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:

Axonal transport and neurological disease

Abstract

Axonal transport is the process whereby motor proteins actively navigate microtubules to deliver diverse cargoes, such as organelles, from one end of the axon to the other, and is widely regarded as essential for nerve development, function and survival. Mutations in genes encoding key components of the transport machinery, including motor proteins, motor adaptors and microtubules, have been discovered to cause neurological disease. Moreover, disruptions in axonal cargo trafficking have been extensively reported across a wide range of nervous system disorders. However, whether these impairments have a major causative role in, are contributing to or are simply a consequence of neuronal degeneration remains unclear. Therefore, the fundamental relevance of defective trafficking along axons to nerve dysfunction and pathology is often debated. In this article, we review the latest evidence emerging from human and in vivo studies on whether perturbations in axonal transport are indeed integral to the pathogenesis of neurological disease.

Key points

  • Mutations in various genes encoding components of the axonal transport machinery have been implicated in the pathogenesis of neurological diseases.

  • Defective axonal trafficking has been linked to many nervous system disorders, but whether it is a cause or consequence of neuropathology remains largely unresolved.

  • Intravital imaging of transport in axons of live mice provides some of the most compelling evidence that trafficking disturbances contribute to neuronal dysfunction.

  • Targeting of specific mechanisms of axonal transport might be a valid therapeutic strategy to treat neurological disease.

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

Fig. 1: The axonal transport machinery.

Similar content being viewed by others

References

  1. Hirokawa, N. & Tanaka, Y. Kinesin superfamily proteins (KIFs): various functions and their relevance for important phenomena in life and diseases. Exp. Cell Res. 334, 16–25 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Terenzio, M., Schiavo, G. & Fainzilber, M. Compartmentalized signaling in neurons: from cell biology to neuroscience. Neuron 96, 667–679 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Reck-Peterson, S. L., Redwine, W. B., Vale, R. D. & Carter, A. P. The cytoplasmic dynein transport machinery and its many cargoes. Nat. Rev. Mol. Cell Biol. 19, 382–398 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Villarroel-Campos, D., Schiavo, G. & Lazo, O. M. The many disguises of the signalling endosome. FEBS Lett. 592, 3615–3632 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Maday, S. Mechanisms of neuronal homeostasis: autophagy in the axon. Brain Res. 1649, 143–150 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ferguson, S. M. Axonal transport and maturation of lysosomes. Curr. Opin. Neurobiol. 51, 45–51 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rishal, I. & Fainzilber, M. Axon–soma communication in neuronal injury. Nat. Rev. Neurosci. 15, 32–42 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Gibbs, K. L., Greensmith, L. & Schiavo, G. Regulation of axonal transport by protein kinases. Trends Biochem. Sci. 40, 597–610 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Brady, S. T. & Morfini, G. A. Regulation of motor proteins, axonal transport deficits and adult-onset neurodegenerative diseases. Neurobiol. Dis. 105, 273–282 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Dubey, J., Ratnakaran, N. & Koushika, S. P. Neurodegeneration and microtubule dynamics: death by a thousand cuts. Front. Cell. Neurosci. 9, 343 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Barlan, K. & Gelfand, V. I. Microtubule-based transport and the distribution, tethering, and organization of organelles. Cold Spring Harb. Perspect. Biol. 9, a025817 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Hirokawa, N., Niwa, S. & Tanaka, Y. Molecular motors in neurons: transport mechanisms and roles in brain function, development, and disease. Neuron 68, 610–638 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Hinckelmann, M.-V., Zala, D. & Saudou, F. Releasing the brake: restoring fast axonal transport in neurodegenerative disorders. Trends Cell Biol. 23, 634–643 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Maday, S., Twelvetrees, A. E., Moughamian, A. J. & Holzbaur, E. L. F. Axonal transport: cargo-specific mechanisms of motility and regulation. Neuron 84, 292–309 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Neefjes, J. & van der Kant, R. Stuck in traffic: an emerging theme in diseases of the nervous system. Trends Neurosci. 37, 66–76 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. DiMauro, S., Schon, E. A., Carelli, V. & Hirano, M. The clinical maze of mitochondrial neurology. Nat. Rev. Neurol. 9, 429–444 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Millecamps, S. & Julien, J.-P. Axonal transport deficits and neurodegenerative diseases. Nat. Rev. Neurosci. 14, 161–176 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Prior, R., Van Helleputte, L., Benoy, V. & Van Den Bosch, L. Defective axonal transport: a common pathological mechanism in inherited and acquired peripheral neuropathies. Neurobiol. Dis. 105, 300–320 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Milde, S., Adalbert, R., Elaman, M. H. & Coleman, M. P. Axonal transport declines with age in two distinct phases separated by a period of relative stability. Neurobiol. Aging 36, 971–981 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Vagnoni, A., Hoffmann, P. C. & Bullock, S. L. Reducing Lissencephaly-1 levels augments mitochondrial transport and has a protective effect in adult Drosophila neurons. J. Cell Sci. 129, 178–190 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Sleigh, J. N. & Schiavo, G. Older but not slower: aging does not alter axonal transport dynamics of signalling endosomes in vivo. Matters https://doi.org/10.19185/matters.201605000018 (2016).

  22. Brouhard, G. J. & Rice, L. M. Microtubule dynamics: an interplay of biochemistry and mechanics. Nat. Rev. Mol. Cell Biol. 19, 451–463 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rao, A. N. & Baas, P. W. Polarity sorting of microtubules in the axon. Trends Neurosci. 41, 77–88 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Baas, P. W., Rao, R. N., Matamoros, A. J. & Leo, L. Stability properties of neuronal microtubules. Cytoskeleton 73, 442–460 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Miki, H., Setou, M., Kaneshiro, K. & Hirokawa, N. All kinesin superfamily protein, KIF, genes in mouse and human. Proc. Natl Acad. Sci. USA 98, 7004–7011 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Carter, A. P., Diamant, A. G. & Urnavicius, L. How dynein and dynactin transport cargos: a structural perspective. Curr. Opin. Struct. Biol. 37, 62–70 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Zhang, K. et al. Cryo-EM reveals how human cytoplasmic dynein is auto-inhibited and activated. Cell 169, 1303–1314 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. McKenney, R. J., Huynh, W., Tanenbaum, M. E., Bhabha, G. & Vale, R. D. Activation of cytoplasmic dynein motility by dynactin–cargo adapter complexes. Science 345, 337–341 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Schlager, M. A. et al. Bicaudal D family adaptor proteins control the velocity of Dynein-based movements. Cell Rep. 8, 1248–1256 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Budzinska, M., Wicher, K. B. & Terenzio, M. Neuronal roles of the bicaudal D family of motor adaptors. Vitam. Horm. 104, 133–152 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. DeSantis, M. E. et al. Lis1 has two opposing modes of regulating cytoplasmic dynein. Cell 170, 1197–1208 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Huang, J., Roberts, A. J., Leschziner, A. E. & Reck-Peterson, S. L. Lis1 acts as a “clutch” between the ATPase and microtubule-binding domains of the dynein motor. Cell 150, 975–986 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Baumbach, J. et al. Lissencephaly-1 is a context-dependent regulator of the human dynein complex. eLife 6, e21768 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Yi, J. Y. et al. High-resolution imaging reveals indirect coordination of opposite motors and a role for LIS1 in high-load axonal transport. J. Cell Biol. 195, 193–201 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zyłkiewicz, E. et al. The N-terminal coiled-coil of Ndel1 is a regulated scaffold that recruits LIS1 to dynein. J. Cell Biol. 192, 433–445 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Olenick, M. A. & Holbaur, E. L. F. Dynein activators and adaptors at a glance. J. Cell Sci. 132, jcs227132 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Griffin, J. W., Price, D. L., Drachman, D. B. & Engel, W. K. Axonal transport to and from the motor nerve ending. Ann. N. Y. Acad. Sci. 274, 31–45 (1976).

    Article  CAS  PubMed  Google Scholar 

  38. Roy, S. Seeing the unseen: the hidden world of slow axonal transport. Neuroscientist 20, 71–81 (2014).

    Article  PubMed  Google Scholar 

  39. Brown, A., Wang, L. & Jung, P. Stochastic simulation of neurofilament transport in axons: the “stop-and-go” hypothesis. Mol. Biol. Cell 16, 4243–4255 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Garner, J. A. & Mahler, H. R. Biogenesis of presynaptic terminal proteins. J. Neurochem. 49, 905–915 (1987).

    Article  CAS  PubMed  Google Scholar 

  41. Twelvetrees, A. E. et al. The dynamic localization of cytoplasmic dynein in neurons is driven by kinesin-1. Neuron 90, 1000–1015 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Allen, R. D., Metuzals, J., Tasaki, I., Brady, S. T. & Gilbert, S. P. Fast axonal transport in squid giant axon. Science 218, 1127–1129 (1982).

    Article  CAS  PubMed  Google Scholar 

  43. Brady, S. T., Lasek, R. J. & Allen, R. D. Fast axonal transport in extruded axoplasm from squid giant axon. Science 218, 1129–1131 (1982).

    Article  CAS  PubMed  Google Scholar 

  44. Klinman, E. & Holzbaur, E. L. F. Walking forward with kinesin. Trends Neurosci. 41, 555–556 (2018).

    Article  CAS  PubMed  Google Scholar 

  45. Schiavo, G., Greensmith, L., Hafezparast, M. & Fisher, E. M. C. Cytoplasmic dynein heavy chain: the servant of many masters. Trends Neurosci. 36, 641–651 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Poirier, K. et al. Mutations in TUBG1, DYNC1H1, KIF5C and KIF2A cause malformations of cortical development and microcephaly. Nat. Genet. 45, 639–647 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Tsai, J.-W., Lian, W.-N., Kemal, S., Kriegstein, A. R. & Vallee, R. B. Kinesin 3 and cytoplasmic dynein mediate interkinetic nuclear migration in neural stem cells. Nat. Neurosci. 13, 1463–1471 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Doobin, D. J., Kemal, S., Dantas, T. J. & Vallee, R. B. Severe NDE1-mediated microcephaly results from neural progenitor cell cycle arrests at multiple specific stages. Nat. Commun. 7, 12551 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hu, D. J.-K. et al. Dynein recruitment to nuclear pores activates apical nuclear migration and mitotic entry in brain progenitor cells. Cell 154, 1300–1313 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Tsai, J.-W., Bremner, K. H. & Vallee, R. B. Dual subcellular roles for LIS1 and dynein in radial neuronal migration in live brain tissue. Nat. Neurosci. 10, 970–979 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Ori-McKenney, K. M. & Vallee, R. B. Neuronal migration defects in the Loa dynein mutant mouse. Neural Dev. 6, 26 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Poirier, K. et al. Mutations in the neuronal β-tubulin subunit TUBB3 result in malformation of cortical development and neuronal migration defects. Hum. Mol. Genet. 19, 4462–4473 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Jaglin, X. H. et al. Mutations in the β-tubulin gene TUBB2B result in asymmetrical polymicrogyria. Nat. Genet. 41, 746–752 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Yamada, K. et al. Heterozygous mutations of the kinesin KIF21A in congenital fibrosis of the extraocular muscles type 1 (CFEOM1). Nat. Genet. 35, 318–321 (2003).

    Article  CAS  PubMed  Google Scholar 

  55. Cheng, L. et al. Human CFEOM1 mutations attenuate KIF21A autoinhibition and cause oculomotor axon stalling. Neuron 82, 334–349 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Scoto, M. et al. Novel mutations expand the clinical spectrum of DYNC1H1-associated spinal muscular atrophy. Neurol. 84, 668–679 (2015).

    Article  CAS  Google Scholar 

  57. Rossor, A. M. et al. Phenotypic and molecular insights into spinal muscular atrophy due to mutations in BICD2. Brain 138, 293–310 (2015).

    Article  PubMed  Google Scholar 

  58. Huynh, W. & Vale, R. D. Disease-associated mutations in human BICD2 hyperactivate motility of dynein–dynactin. J. Cell Biol. 216, 3051–3060 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hoang, H. T., Schlager, M. A., Carter, A. P. & Bullock, S. L. DYNC1H1 mutations associated with neurological diseases compromise processivity of dynein–dynactin–cargo adaptor complexes. Proc. Natl Acad. Sci. USA 114, E1597–E1606 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Chen, X.-J. et al. Proprioceptive sensory neuropathy in mice with a mutation in the cytoplasmic Dynein heavy chain 1 gene. J. Neurosci. 27, 14515–14524 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hafezparast, M. et al. Mutations in dynein link motor neuron degeneration to defects in retrograde transport. Science 300, 808–812 (2003).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Zhao, J. et al. Dync1h1 mutation causes proprioceptive sensory neuron loss and impaired retrograde axonal transport of dorsal root ganglion neurons. CNS Neurosci. Ther. 22, 593–601 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hwang, S. H. et al. Distal hereditary motor neuropathy type 7B with Dynactin 1 mutation. Mol. Med. Rep. 14, 3362–3368 (2016).

    Article  CAS  PubMed  Google Scholar 

  65. Farrer, M. J. et al. DCTN1 mutations in Perry syndrome. Nat. Genet. 41, 163–165 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Puls, I. et al. Mutant dynactin in motor neuron disease. Nat. Genet. 33, 455–456 (2003).

    Article  CAS  PubMed  Google Scholar 

  67. Mishima, T. et al. Perry syndrome: a distinctive type of TDP-43 proteinopathy. J. Neuropathol. Exp. Neurol. 76, 676–682 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lloyd, T. E. et al. The p150Glued CAP-Gly domain regulates initiation of retrograde transport at synaptic termini. Neuron 74, 344–360 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Chevalier-Larsen, E. S., Wallace, K. E., Pennise, C. R. & Holzbaur, E. L. F. Lysosomal proliferation and distal degeneration in motor neurons expressing the G59S mutation in the p150Glued subunit of dynactin. Hum. Mol. Genet. 17, 1946–1955 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Moughamian, A. J. & Holzbaur, E. L. F. Dynactin is required for transport initiation from the distal axon. Neuron 74, 331–343 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Weedon, M. N. et al. Exome sequencing identifies a dync1h1 mutation in a large pedigree with dominant axonal Charcot–Marie–Tooth disease. Am. J. Hum. Genet. 89, 308–312 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Rivière, J.-B. et al. KIF1A, an axonal transporter of synaptic vesicles, is mutated in hereditary sensory and autonomic neuropathy type 2. Am. J. Hum. Genet. 89, 219–230 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Lee, J.-R. et al. De novo mutations in the motor domain of KIF1A cause cognitive impairment, spastic paraparesis, axonal neuropathy, and cerebellar atrophy. Hum. Mutat. 36, 69–78 (2015).

    Article  CAS  PubMed  Google Scholar 

  74. Tanaka, Y. et al. The molecular motor KIF1A transports the TrkA neurotrophin receptor and is essential for sensory neuron survival and function. Neuron 90, 1215–1229 (2016).

    Article  CAS  PubMed  Google Scholar 

  75. Chiba, K. et al. Disease-associated mutations hyperactivate KIF1A motility and anterograde axonal transport of synaptic vesicle precursors. Proc. Natl Acad. Sci. USA https://doi.org/10.1073/pnas.1905690116 (2019).

    Article  CAS  Google Scholar 

  76. Reid, E. et al. A kinesin heavy chain (KIF5A) mutation in hereditary spastic paraplegia (SPG10). Am. J. Hum. Genet. 71, 1189–1194 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Füger, P. et al. Spastic paraplegia mutation N256S in the neuronal microtubule motor KIF5A disrupts axonal transport in a Drosophila HSP model. PLOS Genet. 8, e1003066 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Brenner, D. et al. Hot-spot KIF5A mutations cause familial ALS. Brain 141, 688–697 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Nicolas, A. et al. Genome-wide analyses identify KIF5A as a novel ALS gene. Neuron 97, 1268–1283 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Zhao, C. et al. Charcot–Marie–Tooth disease type 2A caused by mutation in a microtubule motor KIF1Bβ. Cell 105, 587–597 (2001).

    Article  CAS  PubMed  Google Scholar 

  81. Martin, M. et al. Cytoplasmic dynein, the dynactin complex, and kinesin are interdependent and essential for fast axonal transport. Mol. Biol. Cell 10, 3717–3728 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. LaMonte, B. H. et al. Disruption of dynein/dynactin inhibits axonal transport in motor neurons causing late-onset progressive degeneration. Neuron 34, 715–727 (2002).

    Article  CAS  PubMed  Google Scholar 

  83. Stokin, G. B. et al. Axonopathy and transport deficits early in the pathogenesis of Alzheimer’s disease. Science 307, 1282–1288 (2005).

    Article  CAS  PubMed  Google Scholar 

  84. De Vos, K. J. & Hafezparast, M. Neurobiology of axonal transport defects in motor neuron diseases: opportunities for translational research? Neurobiol. Dis. 105, 283–299 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Toyoshima, I. et al. Kinesin and cytoplasmic dynein in spinal spheroids with motor neuron disease. J. Neurol. Sci. 159, 38–44 (1998).

    Article  CAS  PubMed  Google Scholar 

  86. Galvin, J. E., Uryu, K., Lee, V. M. & Trojanowski, J. Q. Axon pathology in Parkinson’s disease and Lewy body dementia hippocampus contains α-, β-, and γ-synuclein. Proc. Natl Acad. Sci. USA 96, 13450–13455 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Fanara, P. et al. Cerebrospinal fluid-based kinetic biomarkers of axonal transport in monitoring neurodegeneration. J. Clin. Invest. 122, 3159–3169 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Hares, K. et al. Overexpression of kinesin superfamily motor proteins in Alzheimer’s disease. J. Alzheimers Dis. 60, 1511–1524 (2017).

    Article  CAS  PubMed  Google Scholar 

  89. Pantelidou, M. et al. Differential expression of molecular motors in the motor cortex of sporadic ALS. Neurobiol. Dis. 26, 577–589 (2007).

    Article  CAS  PubMed  Google Scholar 

  90. Hares, K. et al. Axonal motor protein KIF5A and associated cargo deficits in multiple sclerosis lesional and normal-appearing white matter. Neuropathol. Appl. Neurobiol. 43, 227–241 (2017).

    Article  CAS  PubMed  Google Scholar 

  91. Chu, Y. et al. Alterations in axonal transport motor proteins in sporadic and experimental Parkinson’s disease. Brain 135, 2058–2073 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  92. Cash, A. D. et al. Microtubule reduction in Alzheimer’s disease and aging is independent of tau filament formation. Am. J. Pathol. 162, 1623–1627 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zhang, F. et al. Posttranslational modifications of α-tubulin in Alzheimer disease. Transl. Neurodegener. 4, 9 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Ren, Y. et al. Parkin mutations reduce the complexity of neuronal processes in iPSC-derived human neurons. Stem Cells 33, 68–78 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Cartelli, D. et al. Parkin absence accelerates microtubule aging in dopaminergic neurons. Neurobiol. Aging 61, 66–74 (2018).

    Article  CAS  PubMed  Google Scholar 

  96. Brunden, K. R., Lee, V. M.-Y., Smith, A. B., Trojanowski, J. Q. & Ballatore, C. Altered microtubule dynamics in neurodegenerative disease: therapeutic potential of microtubule-stabilizing drugs. Neurobiol. Dis. 105, 328–335 (2017).

    Article  CAS  PubMed  Google Scholar 

  97. Saporta, M. A. et al. Axonal Charcot–Marie–Tooth disease patient-derived motor neurons demonstrate disease-specific phenotypes including abnormal electrophysiological properties. Exp. Neurol. 263, 190–199 (2015).

    Article  CAS  PubMed  Google Scholar 

  98. Xu, C.-C., Denton, K. R., Wang, Z.-B., Zhang, X. & Li, X.-J. Abnormal mitochondrial transport and morphology as early pathological changes in human models of spinal muscular atrophy. Dis. Model. Mech. 9, 39–49 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Sleigh, J. N., Vagnoni, A., Twelvetrees, A. E. & Schiavo, G. Methodological advances in imaging intravital axonal transport. F1000Res. 6, 200 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Guo, W. et al. HDAC6 inhibition reverses axonal transport defects in motor neurons derived from FUS-ALS patients. Nat. Commun. 8, 861 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Plucińska, G. & Misgeld, T. Imaging of neuronal mitochondria in situ. Curr. Opin. Neurobiol. 39, 152–163 (2016).

    Article  PubMed  CAS  Google Scholar 

  102. Hinckelmann, M.-V. et al. Self-propelling vesicles define glycolysis as the minimal energy machinery for neuronal transport. Nat. Commun. 7, 13233 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Alami, N. H. et al. Axonal transport of TDP-43 mRNA granules is impaired by ALS-causing mutations. Neuron 81, 536–543 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Baldwin, K. R., Godena, V. K., Hewitt, V. L. & Whitworth, A. J. Axonal transport defects are a common phenotype in Drosophila models of ALS. Hum. Mol. Genet. 25, 2378–2392 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Lacovich, V. et al. Tau isoforms imbalance impairs the axonal transport of the amyloid precursor protein in human neurons. J. Neurosci. 37, 58–69 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Moutaux, E. et al. Neuronal network maturation differently affects secretory vesicles and mitochondria transport in axons. Sci. Rep. 8, 13429 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Pal, A. et al. High content organelle trafficking enables disease state profiling as powerful tool for disease modelling. Sci. Data 5, 180241 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Berry, B. J., Smith, A. S. T., Young, J. E. & Mack, D. L. Advances and current challenges associated with the use of human induced pluripotent stem cells in modeling neurodegenerative disease. Cells Tissues Organs 205, 331–349 (2018).

    Article  CAS  PubMed  Google Scholar 

  109. Drouin-Ouellet, J., Pircs, K., Barker, R. A., Jakobsson, J. & Parmar, M. Direct neuronal reprogramming for disease modeling studies using patient-derived neurons: what have we learned? Front. Neurosci. 11, 530 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  110. Gibbs, K. L., Kalmar, B., Sleigh, J. N., Greensmith, L. & Schiavo, G. In vivo imaging of axonal transport in murine motor and sensory neurons. J. Neurosci. Methods 257, 26–33 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Lewis, T. L., Turi, G. F., Kwon, S.-K., Losonczy, A. & Polleux, F. Progressive decrease of mitochondrial motility during maturation of cortical axons in vitro and in vivo. Curr. Biol. 26, 2602–2608 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Smit-Rigter, L. et al. Mitochondrial dynamics in visual cortex are limited in vivo and not affected by axonal structural plasticity. Curr. Biol. 26, 2609–2616 (2016).

    Article  CAS  PubMed  Google Scholar 

  113. Knabbe, J., Nassal, J. P., Verhage, M. & Kuner, T. Secretory vesicle trafficking in awake and anaesthetized mice: differential speeds in axons versus synapses. J. Physiol. 596, 3759–3773 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Mitchell, D. J. et al. Trk activation of the ERK1/2 kinase pathway stimulates intermediate chain phosphorylation and recruits cytoplasmic dynein to signaling endosomes for retrograde axonal transport. J. Neurosci. 32, 15495–15510 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Pathak, A. et al. Retrograde degenerative signaling mediated by the p75 neurotrophin receptor requires p150Glued deacetylation by axonal HDAC1. Dev. Cell 46, 376–387 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Kiryu-Seo, S., Ohno, N., Kidd, G. J., Komuro, H. & Trapp, B. D. Demyelination increases axonal stationary mitochondrial size and the speed of axonal mitochondrial transport. J. Neurosci. 30, 6658–6666 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Badal, K. K. et al. Synapse formation activates a transcriptional program for persistent enhancement in the bi-directional transport of mitochondria. Cell Rep. 26, 507–517 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Smith, S. E. & Bonni, A. in The Molecular and Cellular Basis of Neurodegenerative Diseases (ed. Wolfe, M. S.) 415–440 (Elsevier, 2018).

  119. Sajic, M. et al. Impulse conduction increases mitochondrial transport in adult mammalian peripheral nerves in vivo. PLOS Biol. 11, e1001754 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Wang, T. et al. Flux of signalling endosomes undergoing axonal retrograde transport is encoded by presynaptic activity and TrkB. Nat. Commun. 7, 12976 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Shidara, Y. & Hollenbeck, P. J. Defects in mitochondrial axonal transport and membrane potential without increased reactive oxygen species production in a Drosophila model of Friedreich ataxia. J. Neurosci. 30, 11369–11378 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Devireddy, S., Liu, A., Lampe, T. & Hollenbeck, P. J. The organization of mitochondrial quality control and life cycle in the nervous system in vivo in the absence of PINK1. J. Neurosci. 35, 9391–9401 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Godena, V. K. et al. Increasing microtubule acetylation rescues axonal transport and locomotor deficits caused by LRRK2 Roc-COR domain mutations. Nat. Commun. 5, 5245 (2014).

    Article  CAS  PubMed  Google Scholar 

  124. Fatouros, C. et al. Inhibition of tau aggregation in a novel Caenorhabditis elegans model of tauopathy mitigates proteotoxicity. Hum. Mol. Genet. 21, 3587–3603 (2012).

    Article  CAS  PubMed  Google Scholar 

  125. Butler, V. J. et al. Tau/MAPT disease-associated variant A152T alters tau function and toxicity via impaired retrograde axonal transport. Hum. Mol. Genet. 28, 1498–1514 (2019).

    Article  CAS  PubMed  Google Scholar 

  126. Bergamin, G., Cieri, D., Vazza, G., Argenton, F. & Mostacciuolo, M. L. Zebrafish Tg(hb9:MTS-Kaede): a new in vivo tool for studying the axonal movement of mitochondria. Biochim. Biophys. Acta 1860, 1247–1255 (2016).

    Article  CAS  PubMed  Google Scholar 

  127. Dukes, A. A. et al. Live imaging of mitochondrial dynamics in CNS dopaminergic neurons in vivo demonstrates early reversal of mitochondrial transport following MPP+ exposure. Neurobiol. Dis. 95, 238–249 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Bothwell, M. Recent advances in understanding neurotrophin signaling. F1000Res. 5, 1885 (2016).

    Article  CAS  Google Scholar 

  129. Zhang, B., Tu, P., Abtahian, F., Trojanowski, J. Q. & Lee, V. M. Neurofilaments and orthograde transport are reduced in ventral root axons of transgenic mice that express human SOD1 with a G93A mutation. J. Cell Biol. 139, 1307–1315 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. 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).

    Article  CAS  PubMed  Google Scholar 

  131. Magrané, J., Cortez, C., Gan, W.-B. & Manfredi, G. Abnormal mitochondrial transport and morphology are common pathological denominators in SOD1 and TDP43 ALS mouse models. Hum. Mol. Genet. 23, 1413–1424 (2014).

    Article  PubMed  CAS  Google Scholar 

  132. Gordon, D. et al. Single-copy expression of an amyotrophic lateral sclerosis-linked TDP-43 mutation (M337V) in BAC transgenic mice leads to altered stress granule dynamics and progressive motor dysfunction. Neurobiol. Dis. 121, 148–162 (2019).

    Article  CAS  PubMed  Google Scholar 

  133. Sleigh, J. N. et al. ALS mice carrying pathological mutant TDP-43, but not mutant FUS, display axonal transport defects in vivo. BioRxiv https://doi.org/10.1101/438812 (2018).

  134. Hardiman, O. et al. Amyotrophic lateral sclerosis. Nat. Rev. Dis. Prim. 3, 17071 (2017).

    Article  PubMed  Google Scholar 

  135. Devoy, A. et al. Humanized mutant FUS drives progressive motor neuron degeneration without aggregation in “FUSDelta14” knockin mice. Brain 140, 2797–2805 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  136. Marinkovic, P. et al. Axonal transport deficits and degeneration can evolve independently in mouse models of amyotrophic lateral sclerosis. Proc. Natl Acad. Sci. USA 109, 4296–4301 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Malik, B. et al. Absence of disturbed axonal transport in spinal and bulbar muscular atrophy. Hum. Mol. Genet. 20, 1776–1786 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Sorbara, C. D. et al. Pervasive axonal transport deficits in multiple sclerosis models. Neuron 84, 1183–1190 (2014).

    Article  CAS  PubMed  Google Scholar 

  139. Morfini, G. et al. JNK mediates pathogenic effects of polyglutamine-expanded androgen receptor on fast axonal transport. Nat. Neurosci. 9, 907–916 (2006).

    Article  CAS  PubMed  Google Scholar 

  140. Halievski, K., Kemp, M. Q., Breedlove, S. M., Miller, K. E. & Jordan, C. L. Non-cell-autonomous regulation of retrograde motoneuronal axonal transport in an SBMA mouse model. eNeuro 3, e0062–16.2016 (2016).

    Article  Google Scholar 

  141. Wang, W. et al. The ALS disease-associated mutant TDP-43 impairs mitochondrial dynamics and function in motor neurons. Hum. Mol. Genet. 22, 4706–4719 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Morfini, G. A. et al. Inhibition of fast axonal transport by pathogenic SOD1 involves activation of p38 MAP kinase. PLOS ONE 8, e65235 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Moller, A., Bauer, C. S., Cohen, R. N., Webster, C. P. & De Vos, K. J. Amyotrophic lateral sclerosis-associated mutant SOD1 inhibits anterograde axonal transport of mitochondria by reducing Miro1 levels. Hum. Mol. Genet. 26, 4668–4679 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Gibbs, K. L. et al. Inhibiting p38 MAPK alpha rescues axonal retrograde transport defects in a mouse model of ALS. Cell Death Dis. 9, 596 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Ruschel, J. et al. Systemic administration of epothilone B promotes axon regeneration after spinal cord injury. Science 348, 347–352 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Mar, F. M., Bonni, A. & Sousa, M. M. Cell intrinsic control of axon regeneration. EMBO Rep. 15, 254–263 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Clark, A. J. et al. Epothilone D accelerates disease progression in the SOD1G93A mouse model of amyotrophic lateral sclerosis. Neuropathol. Appl. Neurobiol. 44, 590–605 (2018).

    Article  CAS  PubMed  Google Scholar 

  148. Reed, N. A. et al. Microtubule acetylation promotes kinesin-1 binding and transport. Curr. Biol. 16, 2166–2172 (2006).

    Article  CAS  PubMed  Google Scholar 

  149. Dompierre, J. P. et al. Histone deacetylase 6 inhibition compensates for the transport deficit in Huntington’s disease by increasing tubulin acetylation. J. Neurosci. 27, 3571–3583 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. d’Ydewalle, C. et al. HDAC6 inhibitors reverse axonal loss in a mouse model of mutant HSPB1-induced Charcot–Marie–Tooth disease. Nat. Med. 17, 968–974 (2011).

    Article  PubMed  CAS  Google Scholar 

  151. Mo, Z. et al. Aberrant GlyRS–HDAC6 interaction linked to axonal transport deficits in Charcot–Marie–Tooth neuropathy. Nat. Commun. 9, 1007 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Taes, I. et al. Hdac6 deletion delays disease progression in the SOD1G93A mouse model of ALS. Hum. Mol. Genet. 22, 1783–1790 (2013).

    Article  CAS  PubMed  Google Scholar 

  153. Kalinski, A. L. et al. Deacetylation of Miro1 by HDAC6 blocks mitochondrial transport and mediates axon growth inhibition. J. Cell Biol. 218, 1871–1890 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Zhang, K. et al. Defective axonal transport of Rab7 GTPase results in dysregulated trophic signaling. J. Neurosci. 33, 7451–7462 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Cioni, J.-M. et al. Late endosomes act as mRNA translation platforms and sustain mitochondria in axons. Cell 176, 56–72 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Zhu, Y. B. & Sheng, Z. H. Increased axonal mitochondrial mobility does not slow amyotrophic lateral sclerosis (ALS)-like disease in mutant SOD1 mice. J. Biol. Chem. 286, 23432–23440 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Perlson, E. et al. A switch in retrograde signaling from survival to stress in rapid-onset neurodegeneration. J. Neurosci. 29, 9903–9917 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Fu, M. & Holzbaur, E. L. F. JIP1 regulates the directionality of APP axonal transport by coordinating kinesin and dynein motors. J. Cell Biol. 202, 495–508 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Lee, S., Pant, H. C. & Shea, T. B. Divergent and convergent roles for kinases and phosphatases in neurofilament dynamics. J. Cell Sci. 127, 4064–4077 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Stevenson, A. et al. Riluzole protects against glutamate-induced slowing of neurofilament axonal transport. Neurosci. Lett. 454, 161–164 (2009).

    Article  CAS  PubMed  Google Scholar 

  161. 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).

    Article  CAS  PubMed  Google Scholar 

  162. 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).

    Article  CAS  PubMed  Google Scholar 

  163. Dewil, M., dela Cruz, V. F., van den Bosch, L. & Robberecht, W. Inhibition of p38 mitogen activated protein kinase activation and mutant SOD1G93A-induced motor neuron death. Neurobiol. Dis. 26, 332–341 (2007).

    Article  CAS  PubMed  Google Scholar 

  164. Chico, L. K., Van Eldik, L. J. & Watterson, D. M. Targeting protein kinases in central nervous system disorders. Nat. Rev. Drug Discov. 8, 892–909 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Hetman, M. & Gozdz, A. Role of extracellular signal regulated kinases 1 and 2 in neuronal survival. Eur. J. Biochem. 271, 2050–2055 (2004).

    Article  CAS  PubMed  Google Scholar 

  166. Hur, E.-M. & Zhou, F.-Q. GSK3 signalling in neural development. Nat. Rev. Neurosci. 11, 539–551 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Coffey, E. T. Nuclear and cytosolic JNK signalling in neurons. Nat. Rev. Neurosci. 15, 285–299 (2014).

    Article  CAS  PubMed  Google Scholar 

  168. Ally, S., Larson, A. G., Barlan, K., Rice, S. E. & Gelfand, V. I. Opposite-polarity motors activate one another to trigger cargo transport in live cells. J. Cell Biol. 187, 1071–1082 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Schuster, M. et al. Kinesin-3 and dynein cooperate in long-range retrograde endosome motility along a nonuniform microtubule array. Mol. Biol. Cell 22, 3645–3657 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Wu, C., Watts, M. E. & Rubin, L. L. MAP4K4 activation mediates motor neuron degeneration in amyotrophic lateral sclerosis. Cell Rep. 26, 1143–1156 (2019).

    Article  CAS  PubMed  Google Scholar 

  171. Xie, Y. et al. Endolysosomal deficits augment mitochondria pathology in spinal motor neurons of asymptomatic fALS mice. Neuron 87, 355–370 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors’ work is supported by the Medical Research Council Career Development Award (MR/S006990/1 to J.N.S.), a Wellcome Trust Postdoctoral Fellowship for Clinicians (110043/Z/15/Z to A.M.R.), a Wellcome Trust Senior Investigator Award (107116/Z/15/Z to G.S.), the European Union’s Horizon 2020 Research and Innovation programme under grant agreement 739572 (to G.S.) and a UK Dementia Research Institute Foundation award (to G.S.).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the researching, writing and editing of this Review.

Corresponding author

Correspondence to Giampietro Schiavo.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Neurology thanks K. De Vos and other anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Databases

Online Mendelian Inheritance in Man: http://www.omim.org/

Glossary

Processivity

The ability of motor–cargo complexes to undergo axonal transport without dissociation from microtubules.

Coiled-coil domains

A structural motif composed of two or more α-helices wrapped around each other to form a supercoil. The coiled-coil domain of the cytoplasmic dynein–dynactin complex connects the ATPase domain with the microtubule-binding domain.

Run length

The total displacement covered by a motor–cargo complex without pausing.

Microtubule gliding assays

An experimental technique that is used to assess the activity of motor proteins, in which microtubules and ATP are applied to motors bound to glass coverslips.

N-Ethyl-N-nitrosourea

(ENU). A potent mutagen that is often used to generate mutant animal models.

Endosomes

Membranous organelles involved in intracellular transport, sorting and delivery of various substances, including growth factors, internalized from the cell exterior.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sleigh, J.N., Rossor, A.M., Fellows, A.D. et al. Axonal transport and neurological disease. Nat Rev Neurol 15, 691–703 (2019). https://doi.org/10.1038/s41582-019-0257-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41582-019-0257-2

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing