Skip to main content

Mechanisms Associated with TDP-43 Neurotoxicity in ALS/FTLD

  • Chapter
  • First Online:
RNA Metabolism in Neurodegenerative Diseases

Part of the book series: Advances in Neurobiology ((NEUROBIOL,volume 20))

Abstract

The discovery of TDP-43 as a major disease protein in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) was first made in 2006. Prior to 2006 there were only 11 publications related to TDP-43, now there are over 2000, indicating the importance of TDP-43 to unraveling the complex molecular mechanisms that underpin the pathogenesis of ALS/FTLD. Subsequent to this discovery, TDP-43 pathology was also found in other neurodegenerative diseases, including Alzheimer’s disease, the significance of which is still in the early stages of exploration. TDP-43 is a predominantly nuclear DNA/RNA-binding protein, one of a number of RNA-binding proteins that are now known to be linked with ALS/FTLD, including Fused in Sarcoma (FUS), heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1), and heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1). However, what sets TDP-43 apart is the vast number of cases in which TDP-43 pathology is present, providing a point of convergence, the understanding of which could lead to broadly applicable therapeutics. Here we will focus on TDP-43 in ALS/FTLD, its nuclear and cytoplasmic functions, and consequences should these functions go awry.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 109.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Kiernan MC, Vucic S, Cheah BC, Turner MR, Eisen A, Hardiman O, et al. Amyotrophic lateral sclerosis. Lancet. 2011;377(9769):942–55.

    Article  PubMed  CAS  Google Scholar 

  2. Chio A, Logroscino G, Traynor BJ, Collins J, Simeone JC, Goldstein LA, et al. Global epidemiology of amyotrophic lateral sclerosis: a systematic review of the published literature. Neuroepidemiology. 2013;41(2):118–30.

    Article  PubMed  CAS  Google Scholar 

  3. Chia R, Chio A, Traynor BJ. Novel genes associated with amyotrophic lateral sclerosis: diagnostic and clinical implications. Lancet Neurol. 2018;17(1):94–102.

    Article  PubMed  CAS  Google Scholar 

  4. Strong MJ. Revisiting the concept of amyotrophic lateral sclerosis as a multisystems disorder of limited phenotypic expression. Curr Opin Neurol. 2017;30(6):599–607.

    Article  PubMed  Google Scholar 

  5. Ng AS, Rademakers R, Miller BL. Frontotemporal dementia: a bridge between dementia and neuromuscular disease. Ann N Y Acad Sci. 2015;1338:71–93.

    Article  PubMed  CAS  Google Scholar 

  6. Bang J, Spina S, Miller BL. Frontotemporal dementia. Lancet. 2015;386(10004):1672–82.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Neary D, Snowden JS, Gustafson L, Passant U, Stuss D, Black S, et al. Frontotemporal lobar degeneration: a consensus on clinical diagnostic criteria. Neurology. 1998;51(6):1546–54.

    Article  PubMed  CAS  Google Scholar 

  8. Warren JD, Rohrer JD, Rossor MN. Clinical review. Frontotemporal dementia. BMJ. 2013;347:f4827.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Phukan J, Elamin M, Bede P, Jordan N, Gallagher L, Byrne S, et al. The syndrome of cognitive impairment in amyotrophic lateral sclerosis: a population-based study. J Neurol Neurosurg Psychiatry. 2012;83(1):102–8.

    Article  PubMed  Google Scholar 

  10. Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, Mori H, et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun. 2006;351(3):602–11.

    Article  CAS  PubMed  Google Scholar 

  11. Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314(5796):130–3.

    Article  CAS  PubMed  Google Scholar 

  12. Kabashi E, Valdmanis PN, Dion P, Spiegelman D, McConkey BJ, Vande Velde C, et al. TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat Genet. 2008;40(5):572–4.

    Article  PubMed  CAS  Google Scholar 

  13. Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319(5870):1668–72.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  14. Buratti E, Baralle FE. The multiple roles of TDP-43 in pre-mRNA processing and gene expression regulation. RNA Biol. 2010;7(4):420–9.

    Article  PubMed  CAS  Google Scholar 

  15. Ratti A, Buratti E. Physiological functions and pathobiology of TDP-43 and FUS/TLS proteins. J Neurochem. 2016;138(Suppl 1):95–111.

    Article  PubMed  CAS  Google Scholar 

  16. Brandmeir NJ, Geser F, Kwong LK, Zimmerman E, Qian J, Lee VM, et al. Severe subcortical TDP-43 pathology in sporadic frontotemporal lobar degeneration with motor neuron disease. Acta Neuropathol. 2008;115(1):123–31.

    Article  PubMed  Google Scholar 

  17. Neumann M, Kwong LK, Truax AC, Vanmassenhove B, Kretzschmar HA, Van Deerlin VM, et al. TDP-43-positive white matter pathology in frontotemporal lobar degeneration with ubiquitin-positive inclusions. J Neuropathol Exp Neurol. 2007;66(3):177–83.

    Article  PubMed  CAS  Google Scholar 

  18. Sanelli T, Xiao S, Horne P, Bilbao J, Zinman L, Robertson J. Evidence that TDP-43 is not the major ubiquitinated target within the pathological inclusions of amyotrophic lateral sclerosis. J Neuropathol Exp Neurol. 2007;66(12):1147–53.

    Article  PubMed  Google Scholar 

  19. Hasegawa M, Arai T, Nonaka T, Kametani F, Yoshida M, Hashizume Y, et al. Phosphorylated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Ann Neurol. 2008;64(1):60–70.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Ling SC, Polymenidou M, Cleveland DW. Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron. 2013;79(3):416–38.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Mackenzie IR, Neumann M. Reappraisal of TDP-43 pathology in FTLD-U subtypes. Acta Neuropathol. 2017b;134(1):79–96.

    Article  PubMed  CAS  Google Scholar 

  22. Mackenzie IR, Bigio EH, Ince PG, Geser F, Neumann M, Cairns NJ, et al. Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol. 2007;61(5):427–34.

    Article  PubMed  CAS  Google Scholar 

  23. Maekawa S, Leigh PN, King A, Jones E, Steele JC, Bodi I, et al. TDP-43 is consistently co-localized with ubiquitinated inclusions in sporadic and Guam amyotrophic lateral sclerosis but not in familial amyotrophic lateral sclerosis with and without SOD1 mutations. Neuropathology. 2009;29(6):672–83.

    Article  PubMed  Google Scholar 

  24. Robertson J, Sanelli T, Xiao S, Yang W, Horne P, Hammond R, et al. Lack of TDP-43 abnormalities in mutant SOD1 transgenic mice shows disparity with ALS. Neurosci Lett. 2007;420(2):128–32.

    Article  PubMed  CAS  Google Scholar 

  25. Braak H, Ludolph AC, Neumann M, Ravits J, Del Tredici K. Pathological TDP-43 changes in Betz cells differ from those in bulbar and spinal alpha-motoneurons in sporadic amyotrophic lateral sclerosis. Acta Neuropathol. 2017;133(1):79–90.

    Article  PubMed  CAS  Google Scholar 

  26. Mackenzie IR, Neumann M. Molecular neuropathology of frontotemporal dementia: insights into disease mechanisms from postmortem studies. J Neurochem. 2016;138(Suppl 1):54–70.

    Article  PubMed  CAS  Google Scholar 

  27. Josephs KA, Hodges JR, Snowden JS, Mackenzie IR, Neumann M, Mann DM, et al. Neuropathological background of phenotypical variability in frontotemporal dementia. Acta Neuropathol. 2011;122(2):137–53.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Mackenzie IR, Neumann M, Baborie A, Sampathu DM, Du Plessis D, Jaros E, et al. A harmonized classification system for FTLD-TDP pathology. Acta Neuropathol. 2011;122(1):111–3.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Mackenzie IRA, Neumann M. Fused in sarcoma neuropathology in neurodegenerative disease. Cold Spring Harb Perspect Med. 2017a;7(12). https://doi.org/10.1101/cshperspect.a024299.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Murray ME, DeJesus-Hernandez M, Rutherford NJ, Baker M, Duara R, Graff-Radford NR, et al. Clinical and neuropathologic heterogeneity of c9FTD/ALS associated with hexanucleotide repeat expansion in C9ORF72. Acta Neuropathol. 2011;122(6):673–90.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Ash PE, Bieniek KF, Gendron TF, Caulfield T, Lin WL, Dejesus-Hernandez M, et al. Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron. 2013;77(4):639–46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Mori K, Arzberger T, Grasser FA, Gijselinck I, May S, Rentzsch K, et al. Bidirectional transcripts of the expanded C9orf72 hexanucleotide repeat are translated into aggregating dipeptide repeat proteins. Acta Neuropathol. 2013;126(6):881–93.

    Article  PubMed  CAS  Google Scholar 

  33. Zu T, Liu Y, Banez-Coronel M, Reid T, Pletnikova O, Lewis J, et al. RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia. Proc Natl Acad Sci U S A. 2013;110(51):E4968–77.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Davidson YS, Barker H, Robinson AC, Thompson JC, Harris J, Troakes C, et al. Brain distribution of dipeptide repeat proteins in frontotemporal lobar degeneration and motor neurone disease associated with expansions in C9ORF72. Acta Neuropathol Commun. 2014;2:70.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Mackenzie IR, Frick P, Grasser FA, Gendron TF, Petrucelli L, Cashman NR, et al. Quantitative analysis and clinico-pathological correlations of different dipeptide repeat protein pathologies in C9ORF72 mutation carriers. Acta Neuropathol. 2015;130(6):845–61.

    Article  PubMed  CAS  Google Scholar 

  36. Davidson Y, Robinson AC, Liu X, Wu D, Troakes C, Rollinson S, et al. Neurodegeneration in frontotemporal lobar degeneration and motor neurone disease associated with expansions in C9orf72 is linked to TDP-43 pathology and not associated with aggregated forms of dipeptide repeat proteins. Neuropathol Appl Neurobiol. 2016;42(3):242–54.

    Article  PubMed  CAS  Google Scholar 

  37. Saberi S, Stauffer JE, Jiang J, Garcia SD, Taylor AE, Schulte D, et al. Sense-encoded poly-GR dipeptide repeat proteins correlate to neurodegeneration and uniquely co-localize with TDP-43 in dendrites of repeat-expanded C9orf72 amyotrophic lateral sclerosis. Acta Neuropathol. 2018;135(3):459–74.

    Article  PubMed  CAS  Google Scholar 

  38. Amador-Ortiz C, Lin WL, Ahmed Z, Personett D, Davies P, Duara R, et al. TDP-43 immunoreactivity in hippocampal sclerosis and Alzheimer’s disease. Ann Neurol. 2007;61(5):435–45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Higashi S, Iseki E, Yamamoto R, Minegishi M, Hino H, Fujisawa K, et al. Concurrence of TDP-43, tau and alpha-synuclein pathology in brains of Alzheimer’s disease and dementia with Lewy bodies. Brain Res. 2007;1184:284–94.

    Google Scholar 

  40. Josephs KA, Murray ME, Whitwell JL, Parisi JE, Petrucelli L, Jack CR, et al. Staging TDP-43 pathology in Alzheimer’s disease. Acta Neuropathol. 2014;127(3):441–50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Josephs KA, Whitwell JL, Knopman DS, Hu WT, Stroh DA, Baker M, et al. Abnormal TDP-43 immunoreactivity in AD modifies clinicopathologic and radiologic phenotype. Neurology. 2008;70(19 Pt 2):1850–7.

    Article  PubMed  CAS  Google Scholar 

  42. Schwab C, Arai T, Hasegawa M, Yu S, McGeer PL. Colocalization of transactivation-responsive DNA-binding protein 43 and huntingtin in inclusions of Huntington disease. J Neuropathol Exp Neurol. 2008;67(12):1159–65.

    Article  Google Scholar 

  43. Nakashima-Yasuda H, Uryu K, Robinson J, Xie SX, Hurtig H, Duda JE, et al. Co-morbidity of TDP-43 proteinopathy in Lewy body related diseases. Acta Neuropathol (Berl). 2007; 114(3):221–9.

    Article  PubMed  CAS  Google Scholar 

  44. Lin WL, Dickson DW. Ultrastructural localization of TDP-43 in filamentous neuronal inclusions in various neurodegenerative diseases. Acta Neuropathol. 2008;116(2):205–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Arai T, Mackenzie IR, Hasegawa M, Nonoka T, Niizato K, Tsuchiya K, et al. Phosphorylated TDP-43 in Alzheimer’s disease and dementia with Lewy bodies. Acta Neuropathol. 2009;117(2):125–36.

    Article  PubMed  CAS  Google Scholar 

  46. Fujishiro H, Uchikado H, Arai T, Hasegawa M, Akiyama H, Yokota O, et al. Accumulation of phosphorylated TDP-43 in brains of patients with argyrophilic grain disease. Acta Neuropathol. 2009;117(2):151–8.

    Article  PubMed  CAS  Google Scholar 

  47. McKee AC, Daneshvar DH, Alvarez VE, Stein TD. The neuropathology of sport. Acta Neuropathol. 2014;127(1):29–51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Ou SH, Wu F, Harrich D, Garcia-Martinez LF, Gaynor RB. Cloning and characterization of a novel cellular protein, TDP-43, that binds to human immunodeficiency virus type 1 TAR DNA sequence motifs. J Virol. 1995;69(6):3584–96.

    PubMed  PubMed Central  CAS  Google Scholar 

  49. Buratti E, Baralle FE. Characterization and functional implications of the RNA binding properties of nuclear factor TDP-43, a novel splicing regulator of CFTR exon 9. J Biol Chem. 2001;276(39):36337–43.

    Article  PubMed  CAS  Google Scholar 

  50. Buratti E, Dork T, Zuccato E, Pagani F, Romano M, Baralle FE. Nuclear factor TDP-43 and SR proteins promote in vitro and in vivo CFTR exon 9 skipping. EMBO J. 2001;20(7):1774–84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Ayala YM, Pantano S, D’Ambrogio A, Buratti E, Brindisi A, Marchetti C, et al. Human, Drosophila, and C. elegans TDP43: nucleic acid binding properties and splicing regulatory function. J Mol Biol. 2005;348(3):575–88.

    Article  PubMed  CAS  Google Scholar 

  52. Kraemer BC, Schuck T, Wheeler JM, Robinson LC, Trojanowski JQ, Lee VM, et al. Loss of murine TDP-43 disrupts motor function and plays an essential role in embryogenesis. Acta Neuropathol. 2010;119(4):409–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Sephton CF, Good SK, Atkin S, Dewey CM, Mayer P 3rd, Herz J, et al. TDP-43 is a developmentally regulated protein essential for early embryonic development. J Biol Chem. 2010;285(9):6826–34.

    Article  PubMed  CAS  Google Scholar 

  54. Wu LS, Cheng WC, Hou SC, Yan YT, Jiang ST, Shen CK. TDP-43, a neuro-pathosignature factor, is essential for early mouse embryogenesis. Genesis. 2010;48(1):56–62.

    PubMed  CAS  Google Scholar 

  55. Lim L, Wei Y, Lu Y, Song J. ALS-causing mutations significantly perturb the self-assembly and interaction with nucleic acid of the intrinsically disordered prion-like domain of TDP-43. PLoS Biol. 2016;14(1):e1002338.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Qin H, Lim LZ, Wei Y, Song J. TDP-43 N terminus encodes a novel ubiquitin-like fold and its unfolded form in equilibrium that can be shifted by binding to ssDNA. Proc Natl Acad Sci U S A. 2014;111(52):18619–24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Mompean M, Baralle M, Buratti E, Laurents DV. An amyloid-like pathological conformation of TDP-43 is stabilized by hypercooperative hydrogen bonds. Front Mol Neurosci. 2016b;9:125.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Budini M, Baralle FE, Buratti E. Targeting TDP-43 in neurodegenerative diseases. Expert Opin Ther Targets. 2014;18(6):617–32.

    Article  PubMed  CAS  Google Scholar 

  59. Buratti E, Brindisi A, Giombi M, Tisminetzky S, Ayala YM, Baralle FE. TDP-43 binds heterogeneous nuclear ribonucleoprotein A/B through its C-terminal tail: an important region for the inhibition of cystic fibrosis transmembrane conductance regulator exon 9 splicing. J Biol Chem. 2005;280(45):37572–84.

    Article  PubMed  CAS  Google Scholar 

  60. Cushman M, Johnson BS, King OD, Gitler AD, Shorter J. Prion-like disorders: blurring the divide between transmissibility and infectivity. J Cell Sci. 2010;123(Pt 8):1191–201.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. D’Ambrogio A, Buratti E, Stuani C, Guarnaccia C, Romano M, Ayala YM, et al. Functional mapping of the interaction between TDP-43 and hnRNP A2 in vivo. Nucleic Acids Res. 2009;37(12):4116–26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Jiang LL, Che MX, Zhao J, Zhou CJ, Xie MY, Li HY, et al. Structural transformation of the amyloidogenic core region of TDP-43 protein initiates its aggregation and cytoplasmic inclusion. J Biol Chem. 2013;288(27):19614–24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Budini M, Buratti E, Stuani C, Guarnaccia C, Romano V, De Conti L, et al. Cellular model of TAR DNA-binding protein 43 (TDP-43) aggregation based on its C-terminal Gln/Asn-rich region. J Biol Chem. 2012;287(10):7512–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Fuentealba RA, Udan M, Bell S, Wegorzewska I, Shao J, Diamond MI, et al. Interaction with polyglutamine aggregates reveals a Q/N-rich domain in TDP-43. J Biol Chem. 2010;285(34):26304–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Mompean M, Buratti E, Guarnaccia C, Brito RM, Chakrabartty A, Baralle FE, et al. Structural characterization of the minimal segment of TDP-43 competent for aggregation. Arch Biochem Biophys. 2014;545:53–62.

    Article  PubMed  CAS  Google Scholar 

  66. Mompean M, Hervas R, Xu Y, Tran TH, Guarnaccia C, Buratti E, et al. Structural evidence of amyloid fibril formation in the putative aggregation domain of TDP-43. J Phys Chem Lett. 2015;6(13):2608–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Ayala YM, Zago P, Ambrogio A, Xu Y-F, Petrucelli L, Buratti E, et al. Structural determinants of the cellular localization and shuttling of TDP-43. J Cell Sci. 2008;121(22):3778.

    Article  PubMed  CAS  Google Scholar 

  68. Winton MJ, Igaz LM, Wong MM, Kwong LK, Trojanowski JQ, Lee VM. Disturbance of nuclear and cytoplasmic TAR DNA-binding protein (TDP-43) induces disease-like redistribution, sequestration, and aggregate formation. J Biol Chem. 2008a;283(19):13302–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Winton MJ, Van Deerlin VM, Kwong LK, Yuan W, Wood EM, Yu CE, et al. A90V TDP-43 variant results in the aberrant localization of TDP-43 in vitro. FEBS Lett. 2008b;582(15):2252–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Ayala YM, De Conti L, Avendano-Vazquez SE, Dhir A, Romano M, D’Ambrogio A, et al. TDP-43 regulates its mRNA levels through a negative feedback loop. EMBO J. 2011;30(2):277–88.

    Article  PubMed  CAS  Google Scholar 

  71. Polymenidou M, Lagier-Tourenne C, Hutt KR, Huelga SC, Moran J, Liang TY, et al. Long pre-mRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43. Nat Neurosci. 2011;14(4):459–68.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Lukavsky PJ, Daujotyte D, Tollervey JR, Ule J, Stuani C, Buratti E, et al. Molecular basis of UG-rich RNA recognition by the human splicing factor TDP-43. Nat Struct Mol Biol. 2013;20(12):1443–9.

    Article  PubMed  CAS  Google Scholar 

  73. Afroz T, Hock EM, Ernst P, Foglieni C, Jambeau M, Gilhespy LAB, et al. Functional and dynamic polymerization of the ALS-linked protein TDP-43 antagonizes its pathologic aggregation. Nat Commun. 2017;8(1):45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Jiang LL, Xue W, Hong JY, Zhang JT, Li MJ, Yu SN, et al. The N-terminal dimerization is required for TDP-43 splicing activity. Sci Rep. 2017;7(1):6196.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Shiina Y, Arima K, Tabunoki H, Satoh J. TDP-43 dimerizes in human cells in culture. Cell Mol Neurobiol. 2010;30(4):641–52.

    Article  PubMed  CAS  Google Scholar 

  76. Zhang YJ, Caulfield T, Xu YF, Gendron TF, Hubbard J, Stetler C, et al. The dual functions of the extreme N-terminus of TDP-43 in regulating its biological activity and inclusion formation. Hum Mol Genet. 2013;22(15):3112–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Chang CK, Wu TH, Wu CY, Chiang MH, Toh EK, Hsu YC, et al. The N-terminus of TDP-43 promotes its oligomerization and enhances DNA binding affinity. Biochem Biophys Res Commun. 2012;425(2):219–24.

    Article  PubMed  CAS  Google Scholar 

  78. Kralovicova J, Patel A, Searle M, Vorechovsky I. The role of short RNA loops in recognition of a single-hairpin exon derived from a mammalian-wide interspersed repeat. RNA Biol. 2015;12(1):54–69.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Moreno F, Rabinovici GD, Karydas A, Miller Z, Hsu SC, Legati A, et al. A novel mutation P112H in the TARDBP gene associated with frontotemporal lobar degeneration without motor neuron disease and abundant neuritic amyloid plaques. Acta Neuropathol Commun. 2015;3:19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Maurel C, Madji-Hounoum B, Thepault RA, Marouillat S, Brulard C, Danel-Brunaud V, et al. Mutation in the RRM2 domain of TDP-43 in Amyotrophic Lateral Sclerosis with rapid progression associated with ubiquitin positive aggregates in cultured motor neurons. Amyotroph Lateral Scler Frontotemporal Degener. 2018;19(1–2):149–51.

    Article  PubMed  CAS  Google Scholar 

  81. Kuo PH, Chiang CH, Wang YT, Doudeva LG, Yuan HS. The crystal structure of TDP-43 RRM1-DNA complex reveals the specific recognition for UG- and TG-rich nucleic acids. Nucleic Acids Res. 2014;42(7):4712–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Chiang CH, Grauffel C, Wu LS, Kuo PH, Doudeva LG, Lim C, et al. Structural analysis of disease-related TDP-43 D169G mutation: linking enhanced stability and caspase cleavage efficiency to protein accumulation. Sci Rep. 2016;6:21581.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Zhang YJ, Xu YF, Cook C, Gendron TF, Roettges P, Link CD, et al. Aberrant cleavage of TDP-43 enhances aggregation and cellular toxicity. Proc Natl Acad Sci U S A. 2009;106(18):7607–12.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Sephton CF, Cenik C, Kucukural A, Dammer EB, Cenik B, Han Y, et al. Identification of neuronal RNA targets of TDP-43-containing ribonucleoprotein complexes. J Biol Chem. 2011;286(2):1204–15.

    Article  PubMed  CAS  Google Scholar 

  85. Tollervey JR, Wang Z, Hortobagyi T, Witten JT, Zarnack K, Kayikci M, et al. Analysis of alternative splicing associated with aging and neurodegeneration in the human brain. Genome Res. 2011;21(10):1572–82.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Xiao S, Sanelli T, Dib S, Sheps D, Findlater J, Bilbao J, et al. RNA targets of TDP-43 identified by UV-CLIP are deregulated in ALS. Mol Cell Neurosci. 2011;47(3):167–80.

    Article  PubMed  CAS  Google Scholar 

  87. Prudencio M, Jansen-West KR, Lee WC, Gendron TF, Zhang YJ, Xu YF, et al. Misregulation of human sortilin splicing leads to the generation of a nonfunctional progranulin receptor. Proc Natl Acad Sci U S A. 2012;109(52):21510–5.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Pickering-Brown SM. Progranulin and frontotemporal lobar degeneration. Acta Neuropathol. 2007;114(1):39–47.

    Article  PubMed  Google Scholar 

  89. Rosenblum LT, Trotti D. EAAT2 and the molecular signature of amyotrophic lateral sclerosis. Adv Neurobiol. 2017;16:117–36.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Rothstein JD, Van Kammen M, Levey AI, Martin LJ, Kuncl RW. Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis. Ann Neurol. 1995;38(1):73–84.

    Article  PubMed  CAS  Google Scholar 

  91. Colombrita C, Onesto E, Megiorni F, Pizzuti A, Baralle FE, Buratti E, et al. TDP-43 and FUS RNA-binding proteins bind distinct sets of cytoplasmic messenger RNAs and differently regulate their post-transcriptional fate in motoneuron-like cells. J Biol Chem. 2012;287(19):15635–47.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Honda D, Ishigaki S, Iguchi Y, Fujioka Y, Udagawa T, Masuda A, et al. The ALS/FTLD-related RNA-binding proteins TDP-43 and FUS have common downstream RNA targets in cortical neurons. FEBS Open Bio. 2013;4:1–10.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Wang IF, Wu LS, Chang HY, Shen CK. TDP-43, the signature protein of FTLD-U, is a neuronal activity-responsive factor. J Neurochem. 2008;105(3):797–806.

    Article  PubMed  CAS  Google Scholar 

  94. Fallini C, Bassell GJ, Rossoll W. The ALS disease protein TDP-43 is actively transported in motor neuron axons and regulates axon outgrowth. Hum Mol Genet. 2012;21(16):3703–18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Coyne AN, Siddegowda BB, Estes PS, Johannesmeyer J, Kovalik T, Daniel SG, et al. Futsch/MAP1B mRNA is a translational target of TDP-43 and is neuroprotective in a Drosophila model of amyotrophic lateral sclerosis. J Neurosci. 2014;34(48):15962–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Ishiguro A, Kimura N, Watanabe Y, Watanabe S, Ishihama A. TDP-43 binds and transports G-quadruplex-containing mRNAs into neurites for local translation. Genes Cells. 2016;21(5):466–81.

    Article  PubMed  CAS  Google Scholar 

  97. Narayanan RK, Mangelsdorf M, Panwar A, Butler TJ, Noakes PG, Wallace RH. Identification of RNA bound to the TDP-43 ribonucleoprotein complex in the adult mouse brain. Amyotroph Lateral Scler Frontotemporal Degener. 2013;14(4):252–60.

    Article  PubMed  CAS  Google Scholar 

  98. Alami NH, Smith RB, Carrasco MA, Williams LA, Winborn CS, Han SSW, et al. Axonal transport of TDP-43 mRNA granules is impaired by ALS-causing mutations. Neuron. 2014;81(3):536–43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Diaper DC, Adachi Y, Sutcliffe B, Humphrey DM, Elliott CJ, Stepto A, et al. Loss and gain of Drosophila TDP-43 impair synaptic efficacy and motor control leading to age-related neurodegeneration by loss-of-function phenotypes. Hum Mol Genet. 2013;22(8):1539–57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Feiguin F, Godena VK, Romano G, D’Ambrogio A, Klima R, Baralle FE. Depletion of TDP-43 affects Drosophila motoneurons terminal synapsis and locomotive behavior. FEBS Lett. 2009;583(10):1586–92.

    Article  PubMed  CAS  Google Scholar 

  101. Romano G, Klima R, Buratti E, Verstreken P, Baralle FE, Feiguin F. Chronological requirements of TDP-43 function in synaptic organization and locomotive control. Neurobiol Dis. 2014;71:95–109.

    Article  PubMed  CAS  Google Scholar 

  102. Chand KK, Lee KM, Lee JD, Qiu H, Willis EF, Lavidis NA, et al. Defects in synaptic transmission at the neuromuscular junction precedes motor deficits in a TDP-43(Q331K) transgenic mouse model of amyotrophic lateral sclerosis. FASEB J. 2018. https://doi.org/10.1096/fj.201700835R.

  103. Fogarty MJ, Klenowski PM, Lee JD, Drieberg-Thompson JR, Bartlett SE, Ngo ST, et al. Cortical synaptic and dendritic spine abnormalities in a presymptomatic TDP-43 model of amyotrophic lateral sclerosis. Sci Rep. 2016;6:37968.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Handley EE, Pitman KA, Dawkins E, Young KM, Clark RM, Jiang TC, et al. Synapse dysfunction of layer V pyramidal neurons precedes neurodegeneration in a mouse model of TDP-43 proteinopathies. Cereb Cortex. 2017;27(7):3630–47.

    PubMed  Google Scholar 

  105. Medina DX, Orr ME, Oddo S. Accumulation of C-terminal fragments of transactive response DNA-binding protein 43 leads to synaptic loss and cognitive deficits in human TDP-43 transgenic mice. Neurobiol Aging. 2014;35(1):79–87.

    Article  PubMed  CAS  Google Scholar 

  106. Ling JP, Pletnikova O, Troncoso JC, Wong PC. TDP-43 repression of nonconserved cryptic exons is compromised in ALS-FTD. Science. 2015;349(6248):650–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Jeong YH, Ling JP, Lin SZ, Donde AN, Braunstein KE, Majounie E, et al. Tdp-43 cryptic exons are highly variable between cell types. Mol Neurodegener. 2017;12(1):13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. MacNair L, Xiao S, Miletic D, Ghani M, Julien JP, Keith J, et al. MTHFSD and DDX58 are novel RNA-binding proteins abnormally regulated in amyotrophic lateral sclerosis. Brain. 2016;139(Pt 1):86–100.

    Article  PubMed  Google Scholar 

  109. Swarup V, Phaneuf D, Bareil C, Robertson J, Rouleau GA, Kriz J, et al. Pathological hallmarks of amyotrophic lateral sclerosis/frontotemporal lobar degeneration in transgenic mice produced with TDP-43 genomic fragments. Brain. 2011;134(Pt 9):2610–26.

    Article  PubMed  Google Scholar 

  110. Bakkar N, Kovalik T, Lorenzini I, Spangler S, Lacoste A, Sponaugle K, et al. Artificial intelligence in neurodegenerative disease research: use of IBM Watson to identify additional RNA-binding proteins altered in amyotrophic lateral sclerosis. Acta Neuropathol. 2018;135(2):227–47.

    Article  PubMed  CAS  Google Scholar 

  111. Wang W, Wang L, Lu J, Siedlak SL, Fujioka H, Liang J, et al. The inhibition of TDP-43 mitochondrial localization blocks its neuronal toxicity. Nat Med. 2016;22(8):869–78.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Magrane J, Cortez C, Gan WB, Manfredi G. Abnormal mitochondrial transport and morphology are common pathological denominators in SOD1 and TDP43 ALS mouse models. Hum Mol Genet. 2014;23(6):1413–24.

    Article  PubMed  CAS  Google Scholar 

  113. Vande Velde C, McDonald KK, Boukhedimi Y, McAlonis-Downes M, Lobsiger CS, Bel Hadj S, et al. Misfolded SOD1 associated with motor neuron mitochondria alters mitochondrial shape and distribution prior to clinical onset. PLoS One. 2011;6(7):e22031.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Wang W, Li L, Lin WL, Dickson DW, Petrucelli L, Zhang T, et al. The ALS disease-associated mutant TDP-43 impairs mitochondrial dynamics and function in motor neurons. Hum Mol Genet. 2013;22(23):4706–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Hong K, Li Y, Duan W, Guo Y, Jiang H, Li W, et al. Full-length TDP-43 and its C-terminal fragments activate mitophagy in NSC34 cell line. Neurosci Lett. 2012;530(2):144–9.

    Article  PubMed  CAS  Google Scholar 

  116. Onesto E, Colombrita C, Gumina V, Borghi MO, Dusi S, Doretti A, et al. Gene-specific mitochondria dysfunctions in human TARDBP and C9ORF72 fibroblasts. Acta Neuropathol Commun. 2016;4(1):47.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Stribl C, Samara A, Trümbach D, Peis R, Neumann M, Fuchs H, et al. Mitochondrial dysfunction and decrease in body weight of a transgenic knock-in mouse model for TDP-43. J Biol Chem. 2014;289(15):10769–84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Xu Y-F, Zhang Y-J, Lin W-L, Cao X, Stetler C, Dickson DW, et al. Expression of mutant TDP-43 induces neuronal dysfunction in transgenic mice. Mol Neurodegener. 2011;6:73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Xu Y-F, Gendron TF, Zhang Y-J, Lin W-L, Alton S, Sheng H, et al. Wild-type human TDP-43 expression causes TDP-43 phosphorylation, mitochondrial aggregation, motor deficits, and early mortality in transgenic mice. J Neurosci. 2010;30(32):10851.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Duan W, Li X, Shi J, Guo Y, Li Z, Li C. Mutant TAR DNA-binding protein-43 induces oxidative injury in motor neuron-like cell. Neuroscience. 2010;169(4):1621–9.

    Article  PubMed  CAS  Google Scholar 

  121. Lu J, Duan W, Guo Y, Jiang H, Li Z, Huang J, et al. Mitochondrial dysfunction in human TDP-43 transfected NSC34 cell lines and the protective effect of dimethoxy curcumin. Brain Res Bull. 2012;89(5):185–90.

    Article  PubMed  CAS  Google Scholar 

  122. Smith EF, Shaw PJ, De Vos KJ. The role of mitochondria in amyotrophic lateral sclerosis. Neurosci Lett. 2017. https://doi.org/10.1016/j.neulet.2017.06.052.

  123. Buratti E. Functional significance of TDP-43 mutations in disease. Adv Genet. 2015;91:1–53.

    PubMed  Google Scholar 

  124. McGoldrick P, Joyce PI, Fisher EM, Greensmith L. Rodent models of amyotrophic lateral sclerosis. Biochim Biophys Acta. 2013;1832(9):1421–36.

    Article  PubMed  CAS  Google Scholar 

  125. Johnson BS, McCaffery JM, Lindquist S, Gitler AD. A yeast TDP-43 proteinopathy model: exploring the molecular determinants of TDP-43 aggregation and cellular toxicity. Proc Natl Acad Sci U S A. 2008;105(17):6439–44.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Johnson BS, Snead D, Lee JJ, McCaffery JM, Shorter J, Gitler AD. TDP-43 is intrinsically aggregation-prone, and amyotrophic lateral sclerosis-linked mutations accelerate aggregation and increase toxicity. J Biol Chem. 2009;284(30):20329–39.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. King OD, Gitler AD, Shorter J. The tip of the iceberg: RNA-binding proteins with prion-like domains in neurodegenerative disease. Brain Res. 2012;1462:61–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Mackenzie IR, Nicholson AM, Sarkar M, Messing J, Purice MD, Pottier C, et al. TIA1 mutations in amyotrophic lateral sclerosis and frontotemporal dementia promote phase separation and alter stress granule dynamics. Neuron. 2017;95(4):808–16.e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Maziuk B, Ballance HI, Wolozin B. Dysregulation of RNA binding protein aggregation in neurodegenerative disorders. Front Mol Neurosci. 2017;10:89.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Protter DS, Parker R. Principles and properties of stress granules. Trends Cell Biol. 2016;26(9):668–79.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Sephton CF, Yu G. The function of RNA-binding proteins at the synapse: implications for neurodegeneration. Cell Mol Life Sci. 2015;72(19):3621–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Parker R, Sheth U. P bodies and the control of mRNA translation and degradation. Mol Cell. 2007;25(5):635–46.

    Article  PubMed  CAS  Google Scholar 

  133. Anderson P, Kedersha N. Stress granules. Curr Biol. 2009;19(10):R397–8.

    Article  PubMed  CAS  Google Scholar 

  134. Buchan JR, Parker R. Eukaryotic stress granules: the ins and outs of translation. Mol Cell. 2009;36(6):932–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Holt CE, Bullock SL. Subcellular mRNA localization in animal cells and why it matters. Science. 2009;326(5957):1212–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. Brangwynne CP, Eckmann CR, Courson DS, Rybarska A, Hoege C, Gharakhani J, et al. Germline P granules are liquid droplets that localize by controlled dissolution/condensation. Science. 2009;324(5935):1729–32.

    Article  PubMed  CAS  Google Scholar 

  137. Brangwynne CP, Mitchison TJ, Hyman AA. Active liquid-like behavior of nucleoli determines their size and shape in Xenopus laevis oocytes. Proc Natl Acad Sci U S A. 2011;108(11):4334–9.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Conicella AE, Zerze GH, Mittal J, Fawzi NL. ALS mutations disrupt phase separation mediated by alpha-helical structure in the TDP-43 low-complexity C-terminal domain. Structure. 2016;24(9):1537–49.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  139. Kato M, Han TW, Xie S, Shi K, Du X, Wu LC, et al. Cell-free formation of RNA granules: low complexity sequence domains form dynamic fibers within hydrogels. Cell. 2012;149(4):753–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Lin Y, Protter DS, Rosen MK, Parker R. Formation and maturation of phase-separated liquid droplets by RNA-binding proteins. Mol Cell. 2015;60(2):208–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Molliex A, Temirov J, Lee J, Coughlin M, Kanagaraj AP, Kim HJ, et al. Phase separation by low complexity domains promotes stress granule assembly and drives pathological fibrillization. Cell. 2015;163(1):123–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Murakami T, Qamar S, Lin JQ, Schierle GS, Rees E, Miyashita A, et al. ALS/FTD mutation-induced phase transition of FUS liquid droplets and reversible hydrogels into irreversible hydrogels impairs RNP granule function. Neuron. 2015;88(4):678–90.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Patel A, Lee HO, Jawerth L, Maharana S, Jahnel M, Hein MY, et al. A liquid-to-solid phase transition of the ALS protein FUS accelerated by disease mutation. Cell. 2015;162(5):1066–77.

    Article  PubMed  CAS  Google Scholar 

  144. McInerney GM, Kedersha NL, Kaufman RJ, Anderson P, Liljeström P. Importance of eIF2α phosphorylation and stress granule assembly in alphavirus translation regulation. Mol Biol Cell. 2005;16(8):3753–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. White JP, Lloyd RE. Regulation of stress granules in virus systems. Trends Microbiol. 2012;20(4):175–83.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. White JP, Cardenas AM, Marissen WE, Lloyd RE. Inhibition of cytoplasmic mRNA stress granule formation by a viral proteinase. Cell Host Microbe. 2007;2(5):295–305.

    Article  PubMed  CAS  Google Scholar 

  147. Aulas A, Vande Velde C. Alterations in stress granule dynamics driven by TDP-43 and FUS: a link to pathological inclusions in ALS? Front Cell Neurosci. 2015;9:423.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  148. Markmiller S, Soltanieh S, Server KL, Mak R, Jin W, Fang MY, et al. Context-dependent and disease-specific diversity in protein interactions within stress granules. Cell. 2018;172(3):590–604.e13.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  149. Jain S, Wheeler JR, Walters RW, Agrawal A, Barsic A, Parker R. ATPase-modulated stress granules contain a diverse proteome and substructure. Cell. 2016;164(3):487–98.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  150. Wheeler JR, Matheny T, Jain S, Abrisch R, Parker R. Distinct stages in stress granule assembly and disassembly. Elife. 2016;5. https://doi.org/10.7554/eLife.18413.

  151. Colombrita C, Zennaro E, Fallini C, Weber M, Sommacal A, Buratti E, et al. TDP-43 is recruited to stress granules in conditions of oxidative insult. J Neurochem. 2009;111(4):1051–61.

    Article  PubMed  CAS  Google Scholar 

  152. Dewey CM, Cenik B, Sephton CF, Dries DR, Mayer P 3rd, Good SK, et al. TDP-43 is directed to stress granules by sorbitol, a novel physiological osmotic and oxidative stressor. Mol Cell Biol. 2011;31(5):1098–108.

    Article  PubMed  CAS  Google Scholar 

  153. Meyerowitz J, Parker SJ, Vella LJ, Ng D, Price KA, Liddell JR, et al. C-Jun N-terminal kinase controls TDP-43 accumulation in stress granules induced by oxidative stress. Mol Neurodegener. 2011;6:57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Parker SJ, Meyerowitz J, James JL, Liddell JR, Crouch PJ, Kanninen KM, et al. Endogenous TDP-43 localized to stress granules can subsequently form protein aggregates. Neurochem Int. 2012;60(4):415–24.

    Article  PubMed  CAS  Google Scholar 

  155. Liu-Yesucevitz L, Bilgutay A, Zhang YJ, Vanderweyde T, Citro A, Mehta T, et al. Tar DNA binding protein-43 (TDP-43) associates with stress granules: analysis of cultured cells and pathological brain tissue. PLoS One. 2010b;5(10):e13250.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. McDonald KK, Aulas A, Destroismaisons L, Pickles S, Beleac E, Camu W, et al. TAR DNA-binding protein 43 (TDP-43) regulates stress granule dynamics via differential regulation of G3BP and TIA-1. Hum Mol Genet. 2011;20(7):1400–10.

    Article  PubMed  CAS  Google Scholar 

  157. Liu-Yesucevitz L, Lin AY, Ebata A, Boon JY, Reid W, Xu YF, et al. ALS-linked mutations enlarge TDP-43-enriched neuronal RNA granules in the dendritic arbor. J Neurosci. 2014;34(12):4167–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  158. Liu-Yesucevitz L, Bilgutay A, Zhang YJ, Vanderwyde T, Citro A, Mehta T, et al. Tar DNA binding protein-43 (TDP-43) associates with stress granules: analysis of cultured cells and pathological brain tissue. PLoS One. 2010a;5(10):e13250.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  159. Volkening K, Leystra-Lantz C, Yang W, Jaffee H, Strong MJ. Tar DNA binding protein of 43 kDa (TDP-43), 14-3-3 proteins and copper/zinc superoxide dismutase (SOD1) interact to modulate NFL mRNA stability. Implications for altered RNA processing in amyotrophic lateral sclerosis (ALS). Brain Res. 2009;1305:168–82.

    Article  PubMed  CAS  Google Scholar 

  160. Buchan JR, Kolaitis RM, Taylor JP, Parker R. Eukaryotic stress granules are cleared by autophagy and Cdc48/VCP function. Cell. 2013;153(7):1461–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Hardy J, Rogaeva E. Motor neuron disease and frontotemporal dementia: sometimes related, sometimes not. Exp Neurol. 2014;262(Pt B):75–83.

    Article  PubMed  CAS  Google Scholar 

  162. Auburger G, Sen N-E, Meierhofer D, Başak A-N, Gitler AD. Efficient prevention of neurodegenerative diseases by depletion of starvation response factor ataxin-2. Trends Neurosci. 2017;40(8):507–16.

    Article  PubMed  CAS  Google Scholar 

  163. Kaehler C, Isensee J, Nonhoff U, Terrey M, Hucho T, Lehrach H, et al. Ataxin-2-like is a regulator of stress granules and processing bodies. PLoS One. 2012;7(11):e50134.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  164. Nonhoff U, Ralser M, Welzel F, Piccini I, Balzereit D, Yaspo M-L, et al. Ataxin-2 interacts with the DEAD/H-box RNA helicase DDX6 and interferes with P-bodies and stress granules. Mol Biol Cell. 2007;18(4):1385–96.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  165. Lorenzetti D, Bohlega S, Zoghbi HY. The expansion of the CAG repeat in ataxin-2 is a frequent cause of autosomal dominant spinocerebellar ataxia. Neurology. 1997;49(4):1009.

    Article  PubMed  CAS  Google Scholar 

  166. Imbert G, Saudou F, Yvert G, Devys D, Trottier Y, Garnier J-M, et al. Cloning of the gene for spinocerebellar ataxia 2 reveals a locus with high sensitivity to expanded CAG/glutamine repeats. Nat Genet. 1996;14:285.

    Article  CAS  PubMed  Google Scholar 

  167. Sanpei K, Takano H, Igarashi S, Sato T, Oyake M, Sasaki H, et al. Identification of the spinocerebellar ataxia type 2 gene using a direct identification of repeat expansion and cloning technique, DIRECT. Nat Genet. 1996;14:277.

    Article  CAS  PubMed  Google Scholar 

  168. Pulst S-M, Nechiporuk A, Nechiporuk T, Gispert S, Chen X-N, Lopes-Cendes I, et al. Moderate expansion of a normally biallelic trinucleotide repeat in spinocerebellar ataxia type 2. Nat Genet. 1996;14:269.

    Article  CAS  PubMed  Google Scholar 

  169. Nanetti L, Fancellu R, Tomasello C, Gellera C, Pareyson D, Mariotti C. Rare association of motor neuron disease and spinocerebellar ataxia type 2 (SCA2): a new case and review of the literature. J Neurol. 2009;256(11):1926–8.

    Article  PubMed  Google Scholar 

  170. Elden AC, Kim H-J, Hart MP, Chen-Plotkin AS, Johnson BS, Fang X, et al. Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature. 2010;466:1069.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  171. Bonini NM, Gitler AD. Model organisms reveal insight into human neurodegenerative disease: ataxin-2 intermediate-length polyglutamine expansions are a risk factor for ALS. J Mol Neurosci. 2011;45(3):676–83.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  172. Becker LA, Huang B, Bieri G, Ma R, Knowles DA, Jafar-Nejad P, et al. Therapeutic reduction of ataxin-2 extends lifespan and reduces pathology in TDP-43 mice. Nature. 2017;544(7650):367–71.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Lastres-Becker I, Nonis D, Eich F, Klinkenberg M, Gorospe M, Kotter P, et al. Mammalian ataxin-2 modulates translation control at the pre-initiation complex via PI3K/mTOR and is induced by starvation. Biochim Biophys Acta. 2016;1862(9):1558–69.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  174. Xiao S, Sanelli T, Chiang H, Sun Y, Chakrabartty A, Keith J, et al. Low molecular weight species of TDP-43 generated by abnormal splicing form inclusions in amyotrophic lateral sclerosis and result in motor neuron death. Acta Neuropathol. 2015;130(1):49–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  175. Dormann D, Capell A, Carlson AM, Shankaran SS, Rodde R, Neumann M, et al. Proteolytic processing of TAR DNA binding protein-43 by caspases produces C-terminal fragments with disease defining properties independent of progranulin. J Neurochem. 2009;110(3):1082–94.

    Article  PubMed  CAS  Google Scholar 

  176. Igaz LM, Kwong LK, Chen-Plotkin A, Winton MJ, Unger TL, Xu Y, et al. Expression of TDP-43 C-terminal fragments in vitro recapitulates pathological features of TDP-43 proteinopathies. J Biol Chem. 2009;284(13):8516–24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  177. Herskowitz JH, Gozal YM, Duong DM, Dammer EB, Gearing M, Ye K, et al. Asparaginyl endopeptidase cleaves TDP-43 in brain. Proteomics. 2012;12(15–16):2455–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Kametani F, Obi T, Shishido T, Akatsu H, Murayama S, Saito Y, et al. Mass spectrometric analysis of accumulated TDP-43 in amyotrophic lateral sclerosis brains. Sci Rep. 2016;6:23281.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  179. Tsuji H, Arai T, Kametani F, Nonaka T, Yamashita M, Suzukake M, et al. Molecular analysis and biochemical classification of TDP-43 proteinopathy. Brain. 2012a;135(Pt 11):3380–91.

    Article  PubMed  Google Scholar 

  180. Tsuji H, Nonaka T, Yamashita M, Masuda-Suzukake M, Kametani F, Akiyama H, et al. Epitope mapping of antibodies against TDP-43 and detection of protease-resistant fragments of pathological TDP-43 in amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Biochem Biophys Res Commun. 2012b;417(1):116–21.

    Article  PubMed  CAS  Google Scholar 

  181. Mompean M, Romano V, Pantoja-Uceda D, Stuani C, Baralle FE, Buratti E, et al. The TDP-43 N-terminal domain structure at high resolution. FEBS J. 2016a;283(7):1242–60.

    Article  PubMed  CAS  Google Scholar 

  182. Nishimoto Y, Ito D, Yagi T, Nihei Y, Tsunoda Y, Suzuki N. Characterization of alternative isoforms and inclusion body of the TAR DNA-binding protein-43. J Biol Chem. 2010;285(1):608–19.

    Article  PubMed  CAS  Google Scholar 

  183. Che MX, Jiang YJ, Xie YY, Jiang LL, Hu HY. Aggregation of the 35-kDa fragment of TDP-43 causes formation of cytoplasmic inclusions and alteration of RNA processing. FASEB J. 2011;25(7):2344–53.

    Article  PubMed  CAS  Google Scholar 

  184. D’Alton S, Altshuler M, Lewis J. Studies of alternative isoforms provide insight into TDP-43 autoregulation and pathogenesis. RNA. 2015;21(8):1419–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  185. Wang HY, Wang IF, Bose J, Shen CK. Structural diversity and functional implications of the eukaryotic TDP gene family. Genomics. 2004;83(1):130–9.

    Article  PubMed  CAS  Google Scholar 

  186. Che MX, Jiang LL, Li HY, Jiang YJ, Hu HY. TDP-35 sequesters TDP-43 into cytoplasmic inclusions through binding with RNA. FEBS Lett. 2015;589(15):1920–8.

    Article  PubMed  CAS  Google Scholar 

  187. Sasaguri H, Chew J, Xu YF, Gendron TF, Garrett A, Lee CW, et al. The extreme N-terminus of TDP-43 mediates the cytoplasmic aggregation of TDP-43 and associated toxicity in vivo. Brain Res. 2016;1647:57–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  188. Wei Y, Lim L, Wang L, Song J. Inter-domain interactions of TDP-43 as decoded by NMR. Biochem Biophys Res Commun. 2016;473(2):614–9.

    Article  PubMed  CAS  Google Scholar 

  189. Yang C, Tan W, Whittle C, Qiu L, Cao L, Akbarian S, et al. The C-terminal TDP-43 fragments have a high aggregation propensity and harm neurons by a dominant-negative mechanism. PLoS One. 2010;5(12):e15878.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  190. Budini M, Romano V, Quadri Z, Buratti E, Baralle FE. TDP-43 loss of cellular function through aggregation requires additional structural determinants beyond its C-terminal Q/N prion-like domain. Hum Mol Genet. 2015;24(1):9–20.

    Article  PubMed  CAS  Google Scholar 

  191. Brady OA, Meng P, Zheng Y, Mao Y, Hu F. Regulation of TDP-43 aggregation by phosphorylation andp62/SQSTM1. J Neurochem. 2011;116(2):248–59.

    Article  PubMed  CAS  Google Scholar 

  192. Kim SH, Shanware NP, Bowler MJ, Tibbetts RS. Amyotrophic lateral sclerosis-associated proteins TDP-43 and FUS/TLS function in a common biochemical complex to co-regulate HDAC6 mRNA. J Biol Chem. 2010;285(44):34097–105.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  193. Nonaka T, Kametani F, Arai T, Akiyama H, Hasegawa M. Truncation and pathogenic mutations facilitate the formation of intracellular aggregates of TDP-43. Hum Mol Genet. 2009;18(18):3353–64.

    Article  PubMed  CAS  Google Scholar 

  194. Mashiko T, Sakashita E, Kasashima K, Tominaga K, Kuroiwa K, Nozaki Y, et al. Developmentally regulated RNA-binding protein 1 (Drb1)/RNA-binding motif protein 45 (RBM45), a nuclear-cytoplasmic trafficking protein, forms TAR DNA-binding protein 43 (TDP-43)-mediated cytoplasmic aggregates. J Biol Chem. 2016;291(29):14996–5007.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  195. Chen H-J, Mitchell JC, Novoselov S, Miller J, Nishimura AL, Scotter EL, et al. The heat shock response plays an important role in TDP-43 clearance: evidence for dysfunction in amyotrophic lateral sclerosis. Brain. 2016;139(5):1417–32.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Yamashita M, Nonaka T, Arai T, Kametani F, Buchman VL, Ninkina N, et al. Methylene blue and dimebon inhibit aggregation of TDP-43 in cellular models. FEBS Lett. 2009;583(14):2419–24.

    Article  PubMed  CAS  Google Scholar 

  197. Voigt A, Herholz D, Fiesel FC, Kaur K, MĂĽller D, Karsten P, et al. TDP-43-mediated neuron loss in vivo requires RNA-binding activity. PLoS One. 2010;5(8):e12247.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  198. Cohen TJ, Hwang AW, Restrepo CR, Yuan C-X, Trojanowski JQ, Lee VMY. An acetylation switch controls TDP-43 function and aggregation propensity. Nat Commun. 2015;6:5845.

    Article  PubMed  CAS  Google Scholar 

  199. Scotter EL, Vance C, Nishimura AL, Lee Y-B, Chen H-J, Urwin H, et al. Differential roles of the ubiquitin proteasome system and autophagy in the clearance of soluble and aggregated TDP-43 species. J Cell Sci. 2014;127(6):1263–78.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  200. Suzuki H, Shibagaki Y, Hattori S, Matsuoka M. Nuclear TDP-43 causes neuronal toxicity by escaping from the inhibitory regulation by hnRNPs. Hum Mol Genet. 2015;24(6):1513–27.

    Article  PubMed  CAS  Google Scholar 

  201. Bentmann E, Neumann M, Tahirovic S, Rodde R, Dormann D, Haass C. Requirements for stress granule recruitment of fused in sarcoma (FUS) and TAR DNA-binding protein of 43 kDa (TDP-43). J Biol Chem. 2012;287(27):23079–94.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  202. Li HY, Yeh PA, Chiu HC, Tang CY, Tu BP. Hyperphosphorylation as a defense mechanism to reduce TDP-43 aggregation. PLoS One. 2011;6(8):e23075.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  203. Boeynaems S, Bogaert E, Michiels E, Gijselinck I, Sieben A, Jovicic A, et al. Drosophila screen connects nuclear transport genes to DPR pathology in c9ALS/FTD. Sci Rep. 2016;6:20877.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Freibaum BD, Lu Y, Lopez-Gonzalez R, Kim NC, Almeida S, Lee KH, et al. GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature. 2015;525(7567):129–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  205. Jovicic A, Mertens J, Boeynaems S, Bogaert E, Chai N, Yamada SB, et al. Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. Nat Neurosci. 2015;18(9):1226–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  206. Zhang K, Donnelly CJ, Haeusler AR, Grima JC, Machamer JB, Steinwald P, et al. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature. 2015;525(7567):56–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  207. Chou CC, Zhang Y, Umoh ME, Vaughan SW, Lorenzini I, Liu F, et al. TDP-43 pathology disrupts nuclear pore complexes and nucleocytoplasmic transport in ALS/FTD. Nat Neurosci. 2018;21(2):228–39.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  208. Woerner AC, Frottin F, Hornburg D, Feng LR, Meissner F, Patra M, et al. Cytoplasmic protein aggregates interfere with nucleocytoplasmic transport of protein and RNA. Science. 2016;351(6269):173–6.

    Article  CAS  PubMed  Google Scholar 

  209. de Calignon A, Polydoro M, Suarez-Calvet M, William C, Adamowicz DH, Kopeikina KJ, et al. Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron. 2012;73(4):685–97.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  210. Liu L, Drouet V, Wu JW, Witter MP, Small SA, Clelland C, et al. Trans-synaptic spread of tau pathology in vivo. PLoS One. 2012;7(2):e31302.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  211. Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, Trojanowski JQ, et al. Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science. 2012;338(6109):949–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  212. Mougenot AL, Bencsik A, Nicot S, Vulin J, Morignat E, Verchere J, et al. Transmission of prion strains in a transgenic mouse model overexpressing human A53T mutated alpha-synuclein. J Neuropathol Exp Neurol. 2011;70(5):377–85.

    Article  PubMed  CAS  Google Scholar 

  213. Kaufman SK, Sanders DW, Thomas TL, Ruchinskas AJ, Vaquer-Alicea J, Sharma AM, et al. Tau prion strains dictate patterns of cell pathology, progression rate, and regional vulnerability in vivo. Neuron. 2016;92(4):796–812.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  214. Sanders DW, Kaufman SK, DeVos SL, Sharma AM, Mirbaha H, Li A, et al. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron. 2014;82(6):1271–88.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  215. Ishii T, Kawakami E, Endo K, Misawa H, Watabe K. Formation and spreading of TDP-43 aggregates in cultured neuronal and glial cells demonstrated by time-lapse imaging. PLoS One. 2017;12(6):e0179375.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  216. Pokrishevsky E, Grad LI, Cashman NR. TDP-43 or FUS-induced misfolded human wild-type SOD1 can propagate intercellularly in a prion-like fashion. Sci Rep. 2016;6:22155.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  217. Nonaka T, Masuda-Suzukake M, Arai T, Hasegawa Y, Akatsu H, Obi T, et al. Prion-like properties of pathological TDP-43 aggregates from diseased brains. Cell Rep. 2013;4(1):124–34.

    Article  PubMed  CAS  Google Scholar 

  218. Smethurst P, Sidle KC, Hardy J. Review: Prion-like mechanisms of transactive response DNA binding protein of 43 kDa (TDP-43) in amyotrophic lateral sclerosis (ALS). Neuropathol Appl Neurobiol. 2015;41(5):578–97.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Janice Robertson .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer International Publishing AG, part of Springer Nature

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Shenouda, M., Zhang, A.B., Weichert, A., Robertson, J. (2018). Mechanisms Associated with TDP-43 Neurotoxicity in ALS/FTLD. In: Sattler, R., Donnelly, C. (eds) RNA Metabolism in Neurodegenerative Diseases. Advances in Neurobiology, vol 20. Springer, Cham. https://doi.org/10.1007/978-3-319-89689-2_9

Download citation

Publish with us

Policies and ethics