Skip to main content

Advertisement

Log in

New oligodendrocytes exhibit more abundant and accurate myelin regeneration than those that survive demyelination

  • Brief Communication
  • Published:

From Nature Neuroscience

View current issue Submit your manuscript

Abstract

Oligodendrocytes that survive demyelination can remyelinate, including in multiple sclerosis (MS), but how they do so is unclear. In this study, using zebrafish, we found that surviving oligodendrocytes make few new sheaths and frequently mistarget new myelin to neuronal cell bodies, a pathology we also found in MS. In contrast, oligodendrocytes generated after demyelination make abundant and correctly targeted sheaths, indicating that they likely also have a better regenerative potential in MS.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Remyelination by oligodendrocytes that survive demyelination in the Tg(mbp:TRPV1-tagRFPt) zebrafish model.
Fig. 2: Myelin sheath dynamics of surviving oligodendrocytes after demyelination in the Tg(mbp:TRPV1-tagRFPt) zebrafish model.
Fig. 3: Mistargeted myelin profiles are present in remyelinating lesions in motor cortex tissue from people with MS.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Code availability

No code was used in this manuscript.

References

  1. Thompson, A. J., Baranzini, S. E., Geurts, J., Hemmer, B. & Ciccarelli, O. Multiple sclerosis. Lancet 391, 1622–1636 (2018).

    Article  Google Scholar 

  2. Franklin, R. J. M., Frisén, J. & Lyons, D. A. Revisiting remyelination: towards a consensus on the regeneration of CNS myelin. Semin. Cell Dev. Biol. 116, 3–9 (2020).

    Article  Google Scholar 

  3. Cole, K. L. H., Early, J. J. & Lyons, D. A. Drug discovery for remyelination and treatment of MS. Glia 65, 1565–1589 (2017).

    Article  Google Scholar 

  4. Yeung, M. S. Y. et al. Dynamics of oligodendrocyte generation in multiple sclerosis. Nature 566, 538–542 (2019).

    Article  CAS  Google Scholar 

  5. Duncan, I. D. et al. The adult oligodendrocyte can participate in remyelination. Proc. Natl Acad. Sci. USA 115, E11807–E11816 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Bacmeister, C. M. et al. Motor learning promotes remyelination via new and surviving oligodendrocytes. Nat. Neurosci. 23, 819–831 (2020).

    Article  CAS  Google Scholar 

  7. Klingseisen, A. et al. Oligodendrocyte neurofascin independently regulates both myelin targeting and sheath growth in the CNS article oligodendrocyte neurofascin independently regulates both myelin targeting and sheath growth in the CNS. Dev. Cell 51, 730–744 (2019).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. Czopka, T., ffrench-Constant, C. & Lyons, D. A. Individual oligodendrocytes have only a few hours in which to generate new myelin sheaths invivo. Dev. Cell 25, 599–609 (2013).

    Article  CAS  Google Scholar 

  10. Auer, F., Vagionitis, S. & Czopka, T. Evidence for myelin sheath remodeling in the CNS revealed by in vivo imaging. Curr. Biol. 28, 549–559 (2018).

    Article  CAS  Google Scholar 

  11. Harboe, M., Torvund-Jensen, J., Kjaer-Sorensen, K. & Laursen, L. S. Ephrin-A1-EphA4 signaling negatively regulates myelination in the central nervous system. Glia 66, 934–950 (2018).

    Article  Google Scholar 

  12. Wolf, R. M., Wilkes, J. J., Chao, M. V. & Resh, M. D. Tyrosine phosphorylation of p190 RhoGAP by Fyn regulates oligodendrocyte differentiation. J. Neurobiol. 49, 62–78 (2001).

    Article  CAS  Google Scholar 

  13. Albert, M., Antel, J., Brück, W. & Stadelmann, C. Extensive cortical remyelination in patients with chronic multiple sclerosis. Brain Pathol. 17, 129–138 (2007).

    Article  Google Scholar 

  14. Snaidero, N. et al. Myelin membrane wrapping of CNS axons by PI(3,4,5)P3-dependent polarized growth at the inner tongue. Cell 156, 277–290 (2014).

    Article  CAS  Google Scholar 

  15. 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, 696–708 (2018).

    Article  CAS  Google Scholar 

  16. Hill, R. A., Patel, K. D., Goncalves, C. M., Grutzendler, J. & Nishiyama, A. Modulation of oligodendrocyte generation during a critical temporal window after NG2 cell division. Nat. Neurosci. 17, 1518–1529 (2014).

    Article  CAS  Google Scholar 

  17. Orthmann-Murphy, J. et al. Remyelination alters the pattern of myelin in the cerebral cortex. eLife 9, e56621 (2020).

  18. Jäkel, S. et al. Altered human oligodendrocyte heterogeneity in multiple sclerosis. Nature 566, 543–547 (2019).

    Article  Google Scholar 

  19. Kirby, B. B. et al. In vivo time-lapse imaging shows dynamic oligodendrocyte progenitor behavior during zebrafish development. Nat Neurosci. 9, 1506–1511 (2006).

    Article  CAS  Google Scholar 

  20. Hughes, E. G., Kang, S. H., Fukaya, M. & Bergles, D. E. Oligodendrocyte progenitors balance growth with self-repulsion to achieve homeostasis in the adult brain. Nat. Neurosci. 16, 668–679 (2013).

    Article  CAS  Google Scholar 

  21. Almeida, R. G., Czopka, T., ffrench-Constant, C. & Lyons, D. A. Individual axons regulate the myelinating potential of single oligodendrocytes in vivo. Development 138, 4443–4450 (2011).

    Article  CAS  Google Scholar 

  22. Ellett, F., Pase, L., Hayman, J. W., Andrianopoulos, A. & Lieschke, G. J. mpeg1 promoter transgenes direct macrophage-lineage expression in zebrafish. Blood 27, e49–e56 (2011).

    Article  Google Scholar 

  23. Bin, J. M. & Lyons, D. A. Imaging myelination in vivo using transparent animal models. Brain Plast. 2, 3–29 (2016).

    Article  Google Scholar 

  24. Baraban, M., Koudelka, S. & Lyons, D. A. Ca2+ activity signatures of myelin sheath formation and growth in vivo. Nat. Neurosci. 21, 19–25 (2018).

    Article  CAS  Google Scholar 

  25. Hamilton, N. B., Kolodziejczyk, K., Kougioumtzidou, E. & Attwell, D. Proton-gated Ca2+-permeable TRP channels damage myelin in conditions mimicking ischaemia. Nature 529, 523–527 (2016).

    Article  CAS  Google Scholar 

  26. Paez, P. M. & Lyons, D. A. Annual review of neuroscience calcium signaling in the oligodendrocyte lineage: regulators and consequences. Annu. Rev. Neurosci. 43, 163–186 (2020).

    Article  CAS  Google Scholar 

  27. 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  Google Scholar 

  28. 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  Google Scholar 

  29. Karttunen, M. J. et al. Regeneration of myelin sheaths of normal length and thickness in the zebrafish CNS correlates with growth of axons in caliber. PLoS ONE 12, e0178058 (2017).

  30. Early, J. J. et al. An automated high-resolution in vivo screen in zebrafish to identify chemical regulators of myelination. eLife 7, e35156 (2018).

  31. Nicaise, A. M. et al. Cellular senescence in progenitor cells contributes to diminished remyelination potential in progressive multiple sclerosis. Proc. Natl Acad. Sci. USA 116, 9030–9039 (2019).

    Article  CAS  Google Scholar 

  32. Lloyd, A. F. et al. Central nervous system regeneration is driven by microglia necroptosis and repopulation. Nat. Neurosci. 22, 1046–1052 (2019).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank C. ffrench-Constant, E. Hughes and the Lyons Laboratory for feedback, the Bioresearch & Veterinary Services aquatics facility for fish care, S. Mitchell for electron microscopy assistance and C. Melendez-Vasquez for suggesting the ROCK experiment. This work was supported by Wellcome Trust Senior Research Fellowships (102836/Z/13/Z and 214244/Z/18/Z), a Medical Research Council Project Grant (MR/P006272/1) and an MS Society Innovative Grant (95) to D.A.L. S.A.N. and J.M.W. were supported by the Wellcome Trust Four-Year Ph.D. Program in Tissue Repair (grant 108906/Z/15/Z) and J.M.W. by a University of Edinburgh Ph.D. Tissue Repair Studentship Award (MRC Doctoral Training Partnership MR/K501293/1). L.Z. and A.W. were supported by an MS Society UK Centre grant.

Author information

Authors and Affiliations

Authors

Contributions

S.A.N., J.M.W. and D.A.L. conceived the project. S.A.N., J.M.W., A.K. and J.J.E. designed and performed the in vivo experiments. L.Z. and A.W. designed and performed the human tissue experiments. S.A.N. and D.A.L. co-wrote the manuscript, edited by all. D.A.L. managed the project.

Corresponding author

Correspondence to David A. Lyons.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Neuroscience thanks Bruce Appel, Jennifer Orthmann-Murphy, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Characterisation of the Tg(mbp:TRPV1-tagRFPt) zebrafish model.

(a) Schematic illustrating the Tg(mbp:TRPV1-tagRFPt) demyelination model made using Biorender. The rat ortholog of the TRPV1 channel is expressed in myelinating oligodendrocytes and is activated by addition of csn which drives cation influx. Zebrafish TRPV1 channels are insensitive to csn, therefore csn treatment specifically results in damage to myelinating glia which express the rat ortholog of the TRPV1 channel. (b) Confocal images of myelinating oligodendrocytes in the Tg(mbp:EGFP; mbp:TRPV1- tagRFPt) zebrafish line at 4dpf showing oligodendrocytes co-expressing EGFP and tagRFPt in the merged image. Scale bar, 20 µm. (c) Brightfield images of a zebrafish containing the Tg(mbp:TRPV1-tagRFPt) transgene (TRPV1 + ve), or wildtype siblings without the Tg(mbp:TRPV1-tagRFPt) transgene (TRPV1-ve) which show no developmental differences at 4dpf. Scale bars, 500 µm. (d and e) Confocal images of the (d) Tg(mbp:EGFP-CAAX) line and (e) the Tg(mbp:EGFP) line pre-treatment (indicated here as pre-t) at 4dpf, and 3hpt. Zebrafish not containing the Tg(mbp:TRPV1-tagRFPt) transgene show no disruption to myelin or oligodendrocytes following a 2 hour treatment of 10 µM csn. Scale bars, 20 µm. (f) Confocal images of myelin visualised in Tg(mbp:EGFP-CAAX; mbp:TRPV1-tagRFPt) zebrafish, with control (DMSO) and csn treated animals in top and bottom panels respectively pre-treatment (pre-t) at 4dpf, 3hpt, 1dpt and 3dpt. Scale bar, 20 µm.

Extended Data Fig. 2 Csn treatment induces severe demyelination in the Tg(mbp:TRPV1-tagRFPt) zebrafish model.

(a) Transmission electron microscopy images of DMSO and csn treated Tg(mbp:TRPV1-tagRFPt) zebrafish at 1dpt show numerous large calibre myelinated axons (blue) in DMSO treated animals and numerous large calibre unmyelinated axons (yellow) in csn treated animals. Scale bar, 1 µm. (b) Quantification of the number of myelinated axons>0.4 µm diameter in the dorsal spinal cord at 1dpt in DMSO (median = 20.00, IQR = 16.50-22.00) versus csn (median = 0.00, IQR = 0.00-1.50) treated zebrafish, p < 0.0001. Unpaired two-tailed t-test with Welch’s correction. N = 5 zebrafish per condition. Data are presented as median with IQR. (c) Quantification of the number of unmyelinated axons>0.4 µm diameter in the dorsal spinal cord at 1dpt in DMSO (mean = 10.00 ± 5.24 SD) versus csn (mean = 27.40 ± 9.61 SD) treated zebrafish, p = 0.0075. Unpaired two-tailed t-test. N = 5 zebrafish per condition. Data are presented as mean ± SD. (d) Quantification of the number of axons >0.4 µm diameter in the dorsal spinal cord at 1dpt in DMSO (mean = 29.40 ± 8.26 SD) versus csn (mean = 28.00 ± 9.95 SD) treated zebrafish, p = 0.8148. Unpaired two-tailed t-test. N = 5 zebrafish per condition. Data are presented as mean ± SD. (e) Quantification of the number of axons with>3 myelin wraps in the dorsal spinal cord at 1dpt in DMSO (median = 20.00, IQR = 15.00-20.00) versus csn (median = 0.00, IQR = 0.00-1.50) treated zebrafish, p = 0.0079. Two-tailed Mann-Whitney test. N = 5 zebrafish per condition. Data are presented as median with IQR (f) Quantification of the number of axons with ≤3 myelin wraps in the dorsal spinal cord at 1dpt in DMSO (median = 1.00, IQR = 0.00-3.00) versus csn (median = 0.00, IQR = 0.00-0.00) treated zebrafish, p = 0.1667. Two-tailed Kolmogorov-Smirnov test. N = 5 zebrafish per condition. Data is presented as median with IQR. (g) Transmission electron microscopy images of DMSO and csn treated Tg(mbp:TRPV1-tagRFPt) zebrafish at 3dpt show numerous large calibre myelinated axons (>3 myelin wraps highlighted in blue, ≤3 myelin wraps highlighted in orange) and unmyelinated axons (highlighted in yellow) in DMSO and csn treated animals. Scale bar, 1 µm. (h) Quantification of the number of myelinated axons >0.4 µm diameter in the dorsal spinal cord at 3dpt in DMSO (mean = 24.20 ± 9.83 SD) versus csn (mean = 20.83 ± 5.78 SD) treated zebrafish, p = 0.4963. Unpaired two-tailed t-test. N = 5 DMSO treated zebrafish, N = 6 csn treated zebrafish. Data are presented as mean ± SD. (i) Quantification of the number of unmyelinated axons>0.4 µm diameter in the dorsal spinal cord at 3dpt in DMSO (mean = 23.20 ± 6.76 SD) versus csn (mean = 30.17 ± 3.97 SD) treated zebrafish, p = 0.0616. Unpaired two-tailed t-test. N = 5 DMSO treated zebrafish, N = 6 csn treated zebrafish. Data are presented as mean ± SD. (j) Quantification of the number of axons>0.4 µm diameter in the dorsal spinal cord at 3dpt in DMSO (mean = 47.60 ± 10.92 SD) versus csn (mean = 51.00 ± 6.45 SD) treated zebrafish, p = 0.5359. Unpaired two-tailed t-test. N = 5 DMSO treated zebrafish, N = 6 csn treated zebrafish. (k) Quantification of the number of axons with>3 myelin wraps in the dorsal spinal cord at 3dpt in DMSO (median = 16.00, IQR = 15.50-27.00) versus csn (median = 8.50, IQR = 3.75-11.50) treated zebrafish, p = 0.0152. Two-tailed Mann-Whitney test. N = 5 DMSO treated zebrafish, N = 6 csn treated zebrafish. (l) Quantification of the number of axons with≤3 myelin wraps in the dorsal spinal cord at 3dpt in DMSO (mean = 4.00 ± 3.24 SD) versus csn (mean = 12.50 ± 5.01 SD) treated zebrafish, p = 0.0099. Unpaired two-tailed t-test. N = 5 DMSO treated zebrafish, N = 6 csn treated zebrafish.

Extended Data Fig. 3 Csn treatment induces minimal oligodendrocyte loss in the Tg(mbp:TRPV1-tagRFPt) model.

(a) Confocal images of myelinating oligodendrocytes visualised in Tg(mbp:EGFP; mbp:TRPV1-tagRFPt) zebrafish, with control (DMSO) and csn treated animals in top and bottom panels respectively pre-treatment (pre-t) at 4dpf, 3hpt, 1dpt and 3dpt. Scale bar, 20 µm. (b-f) Quantification of myelinating oligodendrocyte number in DMSO and csn treated Tg(mbp:EGFP; mbp:TRPV1-tagRFPt) zebrafish over time. (b) Pre-treatment DMSO (mean = 58.60 ± 13.23 SD) versus csn (mean = 57.20 ± 7.64 SD), p = 0.7753. Data are presented as mean ± SD. (c) 3hpt DMSO (mean = 65.00 ± 14.93 SD) versus csn (mean = 44.11 ± 10.36 SD), p = 0.0041. Data are presented as mean ± SD. (d) 1dpt DMSO (mean = 76.90 ± 14.14 SD) versus csn (mean = 61.70 ± 7.39 SD), p = 0.0075. Data are presented as mean ± SD. (e) 3dpt DMSO (mean = 99.00 ± 15.93 SD) versus csn (mean = 85.70 ± 11.83 SD), p = 0.0444. Data are presented as mean ± SD. (f) 5dpt DMSO (mean = 104.5 ± 12.95 SD) versus csn (mean = 108.5 ± 15.10 SD), p = 0.5328. (b-f) Unpaired two-tailed t-tests. Pre-treatment N = 10 zebrafish per treatment group, 3hpt N = 8 (DMSO) and 9 (csn) treated zebrafish, 1dpt N = 10 zebrafish per treatment group, 3dpt N = 11 zebrafish (DMSO) and 10 zebrafish (csn), 5 dpt N = 10 zebrafish per treatment group. Data are presented as mean ± SD. Each data point represents total (dorsal + ventral) oligodendrocyte number analysed per imaged area per zebrafish.

Extended Data Fig. 4 Characterisation of single oligodendrocyte loss and myelin debris phagocytosis following demyelination in the Tg(mbp:TRPV1-tagRFPt) zebrafish model.

(a) Confocal images of a single oligodendrocyte labelled with mbp:EGFP-CAAX in the Tg(mbp:TRPV1-tagRFPt) line pre-treatment (pre-t) at 4dpf, and 3hpt. An example of oligodendrocyte cell death is demonstrated here by the disappearance of a tagRFPt+ve cell body following csn treatment in the same zebrafish before and after demyelination, whilst 2 tagRFPt+ve oligodendrocytes which survive demyelination are seen neighbouring it at 3hpt. Arrows indicate the location of the oligodendrocyte cell body which undergoes cell death, or where it was prior to demyelination. Scale bar, 20 µm. (b) Confocal images of microglia / macrophage engulfment of myelin debris following demyelination at 1dpt. Arrowheads highlight the location of myelin debris engulfment. Scale bar, 20 µm. (c) Quantification of the number of mpeg+ve cells (macrophages / microglia) in a 4-somite section of the spinal cord at pre-treatment (mean = 1.38 ± 0.74 SD), 1dpt (mean = 2.14 ± 1.07 SD), 2dpt (mean = 3.14 ± 0.38 SD), 3dpt (mean = 2.57 ± 1.90 SD) and 4dpt (mean = 3.00 ± 1.29 SD) (where treatment was a DMSO control). Pre-t vs 1dpt p = 0.7196, pre-t vs 2dpt p = 0.0505, pre-t vs 3dpt p = 0.3106, pre-t vs 4dpt p = 0.0844, 1dpt vs 2dpt p = 0.5190, 1dpt vs 3dpt p = 0.9597, 1dpt vs 4dpt p = 0.6588, 2dpt vs 3dpt p = 0.8930, 2dpt vs 4dpt p = 0.9994, 3dpt vs 4dpt p = 0.9597. Ordinary one-way ANOVA with Tukey’s multiple comparison test. Pre-treatment N = 8 zebrafish, 1–4dpt N = 7 zebrafish. Data are presented as mean ± SD. (D) Quantification of the number of mpeg+ve cells (macrophages / microglia) in a 4-somite section of the spinal cord at pre-treatment (mean = 1.56 ± 1.01 SD), 1dpt (mean = 7.88 ± 2.75 SD), 2dpt (mean = 10.38 ± 3.42 SD), 3dpt (mean = 9.12 ± 2.48 SD) and 4dpt (mean = 6.63 ± 1.92 SD) (where treatment was a demyelinating csn treatment). Pre-t vs 1dpt p < 0.0001, pre-t vs 2dpt p < 0.0001, pre-t vs 3dpt p < 0.0001, pre-t vs 4dpt p = 0.0011, 1dpt vs 2dpt p = 0.2586, 1dpt vs 3dpt p = 0.8394, 1dpt vs 4dpt p = 0.8394, 2dpt vs 3dpt p = 0.8394, 2dpt vs 4dpt p = 0.0294, 3dpt vs 4dpt p = 0.2586. Ordinary one-way ANOVA with Tukey’s multiple comparison test. Pre-treatment N = 9 zebrafish, 1–4dpt N = 8 zebrafish. Data are presented as mean ± SD.

Extended Data Fig. 5 ROCK inhibitor treatment further increases myelin mistargeting by surviving oligodendrocytes in the Tg(mbp:TRPV1-tagRFPt) zebrafish model.

(a) Confocal images of single oligodendrocytes which have not undergone demyelination treated with DMSO (control) or Y27632 ROCK inhibitor and imaged at 4dpf. Scale bar, 20 µm. (b) Confocal images of single surviving oligodendrocytes followed over time from prior to demyelination (csn treatment) at 4dpf through to 3dpt. Following demyelination oligodendrocytes were treated with either DMSO (control) or Y27632 ROCK inhibitor. Scale bars, 20 µm. (c) Quantification of the number of sheaths produced in oligodendrocytes which have not been demyelinated in control (mean = 16.08 ± 5.73 SD) and Y27632 ROCK inhibitor (mean = 19.35 ± 4.08 SD) treated zebrafish, p = 0.0386. Unpaired two-tailed t-test. N = 24 oligodendrocytes from 24 zebrafish (control), N = 20 oligodendrocytes from 20 zebrafish (Y27632). Data are presented as mean ± SD. (d) Quantification of the average sheath length (µm) produced in oligodendrocytes which have not been demyelinated in control (mean = 28.62 ± 7.18 SD) and Y27632 ROCK inhibitor (mean = 27.26 ± 6.91 SD) treated zebrafish, p = 0.5308. Unpaired two-tailed t-test. N = 24 oligodendrocytes from 24 zebrafish (control), N = 20 oligodendrocytes from 20 zebrafish (Y27632). Data are presented as mean ± SD. (e) Quantification of the number of mistargeted myelin profiles produced in oligodendrocytes which have not been demyelinated in control (median = 0.00, IQR = 0.00-0.00) and Y27632 ROCK inhibitor (median = 0.00, IQR = 0.00-0.00) treated zebrafish, p > 0.9999. Two-tailed Mann-Whitney test. N = 24 oligodendrocytes from 24 zebrafish (control), N = 20 oligodendrocytes from 20 zebrafish (Y27632). Data are presented as median with IQR. (f) Quantification of the number of sheaths produced per oligodendrocyte following demyelination in control (median = 3.00, IQR=1.50-3.00) and Y27632 ROCK inhibitor (median = 4.00, IQR = 1.00-4.50) treated zebrafish, p = 0.5964. Two-tailed Mann-Whitney test. N = 5 oligodendrocytes from 5 zebrafish (control), N = 9 oligodendrocytes from 9 zebrafish (Y27632). Data are presented as median with IQR. (g) Quantification of the number of mistargeted myelin profiles produced per oligodendrocyte following demyelination in control (median = 1.00, IQR = 1.00-2.00) and Y27632 ROCK inhibitor (median = 3.00, IQR = 1.50-4.75) treated zebrafish, p = 0.0414. Unpaired two tailed t-test with Welch’s correction. N = 5 oligodendrocytes from 5 zebrafish (control), N = 8 oligodendrocytes from 8 zebrafish (Y27632). Data are presented as mean ± SD.

Extended Data Fig. 6 Mistargeted myelin profiles are present in remyelinating lesions in motor cortex tissue from people with MS.

(a) Low magnification image of chromogenic immunohistochemistry for proteolipid protein (PLP - brown) and NeuN (blue) in human MS motor cortex. Outline of quantified areas shown with lesion area highlighted in red, perilesion area highlighted in green, normal appearing grey matter (NAGM) in purple and white matter indicated by ‘WM’ in white. Images 1 and 2 show examples of quantified areas in 2 different human MS motor cortex samples. Scale bars, 2000 µm. (B and C) High magnification images of chromogenic immunohistochemistry for proteolipid protein (PLP - brown) and NeuN (blue) in human MS motor cortex. (b) Images 1-9 show example images of PLP+ve wrapped NeuN+ve cells (myelinated neuronal cell bodies). Images 1 and 3-9 scale bars, 20 µm. Image 2 scale bar, 10 µm. (c) Images 1-5 show example images of PLP + ve wrapped NeuN-ve cells (oligodendrocytes). Scale bars, 20 µm. (d) Fluorescent immunohistochemistry for NeuN (white), PLP (red) and Hoechst (nuclei-blue) in human MS motor cortex. Arrows indicate the location of PLP + ve wrapped NeuN+ve Hoechst+ve cells (myelinated neuronal cell body). Scale bar, 20 µm. (e) Images 1-5 show example images of CNPase+ve wrapped NeuN+ve cells (myelinated neuronal cell bodies). Scale bars, 20 µm. (f) Fluorescent immunohistochemistry for NeuN (green), CNPase (magenta) and Hoechst (nuclei-blue) in human MS motor cortex. Arrows indicate the location of CNPase+ve wrapped NeuN+ve Hoechst+ve cells. Scale bar, 20 µm.

Extended Data Fig. 7 Extensive remyelination by newly generated oligodendrocytes in the Tg(mbp:TRPV1-tagRFPt) zebrafish model.

(a) Confocal images of csn treated zebrafish with oligodendrocytes newly generated after demyelination. Arrows show position of oligodendrocyte cell bodies. Scale bars, 20 µm. (b) Quantification of the total myelin produced per oligodendrocyte (calculated by multiplying number of sheaths per oligodendrocyte by the average sheath length per oligodendrocyte) (mean = 521.5 ± 138.30 SD), versus the same oligodendrocytes 3dpt (mean = 47.18 ± 26.57 SD) and by newly differentiated oligodendrocytes at 3dpt (mean = 491.80 ± 199.10 SD). Pre-treatment versus surviving p < 0.0001, pre-treatment versus newly differentiated p = 0.8271, surviving versus newly differentiated p < 0.0001. Ordinary one-way ANOVA with Tukey’s multiple comparison test. N = 15 oligodendrocytes from 15 zebrafish (pre-treatment and surviving). N = 20 oligodendrocytes from 11 zebrafish (newly differentiated). Data are presented as mean ± SD.

Extended Data Fig. 8 Summary Schematic.

Summary schematic outlining the responses of oligodendrocytes which survive demyelination and those newly generated after demyelination made using Biorender.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Neely, S.A., Williamson, J.M., Klingseisen, A. et al. New oligodendrocytes exhibit more abundant and accurate myelin regeneration than those that survive demyelination. Nat Neurosci 25, 415–420 (2022). https://doi.org/10.1038/s41593-021-01009-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-021-01009-x

  • Springer Nature America, Inc.

This article is cited by

Navigation