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:

Repopulated microglia are solely derived from the proliferation of residual microglia after acute depletion

An Author Correction to this article was published on 20 November 2020

This article has been updated

Abstract

Newborn microglia rapidly replenish the whole brain after selective elimination of most microglia (>99%) in adult mice. Previous studies reported that repopulated microglia were largely derived from microglial progenitor cells expressing nestin in the brain. However, the origin of these repopulated microglia has been hotly debated. In this study, we investigated the origin of repopulated microglia by a series of fate-mapping approaches. We first excluded the blood origin of repopulated microglia via parabiosis. With different transgenic mouse lines, we then demonstrated that all repopulated microglia were derived from the proliferation of the few surviving microglia (<1%). Despite a transient pattern of nestin expression in newly forming microglia, none of repopulated microglia were derived from nestin-positive non-microglial cells. In summary, we conclude that repopulated microglia are solely derived from residual microglia rather than de novo progenitors, suggesting the absence of microglial progenitor cells in the adult brain.

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: Repopulated microglia rapidly replenish the whole brain after removal of CSF1R inhibition.
Fig. 2: Repopulated microglia do not originate from blood cells.
Fig. 3: Repopulated microglia are not derived from nestin-expressing cells.
Fig. 4: Nestin is transiently expressed in repopulated microglia.
Fig. 5: Repopulated microglia are solely derived from residual microglia.
Fig. 6: Repopulated microglia are derived from the proliferation of residual microglia.
Fig. 7: Transcriptomes of repopulated microglia are distinct from resident microglia and transcriptomes of the brain are little influenced by repopulated microglia.

Similar content being viewed by others

Change history

References

  1. Kettenmann, H., Hanisch, U. K., Noda, M. & Verkhratsky, A. Physiology of microglia. Physiol. Rev. 91, 461–553 (2011).

    Article  CAS  Google Scholar 

  2. del Río Hortega, P. Microglia (Hoeber, New York, 1932).

  3. del Río Hortega, P. & Penfield, W. Cerebral cicatrix: the reaction of neuroglia and microglia to brain wounds. Bull. Johns Hopkins Hosp. 41, 278–303 (1927).

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

    Article  CAS  Google Scholar 

  5. Erblich, B., Zhu, L., Etgen, A. M., Dobrenis, K. & Pollard, J. W. Absence of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain development and olfactory deficits. PLoS One 6, e26317 (2011).

    Article  CAS  Google Scholar 

  6. Chitu, V., Gokhan, Ş., Nandi, S., Mehler, M. F. & Stanley, E. R. Emerging roles for CSF-1 receptor and its ligands in the nervous system. Trends Neurosci. 39, 378–393 (2016).

    Article  CAS  Google Scholar 

  7. Elmore, M. R. 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).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. 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).

    Article  CAS  Google Scholar 

  10. 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).

    Article  CAS  Google Scholar 

  11. Tay, T. L. et al. A new fate mapping system reveals context-dependent random or clonal expansion of microglia. Nat. Neurosci. 20, 793–803 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  13. Varvel, N. H. et al. Microglial repopulation model reveals a robust homeostatic process for replacing CNS myeloid cells. Proc. Natl. Acad. Sci. USA 109, 18150–18155 (2012).

    Article  CAS  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).

    Article  CAS  Google Scholar 

  15. Valdearcos, M., et al. Microglial inflammatory signaling orchestrates the hypothalamic immune response to dietary excess and mediates obesity susceptibility. Cell Metab. 26, 185–197.e183 (2017).

    Article  CAS  Google Scholar 

  16. Valdearcos, M. et al. Microglia dictate the impact of saturated fat consumption on hypothalamic inflammation and neuronal function. Cell Rep. 9, 2124–2138 (2014).

    Article  CAS  Google Scholar 

  17. 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).

    Article  CAS  Google Scholar 

  18. Radu, M. & Chernoff, J. An in vivo assay to test blood vessel permeability. J. Vis. Exp. https://doi.org/10.3791/50062 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Kamran, P. et al. Parabiosis in mice: a detailed protocol. J. Vis. Exp. https://doi.org/10.3791/50556 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Yuan, T. F., Liang, Y. X., Peng, B., Lin, B. & So, K. F. Local proliferation is the main source of rod microglia after optic nerve transection. Sci. Rep. 5, 10788 (2015).

    Article  CAS  Google Scholar 

  21. Miron, V. E. et al. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat. Neurosci. 16, 1211–1218 (2013).

    Article  CAS  Google Scholar 

  22. Villeda, S. A. et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 477, 90–94 (2011).

    Article  CAS  Google Scholar 

  23. 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).

    Article  CAS  Google Scholar 

  24. Okabe, M., Ikawa, M., Kominami, K., Nakanishi, T. & Nishimune, Y. ‘Green mice’ as a source of ubiquitous green cells. FEBS Lett. 407, 313–319 (1997).

    Article  CAS  Google Scholar 

  25. Lagace, D. C. et al. Dynamic contribution of nestin-expressing stem cells to adult neurogenesis. J. Neurosci. 27, 12623–12629 (2007).

    Article  CAS  Google Scholar 

  26. 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  Google Scholar 

  27. Amat, J. A., Ishiguro, H., Nakamura, K. & Norton, W. T. Phenotypic diversity and kinetics of proliferating microglia and astrocytes following cortical stab wounds. Glia 16, 368–382 (1996).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. Mathys, H. et al. Temporal tracking of microglia activation in neurodegeneration at single-cell resolution. Cell Rep. 21, 366–380 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  32. 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).

    Article  CAS  Google Scholar 

  33. Peng, B. et al. Suppression of microglial activation is neuroprotective in a mouse model of human retinitis pigmentosa. J. Neurosci. 34, 8139–8150 (2014).

    Article  Google Scholar 

  34. Beggs, S. & Salter, M. W. Snapshot: microglia in disease. Cell 165, 1294–1294.e1291 (2016).

    Article  CAS  Google Scholar 

  35. Pitulescu, M. E., Schmidt, I., Benedito, R. & Adams, R. H. Inducible gene targeting in the neonatal vasculature and analysis of retinal angiogenesis in mice. Nat. Protoc. 5, 1518–1534 (2010).

    Article  CAS  Google Scholar 

  36. Chapman, A. R. et al. Single cell transcriptome amplification with MALBAC. PLoS One 10, e0120889 (2015).

    Article  Google Scholar 

  37. Sheng, K., Cao, W., Niu, Y., Deng, Q. & Zong, C. Effective detection of variation in single-cell transcriptomes using MATQ-seq. Nat. Methods 14, 267–270 (2017).

    Article  CAS  Google Scholar 

  38. Chen, K. et al. Methyltransferase SETD2-mediated methylation of STAT1 is critical for interferon antiviral activity. Cell 170, 492–506.e414 (2017).

    Article  CAS  Google Scholar 

  39. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  Google Scholar 

  40. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  Google Scholar 

  41. Pertea, M. et al. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 33, 290–295 (2015).

    Article  CAS  Google Scholar 

  42. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  Google Scholar 

  43. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors wish to thank C. Ren, K. Wang and L. Huang (Jinan University), J. Chang, P. Ren, J. Zhang, Z. Yao and W. Zhan (Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences), and Q. Gao, L. Yang, H. Zhong, C. Zhang, W. Zhao, Z. Dong, B. Chen, W. Wu, F. Fan, Z. Liu and M. Xie (BGI) for technical support. They thank C. Liu (Zhejiang University) for donating NG2-CreER mice. They also thank H. Zheng for supporting the establishment of Bo Peng's laboratory. The authors thank P. Lin, B. West, P. Singh and A. Rymar (Plexxikon Inc.) for kindly providing the PLX5622 compound and formulated chow diet. Last but not least, the authors show their gratitude and respect to all animals sacrificed in this study. This study was supported by National Key R&D Program of China (2017YFC0111202; B.P.), National Natural Science Foundation of China (31600839; B.P.), Shenzhen Science and Technology Research Program (JCYJ20170307171222692 and JCYJ20170818163320865 to B.P.; JCYJ20170818161734072 to Y.H.), Guangdong Innovative and Entrepreneurial Research Team Program (2013S046; B.P.) and Shenzhen Peacock Plan (B.P.). This study was also supported by NSFC Grants (31771215, 81501164 and 81611130224; T.-F.Y.) and Young Elite Scientists Sponsorship Program by CAST (YESS; T.Y.).

Author information

Authors and Affiliations

Authors

Contributions

B.P. and Y.R. designed and initiated this study. B.P. conducted quality control on the data and conceptualized the research. B.P. supervised this study. B.P. wrote the manuscript with inputs from T.-F.Y., Y.R., Y.H., S.X. and F.S. B.P., Y.H., S.X., J.W., F.S., Z.X., L.Z., Y.-X.L., Z.L., K.-F.S., T.W., Y.P., N.L., M.S.H. and G.H. performed experiments. Y.H., J.W. Y.R. and B.P. performed most neuroanatomy experiments. S.X. maintained the transgenic animals and performed flow cytometry. Z.X. performed single-cell RT-PCR and FACS-ELISA with the assistance of S.X. and F.S. F.S. and Y.-X.L. performed parabiosis surgery. Z.X., S.X., Y.H., and F.S. performed RNA-seq experiments. G.Q. and B.P. analyzed the RNA-seq results. Y.H., T.Y., S.X. and B.P. performed statistical analysis of results. B.P., Y.H., T.-F.Y. and Y.R. contributed to the interpretation of results. B.P. assembled the figures. All authors discussed results and commented on the manuscript.

Corresponding authors

Correspondence to Ti-Fei Yuan, Yanxia Rao or Bo Peng.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Inhibition of CSF1R by PLX5622 rapidly depletes brain microglia.

(a) Scheme of PLX5622 administration and time points for observation. (b) Representative images show microglial numbers are reduced in the brain after PLX5622 administration. Each white dot represents a microglial cell. (c) Zoom-in images of microglia in somatosensory cortex. (d) Quantification of microglial density in somatosensory cortex of normal brains and the brains on PLX5622 treatment. N (mouse number) = 8, 4, 4, 4, 3, 4, 7 and 7, respectively. PLX5622: PLX5622 formulated diet. Green: GFP; blue: DAPI. The data are presented as mean ± SD; NS: not significant; *: p < 0.05; **: p < 0.01; ***: p < 0.001. One tailed one-way ANOVA with Tukey’s post hoc. p = 0.011, p < 0.001, p < 0.001, p < 0.001, p < 0.001, p < 0.001 and p < 0.001, respectively.

Supplementary Figure 2 Inhibition of CSF1R by PLX5622 rapidly depletes microglia in hippocampus.

(a) Representative confocal images show microglial numbers are reduced in the hippocampus after PLX5622 administration. (b) Quantification of microglial density in hippocampus of normal brains and the brains on PLX5622 treatment. N (mouse number) = 8, 4, 4, 4, 3, 4, 7 and 7, respectively. Green: GFP; blue: DAPI. The data are presented as mean ± SD; NS: not significant; *: p < 0.05; **: p < 0.01; ***: p < 0.001. One tailed one-way ANOVA with Tukey’s post hoc. p < 0.001, p < 0.001, p < 0.001, p < 0.001, p < 0.001, p < 0.001 and p < 0.001, respectively.

Supplementary Figure 3 Repopulated microglia rapidly replenish the hippocampus after removal of CSF1R inhibition.

(a) Representative images show microglia are repopulated in hippocampus after removal of PLX5622. Each white dot indicates a microglial cell. (b) Quantification of microglial density in hippocampus during microglial repopulation. The red line and red area indicate the mean and SD of microglial density in normal brain, respectively. N (mouse number) = 7, 3, 3, 4, 4, 4, 3, 3, 3 and 8, respectively. NS: not significant; *: p < 0.05 to D0; **: p < 0.01 to D0; ***: p < 0.001 to D0; p = 0.983, p = 0.531, p = 0.002, p < 0.001, p < 0.001, p < 0.001, p < 0.001 and p < 0.001, respectively. #: p < 0.05 to normal brain; ##: p < 0.01 to normal brain; ###: p < 0.001 to normal brain; p < 0.001, p < 0.001, p < 0.001, p < 0.001, p > 0.999, p = 0.754, p = 0.995, p > 0.999 and p = 0.988, respectively. (c) Quantification of BrdU-positive microglia among all microglia in S1 during repopulation. N (mouse number) = 4, 3, 3, 4, 3, 3, 3, 3 and 3, respectively. NS: not significant; *: p < 0.05; **: p < 0.01; ***: p < 0.001; p = 0.001, p = 0.001, p = 0.080, p = 0.012, p > 0.999, 0.997, p > 0.999 and p > 0.999, respectively. Green: GFP; blue: DAPI; magenta: BrdU. The data are presented as mean ± SD. One tailed one-way ANOVA with Tukey’s post hoc.

Supplementary Figure 4

Representative gating strategy for blood cell analyses.

Supplementary Figure 5 Circulating progenitors in parabiotic mice can give rise to macrophages in peripheral organs.

GFP-positive macrophages are found in the kidney and spleen of WT parabionts. Arrows: GFP-positive macrophages. Green: GFP; red: Iba1. Each experiment has been independently repeated twice with similar results.

Supplementary Figure 6 Repopulated microglia do not originate from blood cells.

(a-b) Confocal images and quantificative results show there are no GFP-positive repopulated microglia in the hippocampus of WT parabionts. N (mouse number) = 6, 4 and 7, respectively. p < 0.001 (WT vs GFP), p > 0.999 (WT vs parabiotic-WT) and p < 0.001 (GFP vs parabiotic-WT), respectively. (c-d) Confocal images and quantificative results show there are no GFP-positive repopulated microglia in the median eminence of WT parabionts. N (mouse number) = 6, 4 and 7, respectively. p < 0.001 (WT vs GFP), p > 0.999 (WT vs parabiotic-WT) and p < 0.001 (GFP vs parabiotic-WT), respectively. Green: GFP; red: Iba1; blue: DAPI. The data are presented as mean ± SD. N: mouse number for each group; NS: not significant; *: p < 0.05; **: p < 0.01; ***: p < 0.001. One tailed one-way ANOVA with Tukey’s post hoc. Box plot elements: square for mean, box range for percentiles at 25 and 75, inside-box line for median and whisker for min and max. Each experiment in (g-h) has been independently repeated at least twice.

Supplementary Figure 7 Repopulated microglia are not derived from nestin-expressing cells.

(a) No tdTomato-positive microglia are found in hippocampus of treatment protocol Fig. 4a. (b) No tdTomato-positive microglia are found in hippocampus of treatment protocol Fig. 4e. Arrows: tdTomato-positive neurons. Green: GFP; red: tdTomato. Each experiment has been repeated 3 times independently with similar results.

Supplementary Figure 8 Uncropped gels for single-cell RT-PCR.

(a–g) Uncropped gels for single-cell RT-PCR in Fig. 4 of the normal brain (a), microglial depletion for 3 days (b), microglial repopulation for 3 (c), 5 (d) and 14 days (e), developmental P3 brain microglia (f) and microglia in the injured brain (g), respectively.

Supplementary Figure 9 Astrocytes, OPCs or neurons do not differentiate into microglia during repopulation.

(a) Scheme of fate mapping for the origin of repopulated microglia. (b-c) The rationale and hypotheses of tamoxifen triggered fate mapping. (d) Confocal images indicate no tdTomato-positive microglia in repopulated brains of GLAST-CreERT2::Ai14, NG2-CreER::Ai9, CaMK2a-CreERT2::Ai14, GAD2-CreER::Ai14 and TH-IRES-CreER::Ai14 mice, respectively. Each experiment has been independently repeated at least twice with similar results. PLX5622: PLX5622 formulated diet; CD: control diet; TAM: tamoxifen. Green: GFP; red: tdTomato.

Supplementary Figure 10 Repopulated microglia in the hippocampus are derived from residual microglia.

(a) Confocal images show all brain microglia in the hippocampus of tamoxifen administered mice at 2-month old are genetically labeled by tdTomato. (b) Confocal images indicate all repopulated microglia in the hippocampus are labeled with tdTomato in repopulating brain of tamoxifen-treated group. Green: GFP; red: Iba1. Each experiment has been independently repeated three times (7 mice) with similar results.

Supplementary Figure 11 CSF1R inhibition kills brain microglia instead of inducing microglial dedifferentiation.

The number of tdTomato-positive cells in microglia-labeled Cx3cr1-CreER::Ai14 mice is significantly reduced after PLX5622 administration for 10 days. All surviving tdTomato-positive cells are co-labeled with Iba1. Green: Iba1; red: tdTomato; blue: DAPI. The experiment has been independently repeated 3 times with similar results.

Supplementary Figure 12 Model of the origin of repopulated microglia.

(a) Repopulated microglia are not derived from blood cells. (b) Repopulated microglia are not derived from Nestin-positive progenitor cells. (c) Repopulated microglia are not derived from astrocytes, OPCs or neurons. (d) Repopulated microglia are derived from the direct proliferation of residual microglia.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 and Supplementary Table 1

Life Sciences Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, Y., Xu, Z., Xiong, S. et al. Repopulated microglia are solely derived from the proliferation of residual microglia after acute depletion. Nat Neurosci 21, 530–540 (2018). https://doi.org/10.1038/s41593-018-0090-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-018-0090-8

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