (blue) in matched facial nuclei during the time course of injury.

(d) Kinetics of microglial proliferation indicated by Ki67 immunohistochemistry.

(e) Representative confocal images for d. Microglia took on less ramified and more amoeboid morphology at 2 d after the lesion. GFP+ microglia (green), Ki67 (magenta) and DAPI (blue).

(f) Kinetics of microglial activation indicated by MHC class II immunohistochemistry.

(g) Representative confocal images for f. Microglia were highly activated and assumed a rod-like or amoeboid morphology 7d following nerve transection. GFP+ microglia (green), MHC class II (magenta) and DAPI (blue).

Data are represented as mean ± SEM in b, d and f. n = 4 (1 d), 5 (2 d), 6 (7 d), 5 (14 d) and 7 (60 d) mice pooled from five experiments, 3-6 sections each. Two-way ANOVA, * P = 0.0358 in b, ** P = 0.0036 in b, 0.00110 in f, *** P < 0.001 in b, d and f. Scale bars, 50 μm in c, 20 μm in e and g.

Supplementary Figure 4 Distribution of EdU+ GFP+ microglial cells outside of facial nuclei at 7 and 60 d after lesion.

(a) Quantification of EdU+ GFP+ microglial cells external of the facial nuclei (in female Cx3cr1GFP/+ mice injured at 8-14 weeks old) at peak of microgliosis (7 d) and recovery (60 d). Each black and red symbol represents one paired count. Data are pooled from two experiments and shown as mean ± SEM. n = 8 (7 d) and 9 (60 d) mice. Two-way ANOVA, time point *** P = 0.001, lesion *** P < 0.0001.

(b) Representative overview images for a showing lesion and contralateral pons. Dotted lines demarcate the facial nuclei. High magnification insets show EdU+ GFP+ microglial cells in the intermediate reticular nucleus. Scale bars, 500 μm, 50 μm in inset.

Supplementary Figure 5 Graphical abstract of context-dependent microglial renewal and clonal expansion.

Microglial renewal occurs randomly and steadily throughout the healthy CNS with regional differences in rate. The onset of neurodegeneration triggers rapid clonal expansion and activation of microglial cells. During recovery the altered microglial network gradually returns to homeostatic cell density and a non-activated state. Excess microglia associated with disease are eliminated via cell egress and local apoptotic cell death to regain steady state microglial interfaces in the CNS.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 and Supplementary Tables 1–3. (PDF 1716 kb)

Supplementary Methods Checklist (PDF 567 kb)

Supplementary Software

Supplementary Software. (ZIP 10418 kb)

Supplementary Video 1

Thirty-minute time lapse recording of steady state GFP+ adult microglial cells in a CX3CR1GFP/wt mouse via a cranial window. (MOV 562 kb)

Supplementary Video 2

Thirty-minute time lapse recording of a steady state RFP+ adult microglial cell in a Microfetti mouse via a cranial window. (MOV 338 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tay, T., Mai, D., Dautzenberg, J. et al. A new fate mapping system reveals context-dependent random or clonal expansion of microglia. Nat Neurosci 20, 793–803 (2017). https://doi.org/10.1038/nn.4547

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4547

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