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:

Microglia phagocytose myelin sheaths to modify developmental myelination

Abstract

During development, oligodendrocytes contact and wrap neuronal axons with myelin. Similarly to neurons and synapses, excess myelin sheaths are produced and selectively eliminated, but how elimination occurs is unknown. Microglia, the resident immune cells of the central nervous system, engulf surplus neurons and synapses. To determine whether microglia also prune myelin sheaths, we used zebrafish to visualize and manipulate interactions between microglia, oligodendrocytes, and neurons during development. We found that microglia closely associate with oligodendrocytes and specifically phagocytose myelin sheaths. By using a combination of optical, genetic, chemogenetic, and behavioral approaches, we reveal that neuronal activity bidirectionally balances microglial association with neuronal cell bodies and myelin phagocytosis in the optic tectum. Furthermore, multiple strategies to deplete microglia resulted in oligodendrocytes maintaining excessive and ectopic myelin. Our work reveals a neuronal activity-regulated role for microglia in modifying developmental myelin targeting by oligodendrocytes.

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: Microglia dynamically engage with myelin sheaths in the spinal cord.
Fig. 2: Microglia phagocytose myelin sheaths during developmental myelination.
Fig. 3: Microglia phagocytose myelin sheaths with minimal phagocytosis of oligodendrocyte somas.
Fig. 4: Neuronal activity regulates microglia–neuron interactions.
Fig. 5: Neuronal activity regulates myelin phagocytosis by microglia.
Fig. 6: Microglial ablation during myelination increases myelin sheath number.

Similar content being viewed by others

Data, code and materials availability

All data are available in the manuscript, the Source Data files or the Supplementary Information. Fiji scripts are available in the Supplementary Information. All plasmids and transgenic zebrafish are available by request.

References

  1. Hines, J. H., Ravanelli, A. M., Schwindt, R., Scott, E. K. & Appel, B. Neuronal activity biases axon selection for myelination in vivo. Nat. Neurosci. 18, 683–689 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mensch, S. et al. Synaptic vesicle release regulates myelin sheath number of individual oligodendrocytes in vivo. Nat. Neurosci. 18, 628–630 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gibson, E. M. et al. Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science 344, 1252304 (2014).

  4. Mitew, S. et al. Pharmacogenetic stimulation of neuronal activity increases myelination in an axon-specific manner. Nat. Commun. 9, 306 (2018).

  5. Scholz, J., Klein, M. C., Behrens, T. E. J. & Johansen-Berg, H. Training induces changes in white-matter architecture. Nat. Neurosci. 12, 1370–1371 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hughes, E. G., Orthmann-Murphy, J. L., Langseth, A. J. & Bergles, D. E. Myelin remodeling through experience-dependent oligodendrogenesis in the adult somatosensory cortex. Nat. Neurosci. 21, 696–706 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sampaio-Baptista, C. et al. Motor skill learning induces changes in white matter microstructure and myelination. J. Neurosci. 33, 19499–19503 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. McKenzie, I. A. et al. Motor skill learning requires active central myelination. Science 346, 318–322 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Liu, P., Du, J. & He, C. Developmental pruning of early-stage myelin segments during CNS myelination in vivo. Cell Res. 23, 962–964 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Li, Y., Du, X., Liu, C., Wen, Z.-L. & Du, J. Reciprocal regulation between resting microglial dynamics and neuronal activity in vivo. Dev. Cell 23, 1189–1202 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Tremblay, M.-È., Lowery, R. L. & Majewska, A. K. Microglial interactions with synapses are modulated by visual experience. PLoS Biol. 8, e1000527 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Schafer, D. P. et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hong, S. et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352, 712–716 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Neumann, H., Kotter, M. R. & Franklin, R. J. M. Debris clearance by microglia: an essential link between degeneration and regeneration. Brain 132, 288–295 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Uranova, N. A., Vikhreva, O. V., Rachmanova, V. I. & Orlovskaya, D. D. Ultrastructural alterations of myelinated fibers and oligodendrocytes in the prefrontal cortex in schizophrenia: a postmortem morphometric study. Schizophr. Res. Treatment 2011, 325789 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Pozner, A. et al. Intracellular calcium dynamics in cortical microglia responding to focal laser injury in the PC::G5-tdT reporter mouse. Front. Mol. Neurosci. 8, 12 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Eichhoff, G., Brawek, B. & Garaschuk, O. Microglial calcium signal acts as a rapid sensor of single neuron damage in vivo. Biochim. Biophys. Acta Mol. Cell Res. 1813, 1014–1024 (2011).

    Article  CAS  Google Scholar 

  18. Sieger, D., Moritz, C., Ziegenhals, T., Prykhozhij, S. & Peri, F. Long-range Ca2+ waves transmit brain-damage signals to microglia. Dev. Cell 22, 1138–1148 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Bai, Q., Sun, M., Stolz, D. B. & Burton, E. A. Major isoform of zebrafish P0 is a 23.5 kDa myelin glycoprotein expressed in selected white matter tracts of the central nervous system. J. Comp. Neurol. 519, 1580–1596 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Nevin, L. M., Robles, E., Baier, H. & Scott, E. K. Focusing on optic tectum circuitry through the lens of genetics. BMC Biol. 8, 126 (2010).

  21. Langebeck-Jensen, K., Shahar, O. D., Schuman, E. M., Langer, J. D. & Ryu, S. Larval zebrafish proteome regulation in response to an environmental challenge. Proteomics https://doi.org/10.1002/pmic.201900028 (2019).

  22. Shiau, C. E., Kaufman, Z., Meireles, A. M. & Talbot, W. S. Differential requirement for irf8 in formation of embryonic and adult macrophages in zebrafish. PLoS ONE 10, e0117513 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Herbomel, P., Thisse, B. & Thisse, C. Zebrafish early macrophages colonize cephalic mesenchyme and developing brain, retina, and epidermis through a M-CSF receptor-dependent invasive process. Dev. Biol. 238, 274–288 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Green, L. A., Nebiolo, J. C. & Smith, C. J. Microglia exit the CNS in spinal root avulsion. PLoS Biol. 17, e3000159 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Wlodarczyk, A. et al. A novel microglial subset plays a key role in myelinogenesis in developing brain. EMBO J. 36, 3292–3308 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Giera, S. et al. Microglial transglutaminase-2 drives myelination and myelin repair via GPR56/ADGRG1 in oligodendrocyte precursor cells. eLife 7, e33385 (2018).

  27. Almeida, R. G. et al. Myelination of neuronal cell bodies when myelin supply exceeds axonal demand. Curr. Biol. 28, 1296–1305 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Li, Q. et al. Developmental heterogeneity of microglia and brain myeloid cells revealed by deep single-cell RNA sequencing. Neuron 101, 207–223 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Fields, R. D. Volume transmission in activity-dependent regulation of myelinating glia. Neurochem. Int. 45, 503–509 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  31. Koudelka, S. et al. Individual neuronal subtypes exhibit diversity in CNS myelination mediated by synaptic vesicle release. Curr. Biol. https://doi.org/10.1016/j.cub.2016.03.070 (2016).

  32. Nelson, H. N. et al. Individual neuronal subtypes control initial myelin sheath growth and stabilization. Preprint at bioRxiv https://doi.org/10.1101/809996 (2019).

  33. Geraghty, A. C. et al. Loss of adaptive myelination contributes to methotrexate chemotherapy-related cognitive impairment. Neuron 193, 250–265 (2019).

  34. Baraban, M., Koudelka, S. & Lyons, D. A. Ca2+ activity signatures of myelin sheath formation and growth in vivo. Nat. Neurosci. 21, 19–23 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Nawaz, S. et al. Actin filament turnover drives leading edge growth during myelin sheath formation in the central nervous system. Dev. Cell 34, 139–151 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Elazar, N. et al. Axoglial adhesion by Cadm4 regulates CNS myelination. Neuron 101, 224–231 (2018).

  37. Hughes, A. N. & Appel, B. Oligodendrocytes express synaptic proteins that modulate myelin sheath formation. Nat. Commun. 10, 4125 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Stevens, B. et al. The classical complement cascade mediates CNS synapse elimination. Cell 131, 1164–1178 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Brunner, C., Lassmann, H., Waehneldt, T. V., Matthieu, J. M. & Linington, C. Differential ultrastructural localization of myelin basic protein, myelin/oligodendroglial glycoprotein, and 2′,3′-cyclic nucleotide 3′-phosphodiesterase in the CNS of adult rats. J. Neurochem. 52, 296–304 (1989).

    Article  CAS  PubMed  Google Scholar 

  40. van der Laan, L. J. et al. Macrophage phagocytosis of myelin in vitro determined by flow cytometry: phagocytosis is mediated by CR3 and induces production of tumor necrosis factor-alpha and nitric oxide. J. Neuroimmunol. 70, 145–152 (1996).

    Article  PubMed  Google Scholar 

  41. Barnett, M. H., Parratt, J. D. E., Cho, E. S. & Prineas, J. W. Immunoglobulins and complement in postmortem multiple sclerosis tissue. Ann. Neurol. 65, 32–46 (2009).

    Article  PubMed  Google Scholar 

  42. Gunner, G. et al. Sensory lesioning induces microglial synapse elimination via ADAM10 and fractalkine signaling. Nat. Neurosci. 22, 1075–1088 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Rotshenker, S. et al. Galectin-3/MAC-2, ras and PI3K activate complement receptor-3 and scavenger receptor-AI/II mediated myelin phagocytosis in microglia. Glia 56, 1607–1613 (2008).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  45. Stowell, R. D. et al. Noradrenergic signaling in the wakeful state inhibits microglial surveillance and synaptic plasticity in the mouse visual cortex. Nat. Neurosci. 22, 1782–1792 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Liu, Y. U. et al. Neuronal network activity controls microglial process surveillance in awake mice via norepinephrine signaling. Nat. Neurosci. 22, 1771–1781 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Brown, G. C. & Neher, J. J. Microglial phagocytosis of live neurons. Nat. Rev. Neurosci. 15, 209–216 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. De Biase, L. M. et al. Local cues establish and maintain region-specific phenotypes of basal ganglia microglia. Neuron 95, 341–356 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Oosterhof, N. et al. Homozygous mutations in CSF1R cause a pediatric-onset leukoencephalopathy and can result in congenital absence of microglia. Am. J. Hum. Genet. 104, 936–947 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kimmel, C. B., Ballard, W. W., Kimmel, S. R., Ullmann, B. & Schilling, T. F. Stages of embryonic development of the zebrafish. Dev. Dyn. 203, 253–310 (1995).

    Article  CAS  PubMed  Google Scholar 

  51. Higashijima, S.-I., Mandel, G. & Fetcho, J. R. Distribution of prospective glutamatergic, glycinergic, and GABAergic neurons in embryonic and larval zebrafish. J. Comp. Neurol. 480, 1–18 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Kwan, K. M. et al. The Tol2kit: a multisite gateway-based construction Kit for Tol2 transposon transgenesis constructs. Dev. Dyn. 236, 3088–3099 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Ellett, F., Pase, L., Hayman, J. W., Andrianopoulos, A. & Lieschke, G. J. mpeg1 promoter transgenes direct macrophage-lineage expression in zebrafish. Blood 117, e49–e56 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mathias, J. R., Zhang, Z., Saxena, M. T. & Mumm, J. S. Enhanced cell-specific ablation in zebrafish using a triple mutant of Escherichia coli nitroreductase. Zebrafish 11, 85–97 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Chen, S., Chiu, C. N., McArthur, K. L., Fetcho, J. R. & Prober, D. A. TRP channel mediated neuronal activation and ablation in freely behaving zebrafish. Nat. Methods 13, 147–150 (2016).

    Article  PubMed  CAS  Google Scholar 

  56. Shiau, C. E., Monk, K. R., Joo, W. & Talbot, W. S. An anti-inflammatory NOD-like receptor is required for microglia development. Cell Rep. 5, 1342–1352 (2013).

    Article  CAS  PubMed  Google Scholar 

  57. Almeida, R. & Lyons, D. Oligodendrocyte development in the absence of their target axons in vivo. PLoS ONE 11, e0164432 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Li, L., Jin, H., Xu, J., Shi, Y. & Wen, Z. Irf8 regulates macrophage versus neutrophil fate during zebrafish primitive myelopoiesis. Blood 117, 1359–1369 (2011).

    Article  CAS  PubMed  Google Scholar 

  59. Kucenas, S., Wang, W.-D., Knapik, E. W. & Appel, B. A selective glial barrier at motor axon exit points prevents oligodendrocyte migration from the spinal cord. J. Neurosci. 29, 15187–15194 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wang, Y. et al. Accurate quantification of astrocyte and neurotransmitter fluorescence dynamics for single-cell and population-level physiology. Nat. Neurosci. 22, 1936–1944 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Legland, D., Arganda-Carreras, I. & Andrey, P. MorphoLibJ: integrated library and plugins for mathematical morphology with ImageJ. Bioinformatics 32, 3532–3534 (2016).

    PubMed  Google Scholar 

  62. Longair, M. H., Baker, D. A. & Armstrong, J. D. Simple neurite tracer: open source software for reconstruction, visualization and analysis of neuronal processes. Bioinformatics 27, 2453–2454 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Preston (InVivo Biosystems) and J. Hines (Winona State University) for providing pME-NTR and p5E-myrf plasmids, respectively, and W. Macklin, E. Hughes and S. Bromley-Coolidge for comments on the manuscript. This work was supported by US National Institutes of Health (NIH) grant no. R01 NS095679, a gift from the Gates Frontiers Fund to B.A. and a National Science Foundation Graduate Research Fellowship (no. DGE-1553798) to A.N.H. The University of Colorado Anschutz Medical Campus Zebrafish Core Facility was supported by NIH grant no. P30 NS048154.

Author information

Authors and Affiliations

Authors

Contributions

A.N.H. and B.A. conceptualized the project. A.N.H. designed and performed the experiments and collected and analyzed the data. A.N.H. wrote, and B.A. edited, the manuscript.

Corresponding author

Correspondence to Bruce Appel.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks Marie-Ève Tremblay and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Calcium events in microglial processes contacting myelin sheaths compared to processes not contacting sheaths and comparison with event proximity to the soma.

a, b, Max dF/F (a) and event area (b) for large events (dF/F >0.1, area > 3 µm2) at non-sheath-contacting and sheath-contacting processes of microglia at 4 d.p.f. in Tg(mpeg1.1:GCaMP6s-CAAX; sox10:mRFP) larvae. Wilcoxon rank-sum test, n=41 events from 9 microglia in 9 fish. c, d, To distinguish if distance from cell soma is responsible for sheath-contacting vs non-contacting event differences, we split 645 events that occurred in 31 cells (31 fish) into distal and proximal halves and assessed event dF/F (c) and duration (d); ns by Wilcoxon rank-sum test.

Extended Data Fig. 2 Anti-Mbpa detects myelin within microglia.

a, Coronal section of Tg(mpeg1.1:mVenus-CAAX) 6 d.p.f. larval spinal cord stained with anti-Mbpa to detect myelin. Note anti-Mbpa inclusions (magenta) within microglia (yellow) in orthogonal views (right panels) taken at the locations marked (i), (ii), (iii). b, Coronal section of Tg(mbpa:mCherry-CAAX) 6 d.p.f. tectal commissure stained with anti-Mbpa (cyan) to detect myelin. Oligodendrocyte membrane (mCherry-CAAX) colocalizes with anti-Mbpa (cyan), both in the intact commissure and as a limited amount of extracellular debris (inset). Scale bars, 10 µm.

Extended Data Fig. 3 Acute treatment of larvae with bafilomycin A1 does not affect oligodendrocyte myelination.

a, Representative individual oligodendrocytes in transient-transgenic myrf:mScarlet-CAAX expressing larvae treated with bafilomycin A1 or DMSO. Scale bar, 10 µm. b, c, Quantification of sheath length and number of oligodendrocytes in each treatment group. n=12 larvae/12 fish (DMSO), 8 larvae/8 fish (bafilomycin A1). Data in (b) and (c) analyzed by Wilcoxon rank-sum test.

Extended Data Fig. 4 Glutamate uncaging evokes Ca2+ transients most reliably in neurons located < 20 µm away from the uncaging site.

a, Ratio of Ca2+ transient-experiencing neurons to those without transients binned in 10 µm bins from the uncaging site. b, Distance of neurons with Ca2+ transients (defined as dF/F>0.5) or no transients from the uncaging point in larvae treated with DMSO or MNI-glutamate, analyzed by Wilcoxon rank-sum test (n=fish/neurons, n=5/85 DMSO, 9/70 MNI-glutamate).

Extended Data Fig. 5 Microglia ablation does not change oligodendrocyte number or distribution.

a, Max projection images of hemi-spinal cords in 4 d.p.f. Tg(mbpa:TagRFP-T) larvae in each microglia ablation group and controls. Scale bar, 10 µm. b, c, Total number (b) and dorsal/ventral distribution (c) of oligodendrocytes per 3.5 hemisegments in each ablation group paired with corresponding controls, analyzed by Wilcoxon rank sum test (n=fish/oligodendrocytes, groups left to right, n=9/293, 12/442, 7/222, 8/271, 7/225, 6/209, 8/274, 6/208).

Extended Data Fig. 6 Working model of activity-regulated myelin phagocytosis by microglia.

Microglia phagocytose myelin to regulate myelin sheath number and targeting. Neuronal activity attracts microglia to contact neuronal cell bodies and reduces the amount of myelin that microglia can phagocytose, whereas reductions in neuronal activity promote myelin phagocytosis from axons.

Supplementary information

Supplementary Information

Supplementary Tables 1–3 and legends for Supplementary Videos 1–8.

Reporting Summary

Supplementary Video 1

Microglia exhibit Ca2+ transients at sheath- and non-sheath-contacting processes. Microglia in Tg(mpeg1.1:GCaMP6s-CAAX; sox10:mRFP) 4 d.p.f. larvae. Ca2+ events (cyan) are visible at process tips, along branches and in the soma. Note moving engulfed mRFP+ material within microglia and at points of microglia contact with sheaths. Videos are representative of microglial Ca2+ events (31/31 microglia imaged exhibited transients during 10 min). Scale bar, 10 µm.

Supplementary Video 2

Microglia exhibit Ca2+ transients at sheath- and non-sheath-contacting processes. Microglia in Tg(mpeg1.1:GCaMP6s-CAAX; sox10:mRFP) 4 d.p.f. larvae. Ca2+ events (cyan) are visible at process tips, along branches and in the soma. Note moving engulfed mRFP+ material within microglia and at points of microglia contact with sheaths. Videos are representative of microglial Ca2+ events (31/31 microglia imaged exhibited transients during 10 min). Scale bar, 10 µm.

Supplementary Video 3

Microglia Ca2+ transients before phagocytosis. Microglia in Tg(mpeg1.1:GCaMP6s-CAAX; sox10:mRFP), a 4 d.p.f. larva. A Ca2+ event at the distal process is followed by beading, engulfment and intracellular movement of mRFP+ phagocytosed material. Scale bar, 10 µm.

Supplementary Video 4

Microglia Ca2+ transients before phagocytosis. Microglia in Tg(mpeg1.1:GCaMP6s-CAAX; sox10:mRFP), a 4 d.p.f. larva. A Ca2+ event in the first frame (arrowhead) is followed by breakdown of the associated sheath (rectangle); a later event (second arrowhead) is followed by sheath beading but not loss within the acquisition time. Scale bar, 10 µm.

Supplementary Video 5

Engulfment of myelin sheaths by microglia. Top, timelapse imaging (~5 min per frame, timestamped in upper left) of a microglia (yellow) interacting with an oligodendrocyte (magenta) in a Tg(mpeg1.1:mVenus-CAAX; sox10:mRFP) larva. Two myelin sheaths (magenta rectangles) are intact and visibly surrounded by microglial processes initially, but become engulfed by the microglia soon after. Bottom, same video but only the RFP channel is shown to highlight the nascent sheaths that are engulfed. Scale bar, 10 µm.

Supplementary Video 6

3D rotation of a microglia containing myelin inclusions. A microglia (yellow) in a Tg(mpeg1.1:mVenus-CAAX; mbpa:mCherry-CAAX) larva containing phagocytosed myelin (magenta inclusions). Note intact myelin sheaths next to the microglia (magenta tubes). Movie generated with 3D Viewer plugin in Fiji.

Supplementary Video 7

3D rotation of microglia, myelin and neurons in the dorsal optic tectum. A 3D rendering of a ~40-µm z-stack of dorsal optic tectum in a 6 d.p.f. Tg(mpeg1.1:mVenus-CAAX; mbpa:mCherry-CAAX) larva mosaically expressing neuroD:mTagBFP-CAAX to sparsely label neurons. Microglia (yellow) are situated between the tectal commissure (magenta) and tectal neurons (cyan). Movie generated with 3D Viewer plugin in Fiji.

Supplementary Video 8

Microglial response to glutamate uncaging in tectal cell body layer. Timelapse imaging (1 min per frame) of a microglia in a Tg(mpeg1.1:mVenus-CAAX) larva with MNI-glutamate focally uncaged in the tectal cell body layer. Closed circle after the third minute represents the time and area of 405-nm laser application to uncage glutamate; open circle for remaining frames indicates the area where glutamate was uncaged. Scale bar, 20 µm.

Source data

Source Data Fig. 1

Source data for all experiments and plots in Fig. 1.

Source Data Fig. 2

Source data for all experiments and plots in Fig. 2 and Extended Data Figs. 1 and 2.

Source Data Fig. 3

Source data for all experiments and plots in Fig. 3 and Extended Data Fig. 3.

Source Data Fig. 4

Source data for all experiments and plots in Fig. 4 and Extended Data Fig. 4.

Source Data Fig. 5

Source data for all experiments and plots in Fig. 5.

Source Data Fig. 6

Source data for all experiments and plots in Fig. 6 and Extended Data Fig. 5.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hughes, A.N., Appel, B. Microglia phagocytose myelin sheaths to modify developmental myelination. Nat Neurosci 23, 1055–1066 (2020). https://doi.org/10.1038/s41593-020-0654-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-020-0654-2

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