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.

  • Review Article
  • Published:

Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis

Abstract

Amyotrophic lateral sclerosis (ALS) is a genetically diverse disease. At least 15 ALS-associated gene loci have so far been identified, and the causative gene is known in approximately 30% of familial ALS cases. Less is known about the factors underlying the sporadic form of the disease. The molecular mechanisms of motor neuron degeneration are best understood in the subtype of disease caused by mutations in superoxide dismutase 1, with a current consensus that motor neuron injury is caused by a complex interplay between multiple pathogenic processes. A key recent finding is that mutated TAR DNA-binding protein 43 is a major constituent of the ubiquitinated protein inclusions in ALS, providing a possible link between the genetic mutation and the cellular pathology. New insights have also indicated the importance of dysregulated glial cell–motor neuron crosstalk, and have highlighted the vulnerability of the distal axonal compartment early in the disease course. In addition, recent studies have suggested that disordered RNA processing is likely to represent a major contributing factor to motor neuron disease. Ongoing research on the cellular pathways highlighted in this Review is predicted to open the door to new therapeutic interventions to slow disease progression in ALS.

Key Points

  • Multiple cellular events contribute to the pathobiology of amyotrophic lateral sclerosis (ALS), including oxidative stress, mitochondrial dysfunction, excitotoxicity, protein aggregation, impaired axonal transport, neuroinflammation, and dysregulated RNA signaling

  • TAR DNA-binding protein 43 is a major constituent of the ubiquitinated protein inclusions found in surviving motor neurons in most forms of ALS

  • Glial pathology and disruption of glial cell–motor neuron communication contribute to neurodegeneration and the propagation of motor neuron injury

  • Understanding the links between molecular changes and clinical features of the disease should guide future therapeutic efforts

  • Degenerative changes in motor neurons seem to affect the health of the distal axonal compartment at an early stage of disease, highlighting an important neuroprotective target

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: Molecular mechanisms of motor neuron injury in ALS.
Figure 2: Mitochondrial dysfunction in ALS.
Figure 3: Emerging evidence for dysregulation of RNA processing in ALS.

Similar content being viewed by others

References

  1. Wood-Allum, C. & Shaw, P. J. Motor neurone disease: a practical update on diagnosis and management. Clin. Med. 10, 252–258 (2010).

    Google Scholar 

  2. Ince, P. G., Clark, B., Holton, J., Revesz, T. & Wharton, S. B. in Greenfield's Neuropathology (eds Love, S. et al.) 947–971 (Hodder Arnold, London, 2008).

    Google Scholar 

  3. Martin, L. J. Neuronal death in amyotrophic lateral sclerosis is apoptosis: possible contribution of a programmed cell death mechanism. J. Neuropathol. Exp. Neurol. 58, 459–471 (1999).

    CAS  PubMed  Google Scholar 

  4. Ince, P. G., Lowe, J. & Shaw, P. J. Amyotrophic lateral sclerosis: current issues in classification, pathogenesis and molecular pathology. Neuropathol. Appl. Neurobiol. 24, 104–117 (1998).

    CAS  PubMed  Google Scholar 

  5. Ince, P. G., Tomkins, J., Slade, J. Y., Thatcher, N. M. & Shaw, P. J. Amyotrophic lateral sclerosis associated with genetic abnormalities in the gene encoding Cu/Zn superoxide dismutase: molecular pathology of five new cases, and comparison with previous reports and 73 sporadic cases of ALS. J. Neuropathol. Exp. Neurol. 57, 895–904 (1998).

    CAS  PubMed  Google Scholar 

  6. Piao, Y. S. et al. Neuropathology with clinical correlations of sporadic amyotrophic lateral sclerosis: 102 autopsy cases examined between 1962 and 2000. Brain Pathol. 13, 10–22 (2003).

    PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  8. Nishihara, Y. et al. Sporadic amyotrophic lateral sclerosis: two pathological patterns shown by analysis of distribution of TDP-43 immunoreactive neuronal and glial cytoplasmic inclusions. Acta Neuropathol. 116, 169–182 (2008).

    Google Scholar 

  9. Zhang, H. et al. TDP-43 immunoreactive neuronal and glial inclusions in the neostriatum in amyotrophic lateral sclerosis with and without dementia. Acta Neuropathol. 115, 115–122 (2008).

    CAS  PubMed  Google Scholar 

  10. Ilieva, H., Polymenidou, M. & Cleveland, D. W. Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J. Cell Biol. 187, 761–772 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Kirby, J. et al. Mutant SOD1 alters the motor neuronal transcriptome: implications for familial ALS. Brain 128, 1686–1706 (2005).

    PubMed  Google Scholar 

  12. Kirby, J. et al. Phosphatase and tensin homologue/protein kinase B pathway linked to motor neuron survival in human superoxide dismutase 1-related amyotrophic lateral sclerosis. Brain 134, 506–517 (2011).

    PubMed  PubMed Central  Google Scholar 

  13. Tovar, Y. R. & Tapia, R. VEGF protects spinal motor neurons against chronic excitotoxic degeneration in vivo by activation of PI3-K pathway and inhibition of p38 MAPK. J. Neurochem. 115, 1090–1101 (2010).

    Google Scholar 

  14. Brockington, A. et al. Downregulation of genes with a function in axon outgrowth and synapse formation in motor neurones of the VEGFδ/δ mouse model of amyotrophic lateral sclerosis. BMC Genomics 11, 203 (2010).

    PubMed  PubMed Central  Google Scholar 

  15. Murray, L. M., Talbot, K. & Gillingwater, T. H. Review: neuromuscular synaptic vulnerability in motor neurone disease: amyotrophic lateral sclerosis and spinal muscular atrophy. Neuropathol. Appl. Neurobiol. 36, 133–156 (2010).

    CAS  PubMed  Google Scholar 

  16. Aggarwal, S. & Cudkowicz, M. ALS drug development: reflections of the past and a way forward. Neurotherapeutics 5, 516–527 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Schnabel, J. Neuroscience: standard model. Nature 454, 682–685 (2008).

    CAS  PubMed  Google Scholar 

  18. Schymick, J. C., Talbot, K. & Traynor, B. J. Genetics of sporadic amyotrophic lateral sclerosis. Hum. Mol. Genet. 16 (Spec. no. 2), R233–R242 (2007).

    CAS  PubMed  Google Scholar 

  19. Dunckley, T. et al. Whole-genome analysis of sporadic amyotrophic lateral sclerosis. N. Engl. J. Med. 357, 775–788 (2007).

    CAS  PubMed  Google Scholar 

  20. van Es, M. A. et al. ITPR2 as a susceptibility gene in sporadic amyotrophic lateral sclerosis: a genome-wide association study. Lancet Neurol. 6, 869–877 (2007).

    CAS  PubMed  Google Scholar 

  21. van Es, M. A. et al. Genetic variation in DPP6 is associated with susceptibility to amyotrophic lateral sclerosis. Nat. Genet. 40, 29–31 (2008).

    CAS  PubMed  Google Scholar 

  22. Laaksovirta, H. et al. Chromosome 9p21 in amyotrophic lateral sclerosis in Finland: a genome-wide association study. Lancet Neurol. 9, 978–985 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Shatunov, A. et al. Chromosome 9p21 in sporadic amyotrophic lateral sclerosis in the UK and seven other countries: a genome-wide association study. Lancet Neurol. 9, 986–994 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Hosler, B. A. et al. Linkage of familial amyotrophic lateral sclerosis with frontotemporal dementia to chromosome 9q21-q22. JAMA 284, 1664–1669 (2000).

    CAS  PubMed  Google Scholar 

  25. Elden, A. C. et al. Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 466, 1069–1075 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Andersen, P. M. & Al-Chalabi, A. Clinical genetics of amyotrophic lateral sclerosis: what do we really know? Nat. Rev. Neurol. 7, 603–615 (2011).

    CAS  PubMed  Google Scholar 

  27. Rosen, D. R. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 364, 362 (1993).

    CAS  PubMed  Google Scholar 

  28. Smith, R. G., Henry, Y. K., Mattson, M. P. & Appel, S. H. Presence of 4-hydroxynonenal in cerebrospinal fluid of patients with sporadic amyotrophic lateral sclerosis. Ann. Neurol. 44, 696–699 (1998).

    CAS  PubMed  Google Scholar 

  29. Simpson, E. P., Henry, Y. K., Henkel, J. S., Smith, R. G. & Appel, S. H. Increased lipid peroxidation in sera of ALS patients: a potential biomarker of disease burden. Neurology 62, 1758–1765 (2004).

    CAS  PubMed  Google Scholar 

  30. Mitsumoto, H. et al. Oxidative stress biomarkers in sporadic ALS. Amyotroph. Lateral Scler. 9, 177–183 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Shaw, P. J., Ince, P. G., Falkous, G. & Mantle, D. Oxidative damage to protein in sporadic motor neuron disease spinal cord. Ann. Neurol. 38, 691–695 (1995).

    CAS  PubMed  Google Scholar 

  32. Shibata, N. et al. Morphological evidence for lipid peroxidation and protein glycoxidation in spinal cords from sporadic amyotrophic lateral sclerosis patients. Brain Res. 917, 97–104 (2001).

    CAS  PubMed  Google Scholar 

  33. Fitzmaurice, P. S. et al. Evidence for DNA damage in amyotrophic lateral sclerosis. Muscle Nerve 19, 797–798 (1996).

    CAS  PubMed  Google Scholar 

  34. Chang, Y. et al. Messenger RNA oxidation occurs early in disease pathogenesis and promotes motor neuron degeneration in ALS. PLoS ONE 3, e2849 (2008).

    PubMed  PubMed Central  Google Scholar 

  35. Barber, S. C. & Shaw, P. J. Oxidative stress in ALS: key role in motor neuron injury and therapeutic target. Free Radic. Biol. Med. 48, 629–641 (2010).

    CAS  PubMed  Google Scholar 

  36. Andrus, P. K., Fleck, T. J., Gurney, M. E. & Hall, E. D. Protein oxidative damage in a transgenic mouse model of familial amyotrophic lateral sclerosis. J. Neurochem. 71, 2041–2048 (1998).

    CAS  PubMed  Google Scholar 

  37. Duan, W. et al. Mutant TAR DNA-binding protein-43 induces oxidative injury in motor neuron-like cell. Neuroscience 169, 1621–1629 (2010).

    CAS  PubMed  Google Scholar 

  38. Subramaniam, J. R. et al. Mutant SOD1 causes motor neuron disease independent of copper chaperone-mediated copper loading. Nat. Neurosci. 5, 301–307 (2002).

    CAS  PubMed  Google Scholar 

  39. Harraz, M. M. et al. SOD1 mutations disrupt redox-sensitive Rac regulation of NADPH oxidase in a familial ALS model. J. Clin. Invest. 118, 659–670 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Wu, D. C., Re, D. B., Nagai, M., Ischiropoulos, H. & Przedborski, S. The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice. Proc. Natl Acad. Sci. USA 103, 12132–12137 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Marden, J. J. et al. Redox modifier genes in amyotrophic lateral sclerosis in mice. J. Clin. Invest. 117, 2913–2919 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Sarlette, A. et al. Nuclear erythroid 2-related factor 2-antioxidative response element signaling pathway in motor cortex and spinal cord in amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 67, 1055–1062 (2008).

    CAS  PubMed  Google Scholar 

  43. Rao, S. D. & Weiss, J. H. Excitotoxic and oxidative cross-talk between motor neurons and glia in ALS pathogenesis. Trends Neurosci. 27, 17–23 (2004).

    CAS  PubMed  Google Scholar 

  44. Duffy, L. M. et al. The role of mitochondria in the pathogenesis of amyotrophic lateral sclerosis. Neuropathol. Appl. Neurobiol. 37, 336–352 (2011).

    CAS  PubMed  Google Scholar 

  45. Wood, J. D., Beaujeux, T. P. & Shaw, P. J. Protein aggregation in motor neurone disorders. Neuropathol. Appl. Neurobiol. 29, 529–545 (2003).

    CAS  PubMed  Google Scholar 

  46. Kanekura, K., Suzuki, H., Aiso, S. & Matsuoka, M. ER stress and unfolded protein response in amyotrophic lateral sclerosis. Mol. Neurobiol. 39, 81–89 (2009).

    CAS  PubMed  Google Scholar 

  47. Blackburn, D., Sargsyan, S., Monk, P. N. & Shaw, P. J. Astrocyte function and role in motor neuron disease: a future therapeutic target? Glia 57, 1251–1264 (2009).

    PubMed  Google Scholar 

  48. Sargsyan, S. A., Monk, P. N. & Shaw, P. J. Microglia as potential contributors to motor neuron injury in amyotrophic lateral sclerosis. Glia 51, 241–253 (2005).

    PubMed  Google Scholar 

  49. Benatar, M. Lost in translation: treatment trials in the SOD1 mouse and in human ALS. Neurobiol. Dis. 26, 1–13 (2007).

    CAS  PubMed  Google Scholar 

  50. Orrell, R. W., Lane, R. J. & Ross, M. Antioxidant treatment for amyotrophic lateral sclerosis or motor neuron disease. Cochrane Database of Systematic Reviews, Issue 1. Art. No.: CD002829. http://dx.doi.org/10.1002/14651858.CD002829.pub4 (2007).

  51. Wong, P. C. et al. An adverse property of a familial ALS-linked SOD1 mutation causes motor neuron disease characterized by vacuolar degeneration of mitochondria. Neuron 14, 1105–1116 (1995).

    CAS  PubMed  Google Scholar 

  52. Liu, J. et al. Toxicity of familial ALS-linked SOD1 mutants from selective recruitment to spinal mitochondria. Neuron 43, 5–17 (2004).

    CAS  PubMed  Google Scholar 

  53. Pasinelli, P. et al. Amyotrophic lateral sclerosis-associated SOD1 mutant proteins bind and aggregate with Bcl-2 in spinal cord mitochondria. Neuron 43, 19–30 (2004).

    CAS  PubMed  Google Scholar 

  54. Vande Velde, C., Miller, T. M., Cashman, N. R. & Cleveland, D. W. Selective association of misfolded ALS-linked mutant SOD1 with the cytoplasmic face of mitochondria. Proc. Natl Acad. Sci. USA 105, 4022–4027 (2008).

    PubMed  PubMed Central  Google Scholar 

  55. Wiedemann, F. R., Manfredi, G., Mawrin, C., Beal, M. F. & Schon, E. A. Mitochondrial DNA and respiratory chain function in spinal cords of ALS patients. J. Neurochem. 80, 616–625 (2002).

    CAS  PubMed  Google Scholar 

  56. Mattiazzi, M. et al. Mutated human SOD1 causes dysfunction of oxidative phosphorylation in mitochondria of transgenic mice. J. Biol. Chem. 277, 29626–29633 (2002).

    CAS  PubMed  Google Scholar 

  57. Damiano, M. et al. Neural mitochondrial Ca2+ capacity impairment precedes the onset of motor symptoms in G93A Cu/Zn-superoxide dismutase mutant mice. J. Neurochem. 96, 1349–1361 (2006).

    CAS  PubMed  Google Scholar 

  58. Grosskreutz, J., Van Den Bosch, L. & Keller, B. U. Calcium dysregulation in amyotrophic lateral sclerosis. Cell Calcium 47, 165–174 (2010).

    CAS  PubMed  Google Scholar 

  59. Sathasivam, S., Grierson, A. J. & Shaw, P. J. Characterization of the caspase cascade in a cell culture model of SOD1-related familial amyotrophic lateral sclerosis: expression, activation and therapeutic effects of inhibition. Neuropathol. Appl. Neurobiol. 31, 467–485 (2005).

    CAS  PubMed  Google Scholar 

  60. Sasaki, S. & Iwata, M. Mitochondrial alterations in the spinal cord of patients with sporadic amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 66, 10–16 (2007).

    PubMed  Google Scholar 

  61. Menzies, F. M. et al. Mitochondrial dysfunction in a cell culture model of familial amyotrophic lateral sclerosis. Brain 125, 1522–1533 (2002).

    PubMed  Google Scholar 

  62. De Vos, K. J. et al. Familial amyotrophic lateral sclerosis-linked SOD1 mutants perturb fast axonal transport to reduce axonal mitochondria content. Hum. Mol. Genet. 16, 2720–2728 (2007).

    CAS  PubMed  Google Scholar 

  63. De Vos, K. J., Grierson, A. J., Ackerley, S. & Miller, C. C. Role of axonal transport in neurodegenerative diseases. Annu. Rev. Neurosci. 31, 151–173 (2008).

    CAS  PubMed  Google Scholar 

  64. Bordet, T. et al. Identification and characterization of cholest-4-en-3-one, oxime (TRO19622), a novel drug candidate for amyotrophic lateral sclerosis. J. Pharmacol. Exp. Ther. 322, 709–720 (2007).

    CAS  PubMed  Google Scholar 

  65. Van Damme, P., Dewil, M., Robberecht, W. & Van Den Bosch, L. Excitotoxicity and amyotrophic lateral sclerosis. Neurodegener. Dis. 2, 147–159 (2005).

    CAS  PubMed  Google Scholar 

  66. Arundine, M. & Tymianski, M. Molecular mechanisms of calcium-dependent neurodegeneration in excitotoxicity. Cell Calcium 34, 325–337 (2003).

    CAS  PubMed  Google Scholar 

  67. Carriedo, S. G., Yin, H. Z. & Weiss, J. H. Motor neurons are selectively vulnerable to AMPA/kainate receptor-mediated injury in vitro. J. Neurosci. 16, 4069–4079 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. King, A. E. et al. Excitotoxicity mediated by non-NMDA receptors causes distal axonopathy in long-term cultured spinal motor neurons. Eur. J. Neurosci. 26, 2151–2159 (2007).

    CAS  PubMed  Google Scholar 

  69. Williams, T. L., Day, N. C., Ince, P. G., Kamboj, R. K. & Shaw, P. J. Calcium-permeable α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors: a molecular determinant of selective vulnerability in amyotrophic lateral sclerosis. Ann. Neurol. 42, 200–207 (1997).

    CAS  PubMed  Google Scholar 

  70. Ince, P. et al. Parvalbumin and calbindin D-28k in the human motor system and in motor neuron disease. Neuropathol. Appl. Neurobiol. 19, 291–299 (1993).

    CAS  PubMed  Google Scholar 

  71. Shaw, P. J., Forrest, V., Ince, P. G., Richardson, J. P. & Wastell, H. J. CSF and plasma amino acid levels in motor neuron disease: elevation of CSF glutamate in a subset of patients. Neurodegeneration 4, 209–216 (1995).

    CAS  PubMed  Google Scholar 

  72. 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).

    CAS  PubMed  Google Scholar 

  73. 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).

    CAS  PubMed  Google Scholar 

  74. Fray, A. E. et al. The expression of the glial glutamate transporter protein EAAT2 in motor neuron disease: an immunohistochemical study. Eur. J. Neurosci. 10, 2481–2489 (1998).

    CAS  PubMed  Google Scholar 

  75. Foran, E. & Trotti, D. Glutamate transporters and the excitotoxic path to motor neuron degeneration in amyotrophic lateral sclerosis. Antioxid. Redox Signal. 11, 1587–1602 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Vucic, S., Nicholson, G. A. & Kiernan, M. C. Cortical hyperexcitability may precede the onset of familial amyotrophic lateral sclerosis. Brain 131, 1540–1550 (2008).

    PubMed  Google Scholar 

  77. Vucic, S. & Kiernan, M. C. Novel threshold tracking techniques suggest that cortical hyperexcitability is an early feature of motor neuron disease. Brain 129, 2436–2446 (2006).

    PubMed  Google Scholar 

  78. Kwak, S., Hideyama, T., Yamashita, T. & Aizawa, H. AMPA receptor-mediated neuronal death in sporadic ALS. Neuropathology 30, 182–188 (2010).

    PubMed  Google Scholar 

  79. Mitchell, J. et al. Familial amyotrophic lateral sclerosis is associated with a mutation in D-amino acid oxidase. Proc. Natl Acad. Sci. USA 107, 7556–7561 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Meehan, C. F. et al. Intrinsic properties of lumbar motor neurones in the adult G127insTGGG superoxide dismutase-1 mutant mouse in vivo: evidence for increased persistent inward currents. Acta Physiol. 200, 361–376 (2010).

    CAS  Google Scholar 

  81. Boston-Howes, W. et al. Caspase-3 cleaves and inactivates the glutamate transporter EAAT2. J. Biol. Chem. 281, 14076–14084 (2006).

    CAS  PubMed  Google Scholar 

  82. Milanese, M. et al. Abnormal exocytotic release of glutamate in a mouse model of amyotrophic lateral sclerosis. J. Neurochem. 116, 1028–1042 (2011).

    CAS  PubMed  Google Scholar 

  83. Sunico, C. R. et al. Reduction in the motoneuron inhibitory/excitatory synaptic ratio in an early-symptomatic mouse model of amyotrophic lateral sclerosis. Brain Pathol. 21, 1–15 (2011).

    CAS  PubMed  Google Scholar 

  84. Van Damme, P. et al. Astrocytes regulate GluR2 expression in motor neurons and their vulnerability to excitotoxicity. Proc. Natl Acad. Sci. USA 104, 14825–14830 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Cheah, B. C., Vucic, S., Krishnan, A. V. & Kiernan, M. C. Riluzole, neuroprotection and amyotrophic lateral sclerosis. Curr. Med. Chem. 17, 1942–1959 (2010).

    CAS  PubMed  Google Scholar 

  86. Lacomblez, L., Bensimon, G., Leigh, P. N., Guillet, P. & Meininger, V. Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Amyotrophic Lateral Sclerosis/Riluzole Study Group II. Lancet 347, 1425–1431 (1996).

    CAS  PubMed  Google Scholar 

  87. Siciliano, G. et al. Clinical trials for neuroprotection in ALS. CNS Neurol. Disord. Drug Targets. 9, 305–313 (2010).

    CAS  PubMed  Google Scholar 

  88. Giordana, M. T. et al. TDP-43 redistribution is an early event in sporadic amyotrophic lateral sclerosis. Brain Pathol. 20, 351–360 (2010).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Sobue, G. et al. Phosphorylated high molecular weight neurofilament protein in lower motor neurons in amyotrophic lateral sclerosis and other neurodegenerative diseases involving ventral horn cells. Acta Neuropathol. 79, 402–408 (1990).

    CAS  PubMed  Google Scholar 

  91. Okamoto, K., Hirai, S., Amari, M., Watanabe, M. & Sakurai, A. Bunina bodies in amyotrophic lateral sclerosis immunostained with rabbit anti-cystatin C serum. Neurosci. Lett. 162, 125–128 (1993).

    CAS  PubMed  Google Scholar 

  92. Shibata, N. et al. Cu/Zn superoxide dismutase-like immunoreactivity in Lewy body-like inclusions of sporadic amyotrophic lateral sclerosis. Neurosci. Lett. 179, 149–152 (1994).

    CAS  PubMed  Google Scholar 

  93. Rakhit, R. et al. An immunological epitope selective for pathological monomer-misfolded SOD1 in ALS. Nat. Med. 13, 754–759 (2007).

    CAS  PubMed  Google Scholar 

  94. Bosco, D. A. et al. Wild-type and mutant SOD1 share an aberrant conformation and a common pathogenic pathway in ALS. Nat. Neurosci. 13, 1396–1403 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Groen, E. J. et al. FUS mutations in familial amyotrophic lateral sclerosis in the Netherlands. Arch. Neurol. 67, 224–230 (2010).

    PubMed  Google Scholar 

  96. Hewitt, C. et al. Novel FUS/TLS mutations and pathology in familial and sporadic amyotrophic lateral sclerosis. Arch. Neurol. 67, 455–461 (2010).

    PubMed  Google Scholar 

  97. Ackerley, S. et al. Glutamate slows axonal transport of neurofilaments in transfected neurons. J. Cell Biol. 150, 165–176 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Ackerley, S. et al. Neurofilament heavy chain side arm phosphorylation regulates axonal transport of neurofilaments. J. Cell Biol. 161, 489–495 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Johnson, J. O. et al. Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron 68, 857–864 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Ritson, G. P. et al. TDP-43 mediates degeneration in a novel Drosophila model of disease caused by mutations in VCP/p97. J. Neurosci. 30, 7729–7739 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Deng, H.-X. et al. Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature 477, 211–215 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Yang, Y. et al. The gene encoding alsin, a protein with three guanine-nucleotide exchange factor domains, is mutated in a form of recessive amyotrophic lateral sclerosis. Nat. Genet. 29, 160–165 (2001).

    CAS  PubMed  Google Scholar 

  103. Lai, C. et al. 2006. Amyotrophic lateral sclerosis 2-deficiency leads to neuronal degeneration in amyotrophic lateral sclerosis through altered AMPA receptor trafficking. J. Neurosci. 26, 11798–11806 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Nishimura, A. L. et al. A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. Am. J. Hum. Genet. 75, 822–831 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Maruyama, H. et al. Mutations of optineurin in amyotrophic lateral sclerosis. Nature 465, 223–226 (2010).

    CAS  PubMed  Google Scholar 

  106. Parkinson, N. et al. ALS phenotypes with mutations in CHMP2B (charged multivesicular body protein 2B). Neurology 67, 1074–1077 (2006).

    CAS  PubMed  Google Scholar 

  107. Cox, L. E. et al. Mutations in CHMP2B in lower motor neuron predominant amyotrophic lateral sclerosis (ALS). PLoS One 5, e9872 (2010).

    PubMed  PubMed Central  Google Scholar 

  108. Chow, C. Y. et al. Deleterious variants of FIG4, a phosphoinositide phosphatase, in patients with ALS. Am. J. Hum. Genet. 84, 85–88 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Michell, R. H. & Dove, S. K. A protein complex that regulates PtdIns(3,5)P2 levels. EMBO J. 28, 86–87 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Kieran, D. et al. A mutation in dynein rescues axonal transport defects and extends the life span of ALS mice. J. Cell Biol. 169, 561–567 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Williamson, T. L. & Cleveland, D. W. Slowing of axonal transport is a very early event in the toxicity of ALS-linked SOD1 mutants to motor neurons. Nat. Neurosci. 2, 50–56 (1999).

    CAS  PubMed  Google Scholar 

  112. Bilsland, L. G. et al. Deficits in axonal transport precede ALS symptoms in vivo. Proc. Natl Acad. Sci. USA 107, 20523–20528 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Miller, K. E. & Sheetz, M. P. Axonal mitochondrial transport and potential are correlated. J. Cell Sci. 117, 2791–2804 (2004).

    CAS  PubMed  Google Scholar 

  114. Kiaei, M. et al. Matrix metalloproteinase-9 regulates TNF-α and FasL expression in neuronal, glial cells and its absence extends life in a transgenic mouse model of amyotrophic lateral sclerosis. Exp. Neurol. 205, 74–81 (2007).

    CAS  PubMed  Google Scholar 

  115. De Vos, K. et al. Tumor necrosis factor induces hyperphosphorylation of kinesin light chain and inhibits kinesin-mediated transport of mitochondria. J. Cell Biol. 149, 1207–1214 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Ackerley, S. et al. p38α stress-activated protein kinase phosphorylates neurofilaments and is associated with neurofilament pathology in amyotrophic lateral sclerosis. Mol. Cell Neurosci. 26, 354–364 (2004).

    CAS  PubMed  Google Scholar 

  117. Tortarolo, M. et al. Persistent activation of p38 mitogen-activated protein kinase in a mouse model of familial amyotrophic lateral sclerosis correlates with disease progression. Mol. Cell Neurosci. 23, 180–192 (2003).

    CAS  PubMed  Google Scholar 

  118. Brownlees, J. et al. Phosphorylation of neurofilament heavy chain side-arms by stress activated protein kinase-1b/Jun N-terminal kinase-3. J. Cell Sci. 113, 401–407 (2000).

    CAS  PubMed  Google Scholar 

  119. Guidato, S., Tsai, L. H., Woodgett, J. & Miller, C. C. Differential cellular phosphorylation of neurofilament heavy side-arms by glycogen synthase kinase-3 and cyclin-dependent kinase-5. J. Neurochem. 66, 1698–1706 (1996).

    CAS  PubMed  Google Scholar 

  120. Hutton, M. et al. Association of missense and 5'-splice-site mutations in tau with the inherited dementia FTDP-17. Nature 393, 702–705 (1998).

    CAS  PubMed  Google Scholar 

  121. Figlewicz, D. A. et al. Variants of the heavy neurofilament subunit are associated with the development of amyotrophic lateral sclerosis. Hum. Mol. Genet. 3, 1757–1761 (1994).

    CAS  PubMed  Google Scholar 

  122. Gros-Louis, F. et al. A frameshift deletion in peripherin gene associated with amyotrophic lateral sclerosis. J. Biol. Chem. 279, 45951–45956 (2004).

    CAS  PubMed  Google Scholar 

  123. 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).

    CAS  PubMed  Google Scholar 

  124. Fischer, L. R. et al. Amyotrophic lateral sclerosis is a distal axonopathy: evidence in mice and man. Exp. Neurol. 185, 232–240 (2004).

    PubMed  Google Scholar 

  125. Henkel, J. S. et al. Presence of dendritic cells, MCP-1, and activated microglia/macrophages in amyotrophic lateral sclerosis spinal cord tissue. Ann. Neurol. 55, 221–235 (2004).

    CAS  PubMed  Google Scholar 

  126. Kuhle, J. et al. Increased levels of inflammatory chemokines in amyotrophic lateral sclerosis. Eur. J. Neurol. 16, 771–774 (2009).

    CAS  PubMed  Google Scholar 

  127. Mantovani, S. et al. Immune system alterations in sporadic amyotrophic lateral sclerosis patients suggest an ongoing neuroinflammatory process. J. Neuroimmunol. 210, 73–79 (2009).

    CAS  PubMed  Google Scholar 

  128. Kipnis, J., Avidan, H., Caspi, R. R. & Schwartz, M. Dual effect of CD4+CD25+ regulatory T cells in neurodegeneration: a dialogue with microglia. Proc. Natl Acad. Sci. USA 101 (Suppl. 2), 14663–14669 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Beers, D. R., Henkel, J. S., Zhao, W., Wang, J. & Appel, S. H. CD4+ T cells support glial neuroprotection, slow disease progression, and modify glial morphology in an animal model of inherited ALS. Proc. Natl Acad. Sci. USA 105, 15558–15563 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Lincecum, J. M. et al. From transcriptome analysis to therapeutic anti-CD40L treatment in the SOD1 model of amyotrophic lateral sclerosis. Nat. Genet. 42, 392–399 (2010).

    CAS  PubMed  Google Scholar 

  131. Ferraiuolo, L. et al. Microarray analysis of the cellular pathways involved in the adaptation to and progression of motor neuron injury in the SOD1 G93A mouse model of familial ALS. J. Neurosci. 27, 9201–9219 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Lobsiger, C. S., Boillee, S. & Cleveland, D. W. Toxicity from different SOD1 mutants dysregulates the complement system and the neuronal regenerative response in ALS motor neurons. Proc. Natl Acad. Sci. USA 104, 7319–7326 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Sta, M. et al. Innate and adaptive immunity in amyotrophic lateral sclerosis: evidence of complement activation. Neurobiol. Dis. 42, 211–220 (2011).

    CAS  PubMed  Google Scholar 

  134. Hensley, K. et al. Primary glia expressing the G93A-SOD1 mutation present a neuroinflammatory phenotype and provide a cellular system for studies of glial inflammation. J. Neuroinflammation 3, 2 (2006).

    PubMed  PubMed Central  Google Scholar 

  135. Di Giorgio, F. P., Boulting, G. L., Bobrowicz, S. & Eggan, K. C. Human embryonic stem cell-derived motor neurons are sensitive to the toxic effect of glial cells carrying an ALS-causing mutation. Cell Stem Cell 3, 637–648 (2008).

    CAS  PubMed  Google Scholar 

  136. Marchetto, M. C. et al. Non-cell-autonomous effect of human SOD1 G37R astrocytes on motor neurons derived from human embryonic stem cells. Cell Stem Cell 3, 649–657 (2008).

    CAS  PubMed  Google Scholar 

  137. Kaufman, R. J. Orchestrating the unfolded protein response in health and disease. J. Clin. Invest. 110, 1389–1398 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Yamagishi, S. et al. An in vitro model for Lewy body-like hyaline inclusion/astrocytic hyaline inclusion: induction by ER stress with an ALS-linked SOD1 mutation. PLoS ONE 2, e1030 (2007).

    PubMed  PubMed Central  Google Scholar 

  139. Hitomi, J. et al. Involvement of caspase-4 in endoplasmic reticulum stress-induced apoptosis and Aβ-induced cell death. J. Cell Biol. 165, 347–356 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Atkin, J. D. et al. Induction of the unfolded protein response in familial amyotrophic lateral sclerosis and association of protein-disulfide isomerase with superoxide dismutase 1. J. Biol. Chem. 281, 30152–30165 (2006).

    CAS  PubMed  Google Scholar 

  141. Atkin, J. D. et al. Endoplasmic reticulum stress and induction of the unfolded protein response in human sporadic amyotrophic lateral sclerosis. Neurobiol. Dis. 30, 400–407 (2008).

    CAS  PubMed  Google Scholar 

  142. Saxena, S., Cabuy, E. & Caroni, P. A role for motoneuron subtype-selective ER stress in disease manifestations of FALS mice. Nat. Neurosci. 12, 627–636 (2009).

    CAS  PubMed  Google Scholar 

  143. Vijayalakshmi, K. et al. Evidence of endoplasmic reticular stress in the spinal motor neurons exposed to CSF from sporadic amyotrophic lateral sclerosis patients. Neurobiol. Dis. 41, 695–705 (2011).

    CAS  PubMed  Google Scholar 

  144. Matus, S., Nassif, M., Glimcher, L. H. & Hetz, C. XBP-1 deficiency in the nervous system reveals a homeostatic switch to activate autophagy. Autophagy 5, 1226–1228 (2009).

    PubMed  Google Scholar 

  145. Hetz, C. et al. XBP-1 deficiency in the nervous system protects against amyotrophic lateral sclerosis by increasing autophagy. Genes Dev. 23, 2294–2306 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Lev, S., Ben Halevy, D., Peretti, D. & Dahan, N. The VAP protein family: from cellular functions to motor neuron disease. Trends Cell Biol. 18, 282–290 (2008).

    CAS  PubMed  Google Scholar 

  147. Chen, H. J. et al. Characterization of the properties of a novel mutation in VAPB in familial amyotrophic lateral sclerosis. J. Biol. Chem. 285, 40266–40281 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Lefebvre, S. et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 80, 155–165 (1995).

    CAS  PubMed  Google Scholar 

  149. Burghes, A. H. & Beattie, C. E. Spinal muscular atrophy: why do low levels of survival motor neuron protein make the motor neurons sick? Nat. Rev. Neurosci. 10, 597–609 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Mackenzie, I. R., Rademakers, R. & Neumann, M. TDP-43 and FUS in amyotrophic lateral sclerosis and frontotemporal dementia. Lancet Neurol. 9, 995–1007 (2010).

    CAS  PubMed  Google Scholar 

  151. Liu-Yesucevitz, L. et al. Tar DNA binding protein-43 (TDP-43) associates with stress granules: analysis of cultured cells and pathological brain tissue. PLoS ONE 5, e13250 (2010).

    PubMed  PubMed Central  Google Scholar 

  152. Ito, D., Seki, M., Tsunoda, Y., Uchiyama, H. & Suzuki, N. Nuclear transport impairment of amyotrophic lateral sclerosis-linked mutations in FUS/TLS. Ann. Neurol. 69, 152–162 (2010).

    PubMed  Google Scholar 

  153. Dormann, D. et al. ALS-associated fused in sarcoma (FUS) mutations disrupt transportin-mediated nuclear import. EMBO J. 29, 2841–2857 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Sephton, C. F. et al. Identification of neuronal RNA targets of TDP-43-containing ribonucleoprotein complexes. J. Biol. Chem. 286, 1204–1215 (2011).

    CAS  PubMed  Google Scholar 

  155. Polymenidou, M. et al. Long pre-mRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43. Nat. Neurosci. 14, 459–468 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Tollervey, J. R. et al. Characterizing the RNA targets and position-dependent splicing regulation by TDP-43. Nat. Neurosci. 14, 452–458 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Xiao, S. et al. RNA targets of TDP-43 identified by UV-CLIP are deregulated in ALS. Mol. Cell. Neurosci. 47, 167–180 (2011).

    CAS  PubMed  Google Scholar 

  158. Highley, J. R. et al. TARDBP mutations, amyotrophic lateral sclerosis and alternative splicing in human fibroblasts. Brain Pathol. 20 (Suppl. 1), 32 (2010).

    Google Scholar 

  159. Wegorzewska, I. & Baloh, R. H. TDP-43 based animal models of neurodegeneration: new insights into ALS pathology and pathophysiology. Neurodegen. Dis. 8, 262–274 (2011).

    CAS  Google Scholar 

  160. Joyce, P. I., Fratta, P., Fisher, E. M. & Acevedo-Arozena, A. SOD1 and TDP-43 animal models of amyotrophic lateral sclerosis: recent advances in understanding disease toward the development of clinical treatments. Mamm. Genome 22, 420–448 (2011).

    CAS  Google Scholar 

  161. Greenway, M. J. et al. ANG mutations segregate with familial and 'sporadic' amyotrophic lateral sclerosis. Nat. Genet. 38, 411–3 (2006).

    CAS  PubMed  Google Scholar 

  162. Kieran, D. et al. Control of motoneuron survival by angiogenin. J. Neurosci. 28, 14056–14061 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Li, S., Yu, W. & Hu, G. F. Angiogenin inhibits nuclear translocation of apoptosis inducing factor in a Bcl-2-dependent manner. J. Cell Physiol. http://dx.doi.org/10.1002/jcp.22881.

  164. Chen, Y. Z. et al. DNA/RNA helicase gene mutations in a form of juvenile amyotrophic lateral sclerosis (ALS4). Am. J. Hum. Genet. 74, 1128–1135 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Clement, A. M. et al. Wild-type nonneuronal cells extend survival of SOD1 mutant motor neurons in ALS mice. Science 302, 113–117 (2003).

    CAS  PubMed  Google Scholar 

  166. Boillee, S. et al. Onset and progression in inherited ALS determined by motor neurons and microglia. Science 312, 1389–1392 (2006).

    CAS  PubMed  Google Scholar 

  167. Gong, Y. H., Parsadanian, A. S., Andreeva, A., Snider, W. D. & Elliott, J. L. Restricted expression of G86R Cu/Zn superoxide dismutase in astrocytes results in astrocytosis but does not cause motoneuron degeneration. J. Neurosci. 20, 660–665 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Yamanaka, K. et al. Astrocytes as determinants of disease progression in inherited amyotrophic lateral sclerosis. Nat. Neurosci. 11, 251–253 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Nagai, M. et al. Astrocytes expressing ALS-linked mutated SOD1 release factors selectively toxic to motor neurons. Nat. Neurosci. 10, 615–622 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Pramatarova, A., Laganiere, J., Roussel, J., Brisebois, K. & Rouleau, G. A. Neuron-specific expression of mutant superoxide dismutase 1 in transgenic mice does not lead to motor impairment. J. Neurosci. 21, 3369–3374 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Jaarsma, D., Teuling, E., Haasdijk, E. D., De Zeeuw, C. I. & Hoogenraad, C. C. Neuron-specific expression of mutant superoxide dismutase is sufficient to induce amyotrophic lateral sclerosis in transgenic mice. J. Neurosci. 28, 2075–2088 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Lobsiger, C. S. et al. Schwann cells expressing dismutase active mutant SOD1 unexpectedly slow disease progression in ALS mice. Proc. Natl Acad. Sci. USA 106, 4465–4470 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Turner, B. J., Ackerley, S., Davies, K. E. & Talbot, K. Dismutase-competent SOD1 mutant accumulation in myelinating Schwann cells is not detrimental to normal or transgenic ALS model mice. Hum. Mol. Genet. 19, 815–824 (2010).

    CAS  PubMed  Google Scholar 

  174. Ferraiuolo, L. et al. Dysregulation of astrocyte-motor neuron cross-talk in mutant SOD1 related amyotrophic lateral sclerosis. Brain 134, 2627–2641 (2011).

    PubMed  PubMed Central  Google Scholar 

  175. Shaw, P. J. & Eggett, C. J. Molecular factors underlying selective vulnerability of motor neurons to neurodegeneration in amyotrophic lateral sclerosis. J. Neurol. 247 (Suppl. 1), 17–27 (2000).

    Google Scholar 

  176. Durham, H. D. in Motor Neuron Disorders: Blue Books of Practical Neurology (eds Shaw, P. J. & Strong, M. J.) 379–400 (Butterworth-Heinemann, Philadelphia, 2003).

    Google Scholar 

  177. Sullivan, P. G. et al. Intrinsic differences in brain and spinal cord mitochondria: implication for therapeutic interventions. J. Comp. Neurol. 474, 524–534 (2004).

    PubMed  Google Scholar 

  178. Panov, A. V. et al. Metabolic and functional differences between brain and spinal cord mitochondria underlie different predisposition to pathology. Am. J. Physiol. Regul. Integr. Comp. Physiol. 300, R844–R854 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Vucic, S. & Kiernan, M. C. Axonal excitability properties in amyotrophic lateral sclerosis. Clin. Neurophysiol. 117, 1458–1466 (2006).

    PubMed  Google Scholar 

  180. Wang, X. & Michaelis, E. K. Selective neuronal vulnerability to oxidative stress in the brain. Front. Aging Neurosci. 2, 12 (2010).

    PubMed  PubMed Central  Google Scholar 

  181. Barber, S. C. et al. Contrasting effects of cerebrospinal fluid from motor neurone disease patients on the survival of primary motor neurons cultured with or without glia. Amyotroph. Lateral Scler. 12, 257–263 (2011).

    PubMed  Google Scholar 

  182. Dupuis, L., Pradat, P. F., Ludolph, A. C. & Loeffler, J. P. Energy metabolism in amyotrophic lateral sclerosis. Lancet Neurol. 10, 75–82 (2011).

    CAS  PubMed  Google Scholar 

  183. Douville, R. et al. Identification of active loci of a human endogenous retrovirus in neurons of patients with amyotrophic lateral sclerosis. Ann. Neurol. 69, 141–151 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Orlacchio, A. et al. SPATACSIN mutations cause autosomal recessive juvenile amyotrophic lateral sclerosis. Brain 133, 591–598 (2010).

    PubMed  PubMed Central  Google Scholar 

  185. Luty, A. A. et al. Sigma nonopioid intracellular receptor1 mutations cause fronto-temporal lobar degeneration-motor neuron disease. Ann. Neurol. 68, 639–649 (2010).

    CAS  PubMed  Google Scholar 

  186. Al-Saif, A., Al-Mohanna, F. & Bohlega, S. A mutation in sigma-1 receptor causes juvenile amyotrophic lateral sclerosis. Ann. Neurol. http://dx.doi.org/10.1002/ana.22534.

  187. Mead, R. J. et al. Optimised and rapid pre-clinical screening in the SOD1G93A transgenic mouse model of amyotrophic lateral sclerosis (ALS). PLoS ONE 6, e23244 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. van Zundert, B. et al. Neonatal neuronal circuitry shows hyperexcitable disturbance in a mouse model of the adult-onset neurodegenerative disease amyotrophic lateral sclerosis. J. Neurosci. 28, 10864–10874 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Bories, C. et al. Early electrophysiological abnormalities in lumbar motoneurons in a transgenic mouse model of amyotrophic lateral sclerosis. Eur. J. Neurosci. 25, 451–459 (2008).

    Google Scholar 

  190. Bendotti, C. et al. Early vacuolization and mitochondrial damage in motor neurons of FALS mice are not associated with apoptosis or with changes in cytochrome oxidase histochemical reactivity. J. Neurol. Sci. 191, 25–33 (2001).

    CAS  PubMed  Google Scholar 

  191. Ramesh, T. et al. A genetic model of amyotrophic lateral sclerosis in zebrafish displays phenotypic hallmarks of motoneuron disease. Dis. Model Mech. 3, 652–662 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Kwiatkowski, T. J. Jr et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323, 1205–1208 (2009).

    CAS  PubMed  Google Scholar 

  193. Vance, C. et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 323, 1208–1211 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Hand, C. K. et al. A novel locus for familial amyotrophic lateral sclerosis on chromosome 18q. Am. J. Hum. Genet. 70, 251–256 (2002).

    CAS  PubMed  Google Scholar 

  195. Sapp, P. C. et al. Identification of two novel loci for dominantly inherited familial amyotrophic lateral sclerosis. Am. J. Hum. Genet. 73, 397–403 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Renton, A. E. et al. A hexonucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS–FTD. Neuron http://dx.doi.org/10.1016/j.neuron.2011.09.010.

  197. De Jesus-Hernandez, M. et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron http://dx.doi.org/10.1016/j.neuron.2011.09.011.

Download references

Acknowledgements

The authors gratefully acknowledge financial support provided by the Wellcome Trust, the UK Motor Neurone Disease Association, the Medical Research Council, The Hermann und Lilli Schilling Stiftung, the Deutsche Forschungsgemeinschaft, the European Union under the 7th Framework Programme for RTD—Project MitoTarget (Grant Agreement HEALTH-F2-2008-223388), Project EuroMotor (Grant Agreement FP7/2007-2013 259867), and the ALS Association.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to researching data for the article, discussion of the content, writing the article, and review and/or editing of the manuscript before submission.

Corresponding author

Correspondence to Pamela J. Shaw.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ferraiuolo, L., Kirby, J., Grierson, A. et al. Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis. Nat Rev Neurol 7, 616–630 (2011). https://doi.org/10.1038/nrneurol.2011.152

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrneurol.2011.152

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