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:

EPHA4 is a disease modifier of amyotrophic lateral sclerosis in animal models and in humans

Abstract

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting motor neurons. Disease onset and progression are variable, with survival ranging from months to decades. Factors underlying this variability may represent targets for therapeutic intervention. Here, we have screened a zebrafish model of ALS and identified Epha4, a receptor in the ephrin axonal repellent system, as a modifier of the disease phenotype in fish, rodents and humans. Genetic as well as pharmacological inhibition of Epha4 signaling rescues the mutant SOD1 phenotype in zebrafish and increases survival in mouse and rat models of ALS. Motor neurons that are most vulnerable to degeneration in ALS express higher levels of Epha4, and neuromuscular re-innervation by axotomized motor neurons is inhibited by the presence of Epha4. In humans with ALS, EPHA4 expression inversely correlates with disease onset and survival, and loss-of-function mutations in EPHA4 are associated with long survival. Furthermore, we found that knockdown of Epha4 also rescues the axonopathy induced by expression of mutant TAR DNA-binding protein 43 (TDP-43), another protein causing familial ALS, and the axonopathy induced by knockdown of survival of motor neuron 1, a model for spinomuscular atrophy. This suggests that Epha4 generically modulates the vulnerability of (motor) neurons to axonal degeneration and may represent a new target for therapeutic intervention.

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: Epha4 knockdown rescues a mutant SOD1–induced motor axonopathy.
Figure 2: Genetic knockdown and pharmacological inhibition of Epha4 attenuates disease.
Figure 3: Vulnerable motor neurons in ALS have higher expression of Epha4.
Figure 4: EPHA4 attenuates disease progression in humans with ALS and in motor axonopathies induced by overexpression of mutant TDP-43 and knockdown of Smn1.

Similar content being viewed by others

References

  1. Rosen, D.R. et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 362, 59–62 (1993).

    Article  CAS  Google Scholar 

  2. DeJesus-Hernandez, M. et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72, 245–256 (2011).

    Article  CAS  Google Scholar 

  3. Renton, A.E. et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72, 257–268 (2011).

    Article  CAS  Google Scholar 

  4. Penco, S. et al. Phenotypic heterogeneity in a SOD1 G93D Italian ALS family: an example of human model to study a complex disease. J. Mol. Neurosci. 44, 25–30 (2011).

    Article  CAS  Google Scholar 

  5. Kim, W. et al. Anticipation and phenotypic heterogeneity in Korean familial amyotrophic lateral sclerosis with superoxide dismutase 1 gene mutation. J. Clin. Neurol. 3, 38–44 (2007).

    Article  Google Scholar 

  6. Fogh, I. et al. Age at onset in sod1-mediated amyotrophic lateral sclerosis shows familiality. Neurogenetics 8, 235–236 (2007).

    Article  Google Scholar 

  7. Lemmens, R. et al. Overexpression of mutant superoxide dismutase 1 causes a motor axonopathy in the zebrafish. Hum. Mol. Genet. 16, 2359–2365 (2007).

    Article  CAS  Google Scholar 

  8. Laird, A.S. et al. Progranulin is neurotrophic in vivo and protects against a mutant TDP-43 induced axonopathy. PLoS ONE 5, e13368 (2010).

    Article  Google Scholar 

  9. Cooke, J.E., Kemp, H.A. & Moens, C.B. EphA4 is required for cell adhesion and rhombomere-boundary formation in the zebrafish. Curr. Biol. 15, 536–542 (2005).

    Article  CAS  Google Scholar 

  10. Xu, Q., Alldus, G., Holder, N. & Wilkinson, D.G. Expression of truncated Sek-1 receptor tyrosine kinase disrupts the segmental restriction of gene expression in the Xenopus and zebrafish hindbrain. Development 121, 4005–4016 (1995).

    CAS  Google Scholar 

  11. Klein, R. Bidirectional modulation of synaptic functions by Eph/ephrin signaling. Nat. Neurosci. 12, 15–20 (2009).

    Article  CAS  Google Scholar 

  12. Dottori, M. et al. EphA4 (Sek1) receptor tyrosine kinase is required for the development of the corticospinal tract. Proc. Natl. Acad. Sci. USA 95, 13248–13253 (1998).

    Article  CAS  Google Scholar 

  13. Kullander, K. et al. Role of EphA4 and EphrinB3 in local neuronal circuits that control walking. Science 299, 1889–1892 (2003).

    Article  CAS  Google Scholar 

  14. Noberini, R. et al. Small molecules can selectively inhibit ephrin binding to the EphA4 and EphA2 receptors. J. Biol. Chem. 283, 29461–29472 (2008).

    Article  CAS  Google Scholar 

  15. Goldshmit, Y., Galea, M.P., Wise, G., Bartlett, P.F. & Turnley, A.M. Axonal regeneration and lack of astrocytic gliosis in EphA4-deficient mice. J. Neurosci. 24, 10064–10073 (2004).

    Article  CAS  Google Scholar 

  16. Fabes, J., Anderson, P., Brennan, C. & Bolsover, S. Regeneration-enhancing effects of EphA4 blocking peptide following corticospinal tract injury in adult rat spinal cord. Eur. J. Neurosci. 26, 2496–2505 (2007).

    Article  Google Scholar 

  17. Goldshmit, Y. et al. EphA4 blockers promote axonal regeneration and functional recovery following spinal cord injury in mice. PLoS ONE 6, e24636 (2011).

    Article  CAS  Google Scholar 

  18. Filosa, A. et al. Neuron-glia communication via EphA4/ephrin-A3 modulates LTP through glial glutamate transport. Nat. Neurosci. 12, 1285–1292 (2009).

    Article  CAS  Google Scholar 

  19. Rothstein, J.D., Martin, L.J. & Kuncl, R.W. Decreased glutamate transport by the brain and spinal cord in amyotrophic lateral sclerosis. N. Engl. J. Med. 326, 1464–1468 (1992).

    Article  CAS  Google Scholar 

  20. Rothstein, J.D., Van Kammen, M., Levey, A.I., Martin, L.J. & Kuncl, R.W. Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis. Ann. Neurol. 38, 73–84 (1995).

    Article  CAS  Google Scholar 

  21. Rothstein, J.D. et al. β-lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature 433, 73–77 (2005).

    Article  CAS  Google Scholar 

  22. Bakels, R. & Kernell, D. Matching between motoneurone and muscle unit properties in rat medial gastrocnemius. J. Physiol. (Lond.) 463, 307–324 (1993).

    Article  CAS  Google Scholar 

  23. Gardiner, P.F. Physiological properties of motoneurons innervating different muscle unit types in rat gastrocnemius. J. Neurophysiol. 69, 1160–1170 (1993).

    Article  CAS  Google Scholar 

  24. Frey, D. et al. Early and selective loss of neuromuscular synapse subtypes with low sprouting competence in motoneuron diseases. J. Neurosci. 20, 2534–2542 (2000).

    Article  CAS  Google Scholar 

  25. Pun, S., Santos, A.F., Saxena, S., Xu, L. & Caroni, P. Selective vulnerability and pruning of phasic motoneuron axons in motoneuron disease alleviated by CNTF. Nat. Neurosci. 9, 408–419 (2006).

    Article  CAS  Google Scholar 

  26. Maes, O.C. et al. Transcriptional profiling of Alzheimer blood mononuclear cells by microarray. Neurobiol. Aging 28, 1795–1809 (2007).

    Article  CAS  Google Scholar 

  27. Borovecki, F. et al. Genome-wide expression profiling of human blood reveals biomarkers for Huntington's disease. Proc. Natl. Acad. Sci. USA 102, 11023–11028 (2005).

    Article  CAS  Google Scholar 

  28. Gladkevich, A., Kauffman, H.F. & Korf, J. Lymphocytes as a neural probe: potential for studying psychiatric disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 28, 559–576 (2004).

    Article  Google Scholar 

  29. Glatt, S.J. et al. Comparative gene expression analysis of blood and brain provides concurrent validation of SELENBP1 up-regulation in schizophrenia. Proc. Natl. Acad. Sci. USA 102, 15533–15538 (2005).

    Article  CAS  Google Scholar 

  30. Matigian, N.A. et al. Fibroblast and lymphoblast gene expression profiles in schizophrenia: are non-neural cells informative? PLoS ONE 3, e2412 (2008).

    Article  Google Scholar 

  31. Tsuang, M.T. et al. Assessing the validity of blood-based gene expression profiles for the classification of schizophrenia and bipolar disorder: a preliminary report. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 133B, 1–5 (2005).

    Article  Google Scholar 

  32. Fu, J. et al. Unraveling the regulatory mechanisms underlying tissue-dependent genetic variation of gene expression. PLoS Genet. 8, e1002431 (2012).

    Article  CAS  Google Scholar 

  33. Sreedharan, J. et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 319, 1668–1672 (2008).

    Article  CAS  Google Scholar 

  34. Neumann, M. et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314, 130–133 (2006).

    Article  CAS  Google Scholar 

  35. Gitcho, M.A. et al. TDP-43 A315T mutation in familial motor neuron disease. Ann. Neurol. 63, 535–538 (2008).

    Article  CAS  Google Scholar 

  36. McWhorter, M.L., Monani, U.R., Burghes, A.H. & Beattie, C.E. Knockdown of the survival motor neuron (Smn) protein in zebrafish causes defects in motor axon outgrowth and pathfinding. J. Cell Biol. 162, 919–931 (2003).

    Article  CAS  Google Scholar 

  37. Storkebaum, E. et al. Treatment of motoneuron degeneration by intracerebroventricular delivery of VEGF in a rat model of ALS. Nat. Neurosci. 8, 85–92 (2005).

    Article  CAS  Google Scholar 

  38. Cashman, N.R. et al. Neuroblastoma x spinal cord (NSC) hybrid cell lines resemble developing motor neurons. Dev. Dyn. 194, 209–221 (1992).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work has been supported by grants from the Flanders Institute for Biotechnology (VIB), the University of Leuven (GOA 11/014), Life Sciences Research Partners, Research Foundation Flanders (FWO-Vlaanderen; G.0695.10), the Interuniversity Attraction Poles program P7/16 of the Belgian Federal Science Policy Office, the Robert Packard Center for ALS Research, the Association Belge contre les Maladies Musculaires, the ALS League Belgium and the Thierry Latran Foundation. The research leading to these results has received funding from the European Community's Health Seventh Framework Programme (FP7/2007-2013) under grant agreement number 259867. W.R. is supported by the E. von Behring Chair for Neuromuscular and Neurodegenerative Disorders at the University of Leuven. A.V.H. is supported by the Agency for Innovation by Science and Technology in Flanders (IWT-Vlaanderen). R.L., P.V.D., V.T. and B.D. hold a clinical investigatorship of FWO-Vlaanderen. R.L. is supported by Research Fund KU Leuven. P.G.-P. is supported by the Alfonso Martin Escudero Foundation. A.A.-C. receives salary support from the UK National Institute for Health Research (NIHR) Dementia Biomedical Research Unit at South London and Maudsley National Health Service (NHS) Foundation Trust and King's College London. The views expressed are those of the authors and not necessarily those of the NHS, the NIHR or the Department of Health. R.H.B. Jr. is supported by the US National Institute for Neurological Disease and Stroke (NINDS) (5RO1-NS050557-05) and NINDS American Recovery and Reinvestment Act Award RC2-NS070-342 and acknowledges generous support from the Angel Fund, the ALS Association, P2ALS, Project ALS, the Pierre L. de Bourgknecht ALS Research Foundation and the ALS Therapy Alliance.

Author information

Authors and Affiliations

Authors

Contributions

W.R. designed the study, analyzed data and wrote the manuscript. A.V.H. designed and performed experiments, analyzed data and wrote the manuscript. R.L. collected samples, analyzed data and designed experiments. P.V.D. and L.V.D.B. collected samples and designed experiments. L.S., M.T., P.G.-P., K.A.S., A.S.L., T.P., E.P., A.G. and P.W.v.V. performed experiments. P.M.A., A.A.-C., O.H., J.H.V., L.H.v.d.B., V.T., R.H.B. Jr. and B.D. collected samples and analyzed data. A.M.T. provided mice. All authors discussed the results and implications and commented on the manuscript at all stages.

Corresponding author

Correspondence to Wim Robberecht.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1–10 and Supplementary Figures 1–8 (PDF 1399 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Van Hoecke, A., Schoonaert, L., Lemmens, R. et al. EPHA4 is a disease modifier of amyotrophic lateral sclerosis in animal models and in humans. Nat Med 18, 1418–1422 (2012). https://doi.org/10.1038/nm.2901

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2901

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