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.

  • Letter
  • Published:

A reversible form of axon damage in experimental autoimmune encephalomyelitis and multiple sclerosis

Abstract

In multiple sclerosis, a common inflammatory disease of the central nervous system, immune-mediated axon damage is responsible for permanent neurological deficits1,2. How axon damage is initiated is not known. Here we use in vivo imaging to identify a previously undescribed variant of axon damage in a mouse model of multiple sclerosis. This process, termed 'focal axonal degeneration' (FAD), is characterized by sequential stages, beginning with focal swellings and progressing to axon fragmentation. Notably, most swollen axons persist unchanged for several days, and some recover spontaneously. Early stages of FAD can be observed in axons with intact myelin sheaths. Thus, contrary to the classical view2,3,4,5,6, demyelination—a hallmark of multiple sclerosis—is not a prerequisite for axon damage. Instead, focal intra-axonal mitochondrial pathology is the earliest ultrastructural sign of damage, and it precedes changes in axon morphology. Molecular imaging and pharmacological experiments show that macrophage-derived reactive oxygen and nitrogen species (ROS and RNS) can trigger mitochondrial pathology and initiate FAD. Indeed, neutralization of ROS and RNS rescues axons that have already entered the degenerative process. Finally, axonal changes consistent with FAD can be detected in acute human multiple sclerosis lesions. In summary, our data suggest that inflammatory axon damage might be spontaneously reversible and thus a potential target for therapy.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: In vivo imaging of FAD.
Figure 2: Early FAD stages show mitochondrial alterations but no demyelination.
Figure 3: Activated macrophage/microglia-derived reactive species induce FAD.
Figure 4: Axonal changes consistent with FAD are present in acute human multiple sclerosis lesions.

Similar content being viewed by others

References

  1. Hauser, S.L. & Oksenberg, J.R. The neurobiology of multiple sclerosis: genes, inflammation, and neurodegeneration. Neuron 52, 61–76 (2006).

    Article  CAS  Google Scholar 

  2. Trapp, B.D. & Nave, K.A. Multiple sclerosis: an immune or neurodegenerative disorder? Annu. Rev. Neurosci. 31, 247–269 (2008).

    Article  CAS  Google Scholar 

  3. Trapp, B.D. et al. Axonal transection in the lesions of multiple sclerosis. N. Engl. J. Med. 338, 278–285 (1998).

    Article  CAS  Google Scholar 

  4. Lucchinetti, C. et al. Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann. Neurol. 47, 707–17 (2000).

    Article  CAS  Google Scholar 

  5. Trapp, B.D. & Stys, P.K. Virtual hypoxia and chronic necrosis of demyelinated axons in multiple sclerosis. Lancet Neurol. 8, 280–291 (2009).

    Article  CAS  Google Scholar 

  6. Waxman, S.G. Axonal conduction and injury in multiple sclerosis: the role of sodium channels. Nat. Rev. Neurosci. 7, 932–941 (2006).

    Article  CAS  Google Scholar 

  7. Pitt, D., Werner, P. & Raine, C.S. Glutamate excitotoxicity in a model of multiple sclerosis. Nat. Med. 6, 67–70 (2000).

    Article  CAS  Google Scholar 

  8. Medana, I., Martinic, M.A., Wekerle, H. & Neumann, H. Transection of major histocompatibility complex class I-induced neurites by cytotoxic T lymphocytes. Am. J. Pathol. 159, 809–815 (2001).

    Article  CAS  Google Scholar 

  9. Smith, K.J., Kapoor, R., Hall, S.M. & Davies, M. Electrically active axons degenerate when exposed to nitric oxide. Ann. Neurol. 49, 470–476 (2001).

    Article  CAS  Google Scholar 

  10. Lo, A.C., Saab, C.J., Black, J.A. & Waxman, S.G. Phenytoin protects spinal cord axons and preserves axonal conduction and neurological function in a model of neuroinflammation in vivo. J. Neurophysiol. 90, 3566–3571 (2003).

    Article  CAS  Google Scholar 

  11. Friese, M.A. et al. Acid-sensing ion channel-1 contributes to axonal degeneration in autoimmune inflammation of the central nervous system. Nat. Med. 13, 1483–1489 (2007).

    Article  CAS  Google Scholar 

  12. Mathey, E.K. et al. Neurofascin as a novel target for autoantibody-mediated axonal injury. J. Exp. Med. 204, 2363–2372 (2007).

    Article  CAS  Google Scholar 

  13. Bittner, S. et al. TASK1 modulates inflammation and neurodegeneration in autoimmune inflammation of the central nervous system. Brain 132, 2501–2516 (2009).

    Article  Google Scholar 

  14. Kerschensteiner, M., Schwab, M.E., Lichtman, J.W. & Misgeld, T. In vivo imaging of axonal degeneration and regeneration in the injured spinal cord. Nat. Med. 11, 572–577 (2005).

    Article  CAS  Google Scholar 

  15. Misgeld, T. & Kerschensteiner, M. In vivo imaging of the diseased nervous system. Nat. Rev. Neurosci. 7, 449–463 (2006).

    Article  CAS  Google Scholar 

  16. Bartholomäus, I. et al. Effector T cell interactions with meningeal vascular structure in nascent autoimmune CNS lesions. Nature 462, 94–98 (2009).

    Article  Google Scholar 

  17. Siffrin, V. et al. In vivo imaging of partially reversible TH17 cell–induced neuronal dysfunction in the course of encephalomyelitis. Immunity 33, 424–436 (2010).

    Article  CAS  Google Scholar 

  18. Feng, G. et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron 28, 41–51 (2000).

    Article  CAS  Google Scholar 

  19. Misgeld, T., Kerschensteiner, M., Bareyre, F.M., Burgess, R.W. & Lichtman, J.W. Imaging axonal transport of mitochondria in vivo. Nat. Methods 4, 559–561 (2007).

    Article  CAS  Google Scholar 

  20. Singbartl, K. et al. A CD2-green fluorescence protein–transgenic mouse reveals very late antigen-4–dependent CD8+ lymphocyte rolling in inflamed venules. J. Immunol. 166, 7520–7526 (2001).

    Article  CAS  Google Scholar 

  21. 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 

  22. Kapoor, R., Davies, M., Blaker, P.A., Hall, S.M. & Smith, K.J. Blockers of sodium and calcium entry protect axons from nitric oxide-mediated degeneration. Ann. Neurol. 53, 174–180 (2003).

    Article  CAS  Google Scholar 

  23. Smith, K.J. & Lassmann, H. The role of nitric oxide in multiple sclerosis. Lancet Neurol. 1, 232–241 (2002).

    Article  CAS  Google Scholar 

  24. Lin, M.T. & Beal, M.F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443, 787–795 (2006).

    Article  CAS  Google Scholar 

  25. Coleman, M. Axon degeneration mechanisms: commonality amid diversity. Nat. Rev. Neurosci. 6, 889–898 (2005).

    Article  CAS  Google Scholar 

  26. Franklin, R.J. & Ffrench-Constant, C. Remyelination in the CNS: from biology to therapy. Nat. Rev. Neurosci. 9, 839–855 (2008).

    Article  CAS  Google Scholar 

  27. Coman, I. et al. Nodal, paranodal and juxtaparanodal axonal proteins during demyelination and remyelination in multiple sclerosis. Brain 129, 3186–3195 (2006).

    Article  CAS  Google Scholar 

  28. Aboul-Enein, F., Weiser, P., Höftberger, R., Lassmann, H. & Bradl, M. Transient axonal injury in the absence of demyelination: a correlate of clinical disease in acute experimental autoimmune encephalomyelitis. Acta Neuropathol. 111, 539–547 (2006).

    Article  Google Scholar 

  29. Das Sarma, J., Kenyon, L.C., Hingley, S.T. & Shindler, K.S. Mechanisms of primary axonal damage in a viral model of multiple sclerosis. J. Neurosci. 29, 10272–10280 (2009).

    Article  CAS  Google Scholar 

  30. Kroemer, G., Galluzzi, L. & Brenner, C. Mitochondrial membrane permeabilization in cell death. Physiol. Rev. 87, 99–163 (2007).

    Article  CAS  Google Scholar 

  31. Dutta, R. et al. Mitochondrial dysfunction as a cause of axonal degeneration in multiple sclerosis patients. Ann. Neurol. 59, 478–489 (2006).

    Article  CAS  Google Scholar 

  32. Mahad, D., Ziabreva, I., Lassmann, H. & Turnbull, D. Mitochondrial defects in acute multiple sclerosis lesions. Brain 131, 1722–1735 (2008).

    Article  Google Scholar 

  33. Mahad, D.J. et al. Mitochondrial changes within axons in multiple sclerosis. Brain 132, 1161–1174 (2009).

    Article  Google Scholar 

  34. Forte, M. et al. Cyclophilin D inactivation protects axons in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. Proc. Natl. Acad. Sci. USA 104, 7558–7563 (2007).

    Article  CAS  Google Scholar 

  35. Du, H. et al. Cyclophilin D deficiency attenuates mitochondrial and neuronal perturbation and ameliorates learning and memory in Alzheimer′s disease. Nat. Med. 14, 1097–1105 (2008).

    Article  CAS  Google Scholar 

  36. D'Autréaux, B. & Toledano, M.B. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 8, 813–824 (2007).

    Article  Google Scholar 

  37. Hooper, D.C. et al. Uric acid, a peroxynitrite scavenger, inhibits CNS inflammation, blood-CNS barrier permeability changes and tissue damage in a mouse model of multiple sclerosis. FASEB J. 14, 691–698 (2000).

    Article  CAS  Google Scholar 

  38. Kizelsztein, P., Ovadia, H., Garbuzenko, O., Sigal, A. & Barenholz, Y. Pegylated nanoliposomes remote-loaded with the antioxidant tempamibe ameliorate experimental autoimmune encephalomyelitis. J. Neuroimmunol. 213, 20–25 (2009).

    Article  CAS  Google Scholar 

  39. Lipton, S.A. Pathologically activated therapeutics for neuroprotection. Nat. Rev. Neurosci. 8, 803–808 (2007).

    Article  CAS  Google Scholar 

  40. Abdul-Majid, K.B. et al. Screening of several H-2 congenic mouse strains identified H-2(q) mice as highly susceptible to MOG-induced EAE with minimal adjuvant requirement. J. Neuroimmunol. 111, 23–33 (2000).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank B. Fiedler, G. Heitmann, M. Schedensack, A. Schmalz and S. Knecht for excellent technical assistance; D. Matzek for animal husbandry; A. Dagkalis for help with immunizations; M. Krumbholz for advice on statistical analysis; and R. Hohlfeld, H. Wekerle, J. Sanes, J. Lichtman, L. Godinho, D. Kerschensteiner, P. Williams, T. Dick, E. Meinl and K. Dornmair for discussions or critical reading of the manuscript. Work in M. Kerschensteiner's laboratory is financed through grants from the Deutsche Forschungsgemeinschaft (DFG; Emmy Noether Program and Sonderforschungsbereich 571) and the 'Verein Therapieforschung für MS-Kranke e.V'. M. Kerschensteiner and W.B. are supported by a grant from the German Federal Ministry of Education and Research (Competence Network Multiple Sclerosis). T.M. is supported by the Institute for Advanced Study, Technische Universität München, by the Alexander von Humboldt Foundation and by the Center for Integrated Protein Science (Munich). D.M. and W.B. are supported by grants from the DFG (Sonderforschungsbereich Transregio 43). D.M. is supported by the Swiss National Science Foundation (PP00P3 128372). D.B. is supported by the US National Institutes of Health. This project was further financed by grants to M. Kerschensteiner and T.M. from the Dana Foundation and the Hertie Foundation, and by a grant from the Christopher and Dana Reeve Foundation to T.M. and D.B.

Author information

Authors and Affiliations

Authors

Contributions

M. Kerschensteiner, T.M., D.B., D.M. and I.N. conceived the experiments. I.N. and C.S. did the imaging experiments. I.N., C.S., T.M. and M. Kerschensteiner did image analysis. M.B. and D.B. did and evaluated serial electron microscopy. I.N. and F.M.B. did therapy experiments. D.M., M. Kreutzfeldt and W.B. did histopathological evaluations of EAE and multiple sclerosis tissue. I.N., M. Kerschensteiner and T.M. wrote the paper.

Corresponding authors

Correspondence to Thomas Misgeld or Martin Kerschensteiner.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 and Supplementary Methods (PDF 894 kb)

Supplementary Video 1

In vivo multi-photon time-lapse that illustrates the degeneration of a transgenically labeled stage 1 axon (white) in an acute EAE lesion in a Thy1-CFP-S × Cx3cr1GFP/+ mouse. Axonal degeneration is initiated near a putative node of Ranvier in close proximity to activated macrophages/microglia (magenta). (MOV 409 kb)

Supplementary Video 2

In vivo overview multi-photon time-lapse of the healthy lumbar spinal cord of a Thy1-YFP-16Thy1-MitoCFP-P) mouse in which axons are labeled with YFP (gray). The video illustrates that no obvious morphological changes are induced by our imaging approach. 300 min, 11 frames. (MOV 1533 kb)

Supplementary Video 3

In vivo overview multi-photon time-lapse of the lumbar spinal cord of a Thy1-YFP-16Thy1-MitoCFP-P) mouse, in which axons are labeled with YFP (gray; in some axons, CFP-labeled mitochondria are visible due to spectral cross-talk) 2 d after the EAE onset. The video illustrates stage 1 to stage 2 transitions in three axons during the observation period. 300 min, 11 frames. (MOV 2253 kb)

Supplementary Video 4

In vivo multi-photon time-lapse of the lumbar spinal cord of a Thy1-YFP-16Thy1-MitoCFP-P) mouse in which the axons are labeled with YFP (gray) 3 d after the EAE onset. This video illustrates the recovery of a stage 1 axon during the observation period. 330 min, 11 frames. (MOV 263 kb)

Supplementary Video 5

Video sequence of a stage 1 EAE axon (shown in Fig. 2a–d) that illustrates the correlation between in vivo multi-photon imaging and ssTEM. (MOV 3002 kb)

Supplementary Video 6

In vivo multi-photon microscopy time-lapse of an activated macrophage/microglia (one cell was manually pseudo-colored in magenta based on GFP expression) in apposition to an axon (white) in an acute EAE lesion in a Cx3cr1GFP/+ × Thy1-CFP-S mouse. The video illustrates how immune cells tracts were generated from time-lapse sequences. Note transition of the apposed axon from stage 0 to stage 1 during the time-lapse. Asterisk in first frame marks additional macrophage/microglia. 192 min, 20 frames. (MOV 180 kb)

Supplementary Video 7

In vivo multi-photon time-lapse of a T cell (one cell was manually pseudo-colored in cyan based on GFP expression) in apposition to an axon (white) in an acute EAE lesion in a Thy1-CFP-S × Cd2-GFP mouse. The video illustrates how immune cells tracts were generated from time-lapse sequences. Green asterisk in first frame marks additional T cell, gray asterisk marks axon fragment. 40 min, 27 frames. (MOV 249 kb)

Supplementary Video 8

In vivo multi-photon time-lapse of axonal (white) and mitochondrial (cyan) changes induced after application of H2O2 (330 mM) to the spinal cord of a Thy1-YFP-16 × Thy1-MitoCFP-P mouse. Note that axonal mitochondria change before the transition of the axons from stage 0 to stage 1. (MOV 1088 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nikić, I., Merkler, D., Sorbara, C. et al. A reversible form of axon damage in experimental autoimmune encephalomyelitis and multiple sclerosis. Nat Med 17, 495–499 (2011). https://doi.org/10.1038/nm.2324

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2324

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