Skip to main content

Trends in Understanding the Pathological Roles of TDP-43 and FUS Proteins

  • Chapter
  • First Online:
Frontotemporal Dementias

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 1281))

Abstract

Following the discovery of TDP-43 and FUS involvement in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar dementia (FTLD), the major challenge in the field has been to understand their physiological functions, both in normal and disease conditions. The hope is that this knowledge will improve our understanding of disease and lead to the development of effective therapeutic options. Initially, the focus has been directed at characterizing the role of these proteins in the control of RNA metabolism, because the main function of TDP-43 and FUS is to bind coding and noncoding RNAs to regulate their life cycle within cells. As a result, we now have an in-depth picture of the alterations that occur in RNA metabolism following their aggregation in various ALS/FTLD models and, to a somewhat lesser extent, in patients’ brains. In parallel, progress has been made with regard to understanding how aggregation of these proteins occurs in neurons, how it can spread in different brain regions, and how these changes affect various metabolic cellular pathways to result in neuronal death. The aim of this chapter will be to provide a general overview of the trending topics in TDP-43 and FUS investigations and to highlight what might represent the most promising avenues of research in the years to come.

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 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
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

Similar content being viewed by others

Abbreviations

ALS:

Amyotrophic lateral sclerosis

FTLD:

Frontotemporal lobar dementia

FUS/TLS:

Fused in sarcoma/translocated in liposarcoma

hnRNP:

Heterogeneous ribonucleoproteins

lncRNA:

Long noncoding RNA

mRNA:

Messenger RNA

miRNA:

MicroRNA

TDP-43:

TAR DNA binding protein 43 kDa

References

  1. Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT et al (2006) Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314(5796):130–133

    Article  CAS  PubMed  Google Scholar 

  2. Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, Mori H et al (2006) TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun 351(3):602–611

    Article  CAS  PubMed  Google Scholar 

  3. Kwiatkowski TJ Jr, Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C et al (2009) Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323(5918):1205–1208

    Article  CAS  PubMed  Google Scholar 

  4. Vance C, Rogelj B, Hortobagyi T, De Vos KJ, Nishimura AL, Sreedharan J et al (2009) Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 323(5918):1208–1211

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Neumann M, Bentmann E, Dormann D, Jawaid A, DeJesus-Hernandez M, Ansorge O et al (2011) FET proteins TAF15 and EWS are selective markers that distinguish FTLD with FUS pathology from amyotrophic lateral sclerosis with FUS mutations. Brain 134(Pt 9):2595–2609

    Article  PubMed  PubMed Central  Google Scholar 

  6. Kim HJ, Kim NC, Wang YD, Scarborough EA, Moore J, Diaz Z et al (2013) Mutations in prion-like domains in hnRNPA2B1 and hnRNPA1 cause multisystem proteinopathy and ALS. Nature 495(7442):467–473

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Johnson JO, Pioro EP, Boehringer A, Chia R, Feit H, Renton AE et al (2014) Mutations in the Matrin 3 gene cause familial amyotrophic lateral sclerosis. Nat Neurosci 17(5):664–666

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Elden AC, Kim HJ, Hart MP, Chen-Plotkin AS, Johnson BS, Fang X et al (2010) Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 466(7310):1069–1075

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yuan Z, Jiao B, Hou L, Xiao T, Liu X, Wang J et al (2018) Mutation analysis of the TIA1 gene in Chinese patients with amyotrophic lateral sclerosis and frontotemporal dementia. Neurobiol Aging 64:160 e9–160e12

    Article  CAS  Google Scholar 

  11. Zhang K, Liu Q, Shen D, Tai H, Fu H, Liu S et al (2018) Genetic analysis of TIA1 gene in Chinese patients with amyotrophic lateral sclerosis. Neurobiol Aging 67:201 e9–201e10

    Article  CAS  Google Scholar 

  12. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ et al (2011) Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72(2):245–256

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Renton AE, Majounie E, Waite A, Simon-Sanchez J, Rollinson S, Gibbs JR et al (2011) A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72(2):257–268

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Mori K, Lammich S, Mackenzie IR, Forne I, Zilow S, Kretzschmar H et al (2013) hnRNP A3 binds to GGGGCC repeats and is a constituent of p62-positive/TDP43-negative inclusions in the hippocampus of patients with C9orf72 mutations. Acta Neuropathol 125(3):413–423

    Article  CAS  PubMed  Google Scholar 

  15. Cooper-Knock J, Walsh MJ, Higginbottom A, Robin Highley J, Dickman MJ, Edbauer D et al (2014) Sequestration of multiple RNA recognition motif-containing proteins by C9orf72 repeat expansions. Brain 137(Pt 7):2040–2051

    Article  PubMed  PubMed Central  Google Scholar 

  16. Lee YB, Chen HJ, Peres JN, Gomez-Deza J, Attig J, Stalekar M et al (2013) Hexanucleotide repeats in ALS/FTD form length-dependent RNA foci, sequester RNA binding proteins, and are neurotoxic. Cell Rep 5(5):1178–1186

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Balendra R, Isaacs AM (2018) C9orf72-mediated ALS and FTD: multiple pathways to disease. Nat Rev Neurol 14(9):544–558

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Vatsavayai SC, Nana AL, Yokoyama JS, Seeley WW (2019) C9orf72-FTD/ALS pathogenesis: evidence from human neuropathological studies. Acta Neuropathol 137(1):1–26

    Article  CAS  PubMed  Google Scholar 

  19. Renoux AJ, Todd PK (2012) Neurodegeneration the RNA way. Prog Neurobiol 97(2):173–189

    Article  CAS  PubMed  Google Scholar 

  20. Sephton CF, Yu G (2015) The function of RNA-binding proteins at the synapse: implications for neurodegeneration. Cell Mol Life Sci 72(19):3621–3635

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhao M, Kim JR, van Bruggen R, Park J (2018) RNA-binding proteins in amyotrophic lateral sclerosis. Mol Cells 41(9):818–829

    PubMed  PubMed Central  Google Scholar 

  22. Butti Z, Patten SA (2018) RNA dysregulation in amyotrophic lateral sclerosis. Front Genet 9:712

    Article  CAS  PubMed  Google Scholar 

  23. Nussbacher JK, Tabet R, Yeo GW, Lagier-Tourenne C (2019) Disruption of RNA metabolism in neurological diseases and emerging therapeutic interventions. Neuron 102(2):294–320

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  25. Solomon DA, Mitchell JC, Salcher-Konrad MT, Vance CA, Mizielinska S (2019) Review: modelling the pathology and behaviour of frontotemporal dementia. Neuropathol Appl Neurobiol 45(1):58–80

    Article  CAS  PubMed  Google Scholar 

  26. Penndorf D, Witte OW, Kretz A (2018) DNA plasticity and damage in amyotrophic lateral sclerosis. Neural Regen Res 13(2):173–180

    Article  PubMed  PubMed Central  Google Scholar 

  27. Chabot B, Shkreta L (2016) Defective control of pre-messenger RNA splicing in human disease. J Cell Biol 212(1):13–27

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Fahrenkrog B, Harel A (2018) Perturbations in traffic: aberrant nucleocytoplasmic transport at the heart of neurodegeneration. Cell 7(12):232

    Article  CAS  Google Scholar 

  29. Burk K, Pasterkamp RJ (2019) Disrupted neuronal trafficking in amyotrophic lateral sclerosis. Acta Neuropathol 137(6):859–877

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rot G, Wang Z, Huppertz I, Modic M, Lence T, Hallegger M et al (2017) High-resolution RNA maps suggest common principles of splicing and polyadenylation regulation by TDP-43. Cell Rep 19(5):1056–1067

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Higashi S, Kabuta T, Nagai Y, Tsuchiya Y, Akiyama H, Wada K (2013) TDP-43 associates with stalled ribosomes and contributes to cell survival during cellular stress. J Neurochem 126(2):288–300

    Article  CAS  PubMed  Google Scholar 

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

    Google Scholar 

  33. Majumder P, Chen YT, Bose JK, Wu CC, Cheng WC, Cheng SJ et al (2012) TDP-43 regulates the mammalian spinogenesis through translational repression of Rac1. Acta Neuropathol 124(2):231–245

    Article  CAS  PubMed  Google Scholar 

  34. Fiesel FC, Weber SS, Supper J, Zell A, Kahle PJ (2012) TDP-43 regulates global translational yield by splicing of exon junction complex component SKAR. Nucleic Acids Res 40(6):2668–2682

    Article  CAS  PubMed  Google Scholar 

  35. Liu G, Coyne AN, Pei F, Vaughan S, Chaung M, Zarnescu DC et al (2017) Endocytosis regulates TDP-43 toxicity and turnover. Nat Commun 8(1):2092

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Beers DR, Appel SH (2019) Immune dysregulation in amyotrophic lateral sclerosis: mechanisms and emerging therapies. Lancet Neurol 18(2):211–220

    Article  CAS  PubMed  Google Scholar 

  37. Trageser KJ, Smith C, Herman FJ, Ono K, Pasinetti GM (2019) Mechanisms of immune activation by c9orf72-expansions in amyotrophic lateral sclerosis and frontotemporal dementia. Front Neurosci 13:1298

    Article  PubMed  PubMed Central  Google Scholar 

  38. Borroni B, Alberici A, Buratti E (2019) Review: molecular pathology of frontotemporal lobar degenerations. Neuropathol Appl Neurobiol 45(1):41–57

    Article  CAS  PubMed  Google Scholar 

  39. Briston T, Hicks AR (2018) Mitochondrial dysfunction and neurodegenerative proteinopathies: mechanisms and prospects for therapeutic intervention. Biochem Soc Trans 46(4):829–842

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Aulas A, Vande VC (2015) Alterations in stress granule dynamics driven by TDP-43 and FUS: a link to pathological inclusions in ALS? Front Cell Neurosci 9:423

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Bentmann E, Haass C, Dormann D (2013) Stress granules in neurodegeneration – lessons learnt from TAR DNA binding protein of 43 kDa and fused in sarcoma. FEBS J 280(18):4348–4370

    Article  CAS  PubMed  Google Scholar 

  42. Chew J, Cook C, Gendron TF, Jansen-West K, Del Rosso G, Daughrity LM et al (2019) Aberrant deposition of stress granule-resident proteins linked to C9orf72-associated TDP-43 proteinopathy. Mol Neurodegener 14(1):9

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Zhang P, Fan B, Yang P, Temirov J, Messing J, Kim HJ et al (2020) Chronic optogenetic induction of stress granules is cytotoxic and reveals the evolution of ALS-FTD pathology. Elife 8:e39578

    Google Scholar 

  44. Bennett SA, Tanaz R, Cobos SN, Torrente MP (2019) Epigenetics in amyotrophic lateral sclerosis: a role for histone post-translational modifications in neurodegenerative disease. Transl Res 204:19–30

    Article  CAS  PubMed  Google Scholar 

  45. Jawaid A, Khan R, Polymenidou M, Schulz PE (2018) Disease-modifying effects of metabolic perturbations in ALS/FTLD. Mol Neurodegener 13(1):63

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Joppe K, Roser AE, Maass F, Lingor P (2019) The contribution of iron to protein aggregation disorders in the central nervous system. Front Neurosci 13:15

    Article  PubMed  PubMed Central  Google Scholar 

  47. Francois-Moutal L, Perez-Miller S, Scott DD, Miranda VG, Mollasalehi N, Khanna M (2019) Structural insights Into TDP-43 and effects of post-translational modifications. Front Mol Neurosci 12:301

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Buratti E (2018) TDP-43 post-translational modifications in health and disease. Expert Opin Ther Targets 22(3):279–293

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  50. Afroz T, Perez-Berlanga M, Polymenidou M (2019) Structural transition, function and dysfunction of TDP-43 in neurodegenerative diseases. Chimia (Aarau) 73(6):380–390

    Article  CAS  Google Scholar 

  51. Mompean M, Romano V, Pantoja-Uceda D, Stuani C, Baralle FE, Buratti E et al (2016) The TDP-43 N-terminal domain structure at high resolution. FEBS J 283(7):1242–1260

    Article  CAS  PubMed  Google Scholar 

  52. Qin H, Lim LZ, Wei Y, Song J (2014) 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 111(52):18619–18624

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Winton MJ, Igaz LM, Wong MM, Kwong LK, Trojanowski JQ, Lee VM (2008) Disturbance of nuclear and cytoplasmic TAR DNA-binding protein (TDP-43) induces disease-like redistribution, sequestration, and aggregate formation. J Biol Chem 283(19):13302–13309

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Buratti E, Baralle FE (2001) 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 276(39):36337–36343

    Article  CAS  PubMed  Google Scholar 

  57. Lukavsky PJ, Daujotyte D, Tollervey JR, Ule J, Stuani C, Buratti E et al (2013) Molecular basis of UG-rich RNA recognition by the human splicing factor TDP-43. Nat Struct Mol Biol 20(12):1443–1449

    Article  CAS  PubMed  Google Scholar 

  58. Agrawal S, Kuo PH, Chu LY, Golzarroshan B, Jain M, Yuan HS (2019) RNA recognition motifs of disease-linked RNA-binding proteins contribute to amyloid formation. Sci Rep 9(1):6171

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Babinchak WM, Haider R, Dumm BK, Sarkar P, Surewicz K, Choi JK et al (2019) The role of liquid-liquid phase separation in aggregation of the TDP-43 low-complexity domain. J Biol Chem 294(16):6306–6317

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Crozat A, Aman P, Mandahl N, Ron D (1993) Fusion of CHOP to a novel RNA-binding protein in human myxoid liposarcoma. Nature 363(6430):640–644

    Article  CAS  PubMed  Google Scholar 

  61. Ichikawa H, Shimizu K, Hayashi Y, Ohki M (1994) An RNA-binding protein gene, TLS/FUS, is fused to ERG in human myeloid leukemia with t(16;21) chromosomal translocation. Cancer Res 54(11):2865–2868

    CAS  PubMed  Google Scholar 

  62. Calvio C, Neubauer G, Mann M, Lamond AI (1995) Identification of hnRNP P2 as TLS/FUS using electrospray mass spectrometry. RNA 1(7):724–733

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Morohoshi F, Ootsuka Y, Arai K, Ichikawa H, Mitani S, Munakata N et al (1998) Genomic structure of the human RBP56/hTAFII68 and FUS/TLS genes. Gene 221(2):191–198

    Article  CAS  PubMed  Google Scholar 

  64. Mackenzie IR, Neumann M (2012) FET proteins in frontotemporal dementia and amyotrophic lateral sclerosis. Brain Res 1462:40–43

    Article  CAS  PubMed  Google Scholar 

  65. Deng H, Gao K, Jankovic J (2014) The role of FUS gene variants in neurodegenerative diseases. Nat Rev Neurol 10(6):337–348

    Article  CAS  PubMed  Google Scholar 

  66. Yang L, Gal J, Chen J, Zhu H (2014) Self-assembled FUS binds active chromatin and regulates gene transcription. Proc Natl Acad Sci U S A 111(50):17809–17814

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Liu X, Niu C, Ren J, Zhang J, Xie X, Zhu H et al (2013) The RRM domain of human fused in sarcoma protein reveals a non-canonical nucleic acid binding site. Biochim Biophys Acta 1832(2):375–385

    Article  CAS  PubMed  Google Scholar 

  68. Loughlin FE, Lukavsky PJ, Kazeeva T, Reber S, Hock EM, Colombo M et al (2019) The solution structure of FUS bound to RNA reveals a bipartite mode of RNA recognition with both sequence and shape specificity. Mol Cell 73(3):490–504 e6

    Article  CAS  PubMed  Google Scholar 

  69. Rogelj B, Easton LE, Bogu GK, Stanton LW, Rot G, Curk T et al (2012) Widespread binding of FUS along nascent RNA regulates alternative splicing in the brain. Sci Rep 2:603

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Bhardwaj A, Myers MP, Buratti E, Baralle FE (2013) Characterizing TDP-43 interaction with its RNA targets. Nucleic Acids Res 41(9):5062–5074

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Polymenidou M, Lagier-Tourenne C, Hutt KR, Huelga SC, Moran J, Liang TY et al (2011) Long pre-mRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43. Nat Neurosci 14(4):459–468

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Colombrita C, Onesto E, Megiorni F, Pizzuti A, Baralle FE, Buratti E et al (2012) 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 287(19):15635–15647

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Lu Y, Lim L, Song J (2017) RRM domain of ALS/FTD-causing FUS characteristic of irreversible unfolding spontaneously self-assembles into amyloid fibrils. Sci Rep 7(1):1043

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Wang X, Schwartz JC, Cech TR (2015) Nucleic acid-binding specificity of human FUS protein. Nucleic Acids Res 43(15):7535–7543

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Rothstein JD (2007) TDP-43 in amyotrophic lateral sclerosis: pathophysiology or patho-babel? Ann Neurol 61(5):382–384

    Article  CAS  PubMed  Google Scholar 

  76. Kabashi E, Valdmanis PN, Dion P, Spiegelman D, McConkey BJ, Vande Velde C et al (2008) TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat Genet 40(5):572–574

    Article  CAS  PubMed  Google Scholar 

  77. Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B et al (2008) TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 319(5870):1668–1672

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Gitcho MA, Baloh RH, Chakraverty S, Mayo K, Norton JB, Levitch D et al (2008) TDP-43 A315T mutation in familial motor neuron disease. Ann Neurol 63(4):535–538

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Patel A, Lee HO, Jawerth L, Maharana S, Jahnel M, Hein MY et al (2015) A liquid-to-solid phase transition of the ALS protein FUS accelerated by disease mutation. Cell 162(5):1066–1077

    Article  CAS  PubMed  Google Scholar 

  80. Chen HJ, Topp SD, Hui HS, Zacco E, Katarya M, McLoughlin C et al (2019) RRM adjacent TARDBP mutations disrupt RNA binding and enhance TDP-43 proteinopathy. Brain 142(12):3753–3770

    Article  PubMed  PubMed Central  Google Scholar 

  81. Newell K, Paron F, Mompean M, Murrell J, Salis E, Stuani C et al (2019) Dysregulation of TDP-43 intracellular localization and early onset ALS are associated with a TARDBP S375G variant. Brain Pathol 29(3):397–413

    Article  CAS  PubMed  Google Scholar 

  82. Cohen TJ, Hwang AW, Restrepo CR, Yuan CX, Trojanowski JQ, Lee VM (2015) An acetylation switch controls TDP-43 function and aggregation propensity. Nat Commun 6:5845

    Article  CAS  PubMed  Google Scholar 

  83. Wang P, Wander CM, Yuan CX, Bereman MS, Cohen TJ (2017) Acetylation-induced TDP-43 pathology is suppressed by an HSF1-dependent chaperone program. Nat Commun 8(1):82

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Jiang T, Handley E, Brizuela M, Dawkins E, Lewis KEA, Clark RM et al (2019) Amyotrophic lateral sclerosis mutant TDP-43 may cause synaptic dysfunction through altered dendritic spine function. Dis Model Mech 12(5):dmm038109

    Google Scholar 

  85. Alami NH, Smith RB, Carrasco MA, Williams LA, Winborn CS, Han SS et al (2014) Axonal transport of TDP-43 mRNA granules is impaired by ALS-causing mutations. Neuron 81(3):536–543

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Naumann M, Peikert K, Gunther R, van der Kooi AJ, Aronica E, Hubers A et al (2019) Phenotypes and malignancy risk of different FUS mutations in genetic amyotrophic lateral sclerosis. Ann Clin Transl Neurol 6(12):2384–2394

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Lattante S, Rouleau GA, Kabashi E (2013) TARDBP and FUS mutations associated with amyotrophic lateral sclerosis: summary and update. Hum Mutat 34(6):812–826

    Article  CAS  PubMed  Google Scholar 

  88. Zhou Y, Liu S, Liu G, Ozturk A, Hicks GG (2013) ALS-associated FUS mutations result in compromised FUS alternative splicing and autoregulation. PLoS Genet 9(10):e1003895

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Yamazaki T, Chen S, Yu Y, Yan B, Haertlein TC, Carrasco MA et al (2012) FUS-SMN protein interactions link the motor neuron diseases ALS and SMA. Cell Rep 2(4):799–806

    Google Scholar 

  90. Tsuiji H, Iguchi Y, Furuya A, Kataoka A, Hatsuta H, Atsuta N et al (2013) Spliceosome integrity is defective in the motor neuron diseases ALS and SMA. EMBO Mol Med 5(2):221–234

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Sun S, Ling SC, Qiu J, Albuquerque CP, Zhou Y, Tokunaga S et al (2015) ALS-causative mutations in FUS/TLS confer gain and loss of function by altered association with SMN and U1-snRNP. Nat Commun 6:6171

    Article  CAS  PubMed  Google Scholar 

  92. Yu Y, Chi B, Xia W, Gangopadhyay J, Yamazaki T, Winkelbauer-Hurt ME et al (2015) U1 snRNP is mislocalized in ALS patient fibroblasts bearing NLS mutations in FUS and is required for motor neuron outgrowth in zebrafish. Nucleic Acids Res 43(6):3208–3218

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Hoell JI, Larsson E, Runge S, Nusbaum JD, Duggimpudi S, Farazi TA et al (2011) RNA targets of wild-type and mutant FET family proteins. Nat Struct Mol Biol 18(12):1428–1431

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Nishimoto Y, Nakagawa S, Hirose T, Okano HJ, Takao M, Shibata S et al (2013) The long non-coding RNA nuclear-enriched abundant transcript 1_2 induces paraspeckle formation in the motor neuron during the early phase of amyotrophic lateral sclerosis. Mol Brain 6:31

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Shelkovnikova TA, Robinson HK, Connor-Robson N, Buchman VL (2013) Recruitment into stress granules prevents irreversible aggregation of FUS protein mislocalized to the cytoplasm. Cell Cycle 12(19):3194–3202

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. De Santis R, Alfano V, de Turris V, Colantoni A, Santini L, Garone MG et al (2019) Mutant FUS and ELAVL4 (HuD) aberrant crosstalk in amyotrophic lateral sclerosis. Cell Rep 27(13):3818–31 e5

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Nishimura AL, Shum C, Scotter EL, Abdelgany A, Sardone V, Wright J et al (2014) Allele-specific knockdown of ALS-associated mutant TDP-43 in neural stem cells derived from induced pluripotent stem cells. PLoS One 9(3):e91269

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Guerrero EN, Mitra J, Wang H, Rangaswamy S, Hegde PM, Basu P et al (2019) Amyotrophic lateral sclerosis-associated TDP-43 mutation Q331K prevents nuclear translocation of XRCC4-DNA ligase 4 complex and is linked to genome damage-mediated neuronal apoptosis. Hum Mol Genet 28(18):3161–3162

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Naumann M, Pal A, Goswami A, Lojewski X, Japtok J, Vehlow A et al (2018) Impaired DNA damage response signaling by FUS-NLS mutations leads to neurodegeneration and FUS aggregate formation. Nat Commun 9(1):335

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. White MA, Kim E, Duffy A, Adalbert R, Phillips BU, Peters OM et al (2018) TDP-43 gains function due to perturbed autoregulation in a Tardbp knock-in mouse model of ALS-FTD. Nat Neurosci 21(4):552–563

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Fratta P, Sivakumar P, Humphrey J, Lo K, Ricketts T, Oliveira H et al (2018) Mice with endogenous TDP-43 mutations exhibit gain of splicing function and characteristics of amyotrophic lateral sclerosis. EMBO J 37(11):e98684

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Mackenzie IR, Bigio EH, Ince PG, Geser F, Neumann M, Cairns NJ et al (2007) Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol 61(5):427–434

    Article  CAS  PubMed  Google Scholar 

  103. Tan CF, Eguchi H, Tagawa A, Onodera O, Iwasaki T, Tsujino A et al (2007) TDP-43 immunoreactivity in neuronal inclusions in familial amyotrophic lateral sclerosis with or without SOD1 gene mutation. Acta Neuropathol (Berl) 113(5):535–542

    Article  CAS  Google Scholar 

  104. Ling SC, Polymenidou M, Cleveland DW (2013) Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron 79(3):416–438

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Ling SC, Albuquerque CP, Han JS, Lagier-Tourenne C, Tokunaga S, Zhou H et al (2010) ALS-associated mutations in TDP-43 increase its stability and promote TDP-43 complexes with FUS/TLS. Proc Natl Acad Sci U S A 107(30):13318–13323

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Kryndushkin D, Wickner RB, Shewmaker F (2011) FUS/TLS forms cytoplasmic aggregates, inhibits cell growth and interacts with TDP-43 in a yeast model of amyotrophic lateral sclerosis. Protein Cell 2(3):223–236

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Lee EB, Lee VM, Trojanowski JQ (2012) Gains or losses: molecular mechanisms of TDP43-mediated neurodegeneration. Nat Rev Neurosci 13(1):38–50

    Article  CAS  Google Scholar 

  108. Seyfried NT, Gozal YM, Dammer EB, Xia Q, Duong DM, Cheng D et al (2010) Multiplex SILAC analysis of a cellular TDP-43 proteinopathy model reveals protein inclusions associated with SUMOylation and diverse polyubiquitin chains. Mol Cell Proteomics 9(4):705–718

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Murnyak B, Bodoki L, Vincze M, Griger Z, Csonka T, Szepesi R et al (2015) Inclusion body myositis – pathomechanism and lessons from genetics. Open Med (Wars) 10(1):188–193

    CAS  Google Scholar 

  110. Dardis A, Zampieri S, Canterini S, Newell KL, Stuani C, Murrell JR et al (2016) Altered localization and functionality of TAR DNA Binding Protein 43 (TDP-43) in niemann-pick disease type C. Acta Neuropathol Commun 4(1):52

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Neumann M (2013) Frontotemporal lobar degeneration and amyotrophic lateral sclerosis: molecular similarities and differences. Rev Neurol (Paris) 169(10):793–798

    Article  CAS  Google Scholar 

  112. Monahan Z, Ryan VH, Janke AM, Burke KA, Rhoads SN, Zerze GH et al (2017) Phosphorylation of the FUS low-complexity domain disrupts phase separation, aggregation, and toxicity. EMBO J 36(20):2951–2967

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Capitini C, Conti S, Perni M, Guidi F, Cascella R, De Poli A et al (2014) TDP-43 inclusion bodies formed in bacteria are structurally amorphous, non-amyloid and inherently toxic to neuroblastoma cells. PLoS One 9(1):e86720

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Robinson JL, Geser F, Stieber A, Umoh M, Kwong LK, Van Deerlin VM et al (2013) TDP-43 skeins show properties of amyloid in a subset of ALS cases. Acta Neuropathol 125(1):121–131

    Article  CAS  PubMed  Google Scholar 

  115. Bigio EH, Wu JY, Deng HX, Bit-Ivan EN, Mao Q, Ganti R et al (2013) Inclusions in frontotemporal lobar degeneration with TDP-43 proteinopathy (FTLD-TDP) and amyotrophic lateral sclerosis (ALS), but not FTLD with FUS proteinopathy (FTLD-FUS), have properties of amyloid. Acta Neuropathol 125(3):463–465

    Article  PubMed  PubMed Central  Google Scholar 

  116. Chen AK, Lin RY, Hsieh EZ, Tu PH, Chen RP, Liao TY et al (2010) Induction of amyloid fibrils by the C-terminal fragments of TDP-43 in amyotrophic lateral sclerosis. J Am Chem Soc 132(4):1186–1187

    Article  CAS  PubMed  Google Scholar 

  117. Saini A, Chauhan VS (2011) Delineation of the core aggregation sequences of TDP-43 C-terminal fragment. Chembiochem 12(16):2495–2501

    Article  CAS  PubMed  Google Scholar 

  118. Saini A, Chauhan VS (2014) Self-assembling properties of peptides derived from TDP-43 C-terminal fragment. Langmuir 30(13):3845–3856

    Article  CAS  PubMed  Google Scholar 

  119. Guo W, Chen Y, Zhou X, Kar A, Ray P, Chen X et al (2011) An ALS-associated mutation affecting TDP-43 enhances protein aggregation, fibril formation and neurotoxicity Nat Struct Mol Biol 18(7):822–830

    Google Scholar 

  120. Sun CS, Wang CY, Chen BP, He RY, Liu GC, Wang CH et al (2014) The influence of pathological mutations and proline substitutions in TDP-43 glycine-rich peptides on its amyloid properties and cellular toxicity. PLoS One 9(8):e103644

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  122. Mompean M, Hervas R, Xu Y, Tran TH, Guarnaccia C, Buratti E et al (2015) Structural evidence of amyloid fibril formation in the putative aggregation domain of TDP-43. J Phys Chem Lett 6(13):2608–2615

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Shelkovnikova TA, Peters OM, Deykin AV, Connor-Robson N, Robinson H, Ustyugov AA et al (2013) Fused in sarcoma (FUS) protein lacking nuclear localization signal (NLS) and major RNA binding motifs triggers proteinopathy and severe motor phenotype in transgenic mice. J Biol Chem 288(35):25266–25274

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Ding X, Sun F, Chen J, Chen L, Tobin-Miyaji Y, Xue S et al (2020) Amyloid-forming segment induces aggregation of FUS-LC domain from phase separation modulated by site-specific phosphorylation. J Mol Biol 432(2):467–483

    Article  CAS  PubMed  Google Scholar 

  125. Buratti E, Baralle FE (2012) TDP-43: gumming up neurons through protein-protein and protein-RNA interactions. Trends Biochem Sci 37(6):237–247

    Article  CAS  PubMed  Google Scholar 

  126. Shelkovnikova TA (2013) Modelling FUSopathies: focus on protein aggregation. Biochem Soc Trans 41(6):1613–1617

    Article  CAS  PubMed  Google Scholar 

  127. Hans F, Glasebach H, Kahle PJ (2020) Multiple distinct pathways lead to hyperubiquitylated insoluble TDP-43 protein independent of its translocation into stress granules. J Biol Chem 295(3):673–689

    Article  CAS  PubMed  Google Scholar 

  128. Wolozin B, Ivanov P (2019) Stress granules and neurodegeneration. Nat Rev Neurosci 20(11):649–666

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Baradaran-Heravi Y, Van Broeckhoven C, van der Zee J (2020) Stress granule mediated protein aggregation and underlying gene defects in the FTD-ALS spectrum. Neurobiol Dis 134:104639

    Article  CAS  PubMed  Google Scholar 

  130. Colombrita C, Zennaro E, Fallini C, Weber M, Sommacal A, Buratti E et al (2009) TDP-43 is recruited to stress granules in conditions of oxidative insult. J Neurochem 111(4):1051–1061

    Article  CAS  PubMed  Google Scholar 

  131. Bosco DA, Lemay N, Ko HK, Zhou H, Burke C, Kwiatkowski TJ Jr et al (2010) Mutant FUS proteins that cause amyotrophic lateral sclerosis incorporate into stress granules. Hum Mol Genet 19(21):4160–4175

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Li YR, King OD, Shorter J, Gitler AD (2013) Stress granules as crucibles of ALS pathogenesis. J Cell Biol 201(3):361–372

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Dormann D, Haass C (2013) Fused in sarcoma (FUS): an oncogene goes awry in neurodegeneration. Mol Cell Neurosci 56:475–486

    Google Scholar 

  134. Fang MY, Markmiller S, Vu AQ, Javaherian A, Dowdle WE, Jolivet P et al (2019) Small-molecule modulation of TDP-43 recruitment to stress granules prevents persistent TDP-43 accumulation in ALS/FTD. Neuron 103(5):802–19 e11

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Blokhuis AM, Groen EJ, Koppers M, van den Berg LH, Pasterkamp RJ (2013) Protein aggregation in amyotrophic lateral sclerosis. Acta Neuropathol 125(6):777–794

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Fang YS, Tsai KJ, Chang YJ, Kao P, Woods R, Kuo PH et al (2014) Full-length TDP-43 forms toxic amyloid oligomers that are present in frontotemporal lobar dementia-TDP patients. Nat Commun 5:4824

    Article  CAS  PubMed  Google Scholar 

  137. Johnson BS, Snead D, Lee JJ, McCaffery JM, Shorter J, Gitler AD (2009) TDP-43 is intrinsically aggregation-prone, and amyotrophic lateral sclerosis-linked mutations accelerate aggregation and increase toxicity. J Biol Chem 284(30):20329–20339

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Woerner AC, Frottin F, Hornburg D, Feng LR, Meissner F, Patra M et al (2016) Cytoplasmic protein aggregates interfere with nucleo-cytoplasmic transport of protein and RNA. Science 351(6269):173–176

    Google Scholar 

  139. Dammer EB, Fallini C, Gozal YM, Duong DM, Rossoll W, Xu P et al (2012) Coaggregation of RNA-binding proteins in a model of TDP-43 proteinopathy with selective RGG motif methylation and a role for RRM1 ubiquitination. PLoS One 7(6):e38658

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Collins M, Riascos D, Kovalik T, An J, Krupa K, Hood BL et al (2012) The RNA-binding motif 45 (RBM45) protein accumulates in inclusion bodies in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP) patients. Acta Neuropathol 124(5):717–732

    Google Scholar 

  141. Shelkovnikova TA, Robinson HK, Troakes C, Ninkina N, Buchman VL (2014) Compromised paraspeckle formation as a pathogenic factor in FUSopathies. Hum Mol Genet 23(9):2298–2312

    Article  CAS  PubMed  Google Scholar 

  142. Cragnaz L, Klima R, Skoko N, Budini M, Feiguin F, Baralle FE (2014) Aggregate formation prevents dTDP-43 neurotoxicity in the Drosophila melanogaster eye. Neurobiol Dis 71:74–80

    Article  CAS  PubMed  Google Scholar 

  143. Leitman J, Ulrich Hartl F, Lederkremer GZ (2013) Soluble forms of polyQ-expanded huntingtin rather than large aggregates cause endoplasmic reticulum stress. Nat Commun 4:2753

    Article  PubMed  CAS  Google Scholar 

  144. Bolognesi B, Faure AJ, Seuma M, Schmiedel JM, Tartaglia GG, Lehner B (2019) The mutational landscape of a prion-like domain. Nat Commun 10(1):4162

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Wegorzewska I, Baloh RH (2011) TDP-43-based animal models of neurodegeneration: new insights into ALS pathology and pathophysiology. Neurodegener Dis 8(4):262–274

    Article  CAS  PubMed  Google Scholar 

  146. Armstrong GA, Drapeau P (2013) Loss and gain of FUS function impair neuromuscular synaptic transmission in a genetic model of ALS. Hum Mol Genet 22(21):4282–4292

    Article  CAS  PubMed  Google Scholar 

  147. Xu ZS (2012) Does a loss of TDP-43 function cause neurodegeneration? Mol Neurodegener 7:27

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Vanden Broeck L, Callaerts P, Dermaut B (2014) TDP-43-mediated neurodegeneration: towards a loss-of-function hypothesis? Trends Mol Med 20(2):66–71

    Article  CAS  Google Scholar 

  149. Kino Y, Washizu C, Kurosawa M, Yamada M, Miyazaki H, Akagi T et al (2015) FUS/TLS deficiency causes behavioral and pathological abnormalities distinct from amyotrophic lateral sclerosis. Acta Neuropathol Commun 3:24

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  150. Buratti E, Baralle FE (2009) The molecular links between TDP-43 dysfunction and neurodegeneration. Adv Genet 66:1–34

    Article  CAS  PubMed  Google Scholar 

  151. Halliday G, Bigio EH, Cairns NJ, Neumann M, Mackenzie IR, Mann DM (2012) Mechanisms of disease in frontotemporal lobar degeneration: gain of function versus loss of function effects. Acta Neuropathol 124(3):373–382

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Birsa N, Bentham MP, Fratta P (2020) Cytoplasmic functions of TDP-43 and FUS and their role in ALS. Semin Cell Dev Biol 99:193–201

    Google Scholar 

  153. Kawaguchi T, Rollins MG, Moinpour M, Morera AA, Ebmeier CC, Old WM et al (2020) Changes to the TDP-43 and FUS interactomes induced by DNA damage. J Proteome Res 19(1):360–370

    Article  CAS  PubMed  Google Scholar 

  154. Berson A, Sartoris A, Nativio R, Van Deerlin V, Toledo JB, Porta S et al (2017) TDP-43 promotes neurodegeneration by impairing chromatin remodeling. Curr Biol 27(23):3579–90 e6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Krug L, Chatterjee N, Borges-Monroy R, Hearn S, Liao WW, Morrill K et al (2017) Retrotransposon activation contributes to neurodegeneration in a Drosophila TDP-43 model of ALS. PLoS Genet 13(3):e1006635

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Guerrero EN, Mitra J, Wang H, Rangaswamy S, Hegde PM, Basu P et al (2019) Amyotrophic lateral sclerosis-associated TDP-43 mutation Q331K prevents nuclear translocation of XRCC4-DNA ligase 4 complex and is linked to genome damage-mediated neuronal apoptosis. Hum Mol Genet 28(5):2459–2476

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Mitra J, Guerrero EN, Hegde PM, Liachko NF, Wang H, Vasquez V et al (2019) Motor neuron disease-associated loss of nuclear TDP-43 is linked to DNA double-strand break repair defects. Proc Natl Acad Sci U S A 116(10):4696–4705

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Baechtold H, Kuroda M, Sok J, Ron D, Lopez BS, Akhmedov AT (1999) Human 75-kDa DNA-pairing protein is identical to the pro-oncoprotein TLS/FUS and is able to promote D-loop formation. J Biol Chem 274(48):34337–34342

    Article  CAS  PubMed  Google Scholar 

  159. Hicks GG, Singh N, Nashabi A, Mai S, Bozek G, Klewes L et al (2000) Fus deficiency in mice results in defective B-lymphocyte development and activation, high levels of chromosomal instability and perinatal death. Nat Genet 24(2):175–179

    Article  CAS  PubMed  Google Scholar 

  160. Gardiner M, Toth R, Vandermoere F, Morrice NA, Rouse J (2008) Identification and characterization of FUS/TLS as a new target of ATM. Biochem J 415(2):297–307

    Article  CAS  PubMed  Google Scholar 

  161. Rulten SL, Rotheray A, Green RL, Grundy GJ, Moore DA, Gomez-Herreros F et al (2014) PARP-1 dependent recruitment of the amyotrophic lateral sclerosis-associated protein FUS/TLS to sites of oxidative DNA damage. Nucleic Acids Res 42(1):307–314

    Article  CAS  PubMed  Google Scholar 

  162. Mastrocola AS, Kim SH, Trinh AT, Rodenkirch LA, Tibbetts RS (2013) The RNA-binding protein fused in sarcoma (FUS) functions downstream of poly(ADP-ribose) polymerase (PARP) in response to DNA damage. J Biol Chem 288(34):24731–24741

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Wang WY, Pan L, Su SC, Quinn EJ, Sasaki M, Jimenez JC et al (2013) Interaction of FUS and HDAC1 regulates DNA damage response and repair in neurons. Nat Neurosci 16(10):1383–1391

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Wang X, Arai S, Song X, Reichart D, Du K, Pascual G et al (2008) Induced ncRNAs allosterically modify RNA-binding proteins in cis to inhibit transcription. Nature 454(7200):126–130

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Ou SH, Wu F, Harrich D, Garcia-Martinez LF, Gaynor RB (1995) Cloning and characterization of a novel cellular protein, TDP-43, that binds to human immunodeficiency virus type 1 TAR DNA sequence motifs. J Virol 69(6):3584–3596

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Nehls J, Koppensteiner H, Brack-Werner R, Floss T, Schindler M (2014) HIV-1 replication in human immune cells is independent of TAR DNA binding protein 43 (TDP-43) expression. PLoS One 9(8):e105478

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  167. Abhyankar MM, Urekar C, Reddi PP (2007) A novel CpG-free vertebrate insulator silences the testis-specific SP-10 gene in somatic tissues: role for TDP-43 in insulator function. J Biol Chem 282(50):36143–36154

    Article  CAS  PubMed  Google Scholar 

  168. Murata H, Hattori T, Maeda H, Takashiba S, Takigawa M, Kido J et al (2015) Identification of transactivation-responsive DNA-binding protein 43 (TARDBP43; TDP-43) as a novel factor for TNF-alpha expression upon lipopolysaccharide stimulation in human monocytes. J Periodontal Res 50(4):452–460

    Article  CAS  PubMed  Google Scholar 

  169. Schwenk BM, Hartmann H, Serdaroglu A, Schludi MH, Hornburg D, Meissner F et al (2016) TDP-43 loss of function inhibits endosomal trafficking and alters trophic signaling in neurons. EMBO J 35(21):2350–2370

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Schwartz JC, Ebmeier CC, Podell ER, Heimiller J, Taatjes DJ, Cech TR (2012) FUS binds the CTD of RNA polymerase II and regulates its phosphorylation at Ser2. Genes Dev 26(24):2690–2695

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Schwartz JC, Podell ER, Han SS, Berry JD, Eggan KC, Cech TR (2014) FUS is sequestered in nuclear aggregates in ALS patient fibroblasts. Mol Biol Cell 25(17):2571–2578

    Article  PubMed  PubMed Central  Google Scholar 

  172. Masuda A, Takeda J, Okuno T, Okamoto T, Ohkawara B, Ito M et al (2015) Position-specific binding of FUS to nascent RNA regulates mRNA length. Genes Dev 29(10):1045–1057

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Buratti E, Baralle FE (2011) TDP-43: new aspects of autoregulation mechanisms in RNA binding proteins and their connection with human disease. FEBS J 278(19):3530–3538

    Article  CAS  PubMed  Google Scholar 

  174. Ayala YM, De Conti L, Avendano-Vazquez SE, Dhir A, Romano M, D’Ambrogio A et al (2011) TDP-43 regulates its mRNA levels through a negative feedback loop. EMBO J 30(2):277–288

    Article  CAS  PubMed  Google Scholar 

  175. Sugai A, Kato T, Koyama A, Koike Y, Konno T, Ishihara T et al (2019) Non-genetically modified models exhibit TARDBP mRNA increase due to perturbed TDP-43 autoregulation. Neurobiol Dis 130:104534

    Article  CAS  PubMed  Google Scholar 

  176. Maquat LE (2005) Nonsense-mediated mRNA decay in mammals. J Cell Sci 118(Pt 9):1773–1776

    Article  CAS  PubMed  Google Scholar 

  177. Dini Modigliani S, Morlando M, Errichelli L, Sabatelli M, Bozzoni I (2014) An ALS-associated mutation in the FUS 3’-UTR disrupts a microRNA-FUS regulatory circuitry. Nat Commun 5:4335

    Article  CAS  PubMed  Google Scholar 

  178. Budini M, Buratti E (2011) TDP-43 autoregulation: implications for disease. J Mol Neurosci 45(3):473–479

    Article  CAS  PubMed  Google Scholar 

  179. Buratti E, Dork T, Zuccato E, Pagani F, Romano M, Baralle FE (2001) Nuclear factor TDP-43 and SR proteins promote in vitro and in vivo CFTR exon 9 skipping. EMBO J 20(7):1774–1784

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Sephton CF, Good SK, Atkin S, Dewey CM, Mayer P 3rd, Herz J et al (2010) TDP-43 is a developmentally regulated protein essential for early embryonic development. J Biol Chem 285(9):6826–6834

    Article  CAS  PubMed  Google Scholar 

  181. Tollervey JR, Curk T, Rogelj B, Briese M, Cereda M, Kayikci M et al (2011) Characterizing the RNA targets and position-dependent splicing regulation by TDP-43. Nat Neurosci 14(4):452–458

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Xiao S, Sanelli T, Dib S, Sheps D, Findlater J, Bilbao J et al (2011) RNA targets of TDP-43 identified by UV-CLIP are deregulated in ALS. Mol Cell Neurosci 47(3):167–180

    Article  CAS  PubMed  Google Scholar 

  183. Buratti E, Romano M, Baralle FE (2013) TDP-43 high throughput screening analyses in neurodegeneration: advantages and pitfalls. Mol Cell Neurosci 56C:465–474

    Article  CAS  Google Scholar 

  184. Shiga A, Ishihara T, Miyashita A, Kuwabara M, Kato T, Watanabe N et al (2012) Alteration of POLDIP3 splicing associated with loss of function of TDP-43 in tissues affected with ALS. PLoS One 7(8):e43120

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Prudencio M, Jansen-West KR, Lee WC, Gendron TF, Zhang YJ, Xu YF et al (2012) Misregulation of human sortilin splicing leads to the generation of a nonfunctional progranulin receptor. Proc Natl Acad Sci U S A 109(52):21510–21515

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Mohagheghi F, Prudencio M, Stuani C, Cook C, Jansen-West K, Dickson DW et al (2016) TDP-43 functions within a network of hnRNP proteins to inhibit the production of a truncated human SORT1 receptor. Hum Mol Genet 25(3):534–545

    Google Scholar 

  187. De Conti L, Akinyi MV, Mendoza-Maldonado R, Romano M, Baralle M, Buratti E (2015) TDP-43 affects splicing profiles and isoform production of genes involved in the apoptotic and mitotic cellular pathways. Nucleic Acids Res 43(18):8990–9005

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  188. Colombrita C, Onesto E, Buratti E, de la Grange P, Gumina V, Baralle FE et al (2015) From transcriptomic to protein level changes in TDP-43 and FUS loss-of-function cell models. Biochim Biophys Acta 1849(12):1398–1410

    Article  CAS  PubMed  Google Scholar 

  189. Deshaies JE, Shkreta L, Moszczynski AJ, Sidibe H, Semmler S, Fouillen A et al (2018) TDP-43 regulates the alternative splicing of hnRNP A1 to yield an aggregation-prone variant in amyotrophic lateral sclerosis. Brain 141(5):1320–1333

    Article  PubMed  PubMed Central  Google Scholar 

  190. Gu J, Chen F, Iqbal K, Gong CX, Wang X, Liu F (2017) Transactive response DNA-binding protein 43 (TDP-43) regulates alternative splicing of tau exon 10: implications for the pathogenesis of tauopathies. J Biol Chem 292(25):10600–10612

    Google Scholar 

  191. Bose JK, Wang IF, Hung L, Tarn WY, Shen CK (2008) TDP-43 overexpression enhances exon 7 inclusion during the survival of motor neuron pre-mRNA splicing. J Biol Chem 283(43):28852–28859

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Ling JP, Pletnikova O, Troncoso JC, Wong PC (2015) TDP-43 repression of nonconserved cryptic exons is compromised in ALS-FTD. Science 349(6248):650–655

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Tan Q, Yalamanchili HK, Park J, De Maio A, Lu HC, Wan YW et al (2016) Extensive cryptic splicing upon loss of RBM17 and TDP43 in neurodegeneration models. Hum Mol Genet 25(23):5083–5093

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Donde A, Sun M, Ling JP, Braunstein KE, Pang B, Wen X et al (2019) Splicing repression is a major function of TDP-43 in motor neurons. Acta Neuropathol 138(5):813–826

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Gerbino V, Carri MT, Cozzolino M, Achsel T (2013) Mislocalised FUS mutants stall spliceosomal snRNPs in the cytoplasm. Neurobiol Dis 55:120–128

    Article  CAS  PubMed  Google Scholar 

  196. Yu Y, Reed R (2015) FUS functions in coupling transcription to splicing by mediating an interaction between RNAP II and U1 snRNP. Proc Natl Acad Sci U S A 112(28):8608–8613

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Raczynska KD, Ruepp MD, Brzek A, Reber S, Romeo V, Rindlisbacher B et al (2015) FUS/TLS contributes to replication-dependent histone gene expression by interaction with U7 snRNPs and histone-specific transcription factors. Nucleic Acids Res 43(20):9711–9728

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Lagier-Tourenne C, Polymenidou M, Hutt KR, Vu AQ, Baughn M, Huelga SC et al (2012) Divergent roles of ALS-linked proteins FUS/TLS and TDP-43 intersect in processing long pre-mRNAs. Nat Neurosci 15(11):1488–1497

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Ishigaki S, Masuda A, Fujioka Y, Iguchi Y, Katsuno M, Shibata A et al (2012) Position-dependent FUS-RNA interactions regulate alternative splicing events and transcriptions. Sci Rep 2:529

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  200. van Blitterswijk M, Wang ET, Friedman BA, Keagle PJ, Lowe P, Leclerc AL et al (2013) Characterization of FUS mutations in amyotrophic lateral sclerosis using RNA-Seq. PLoS One 8(4):e60788

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Nakaya T, Alexiou P, Maragkakis M, Chang A, Mourelatos Z (2013) FUS regulates genes coding for RNA-binding proteins in neurons by binding to their highly conserved introns. RNA 19(4):498–509

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  203. Orozco D, Edbauer D (2013) FUS-mediated alternative splicing in the nervous system: consequences for ALS and FTLD. J Mol Med (Berl) 91(12):1343–1354

    Article  Google Scholar 

  204. Reber S, Stettler J, Filosa G, Colombo M, Jutzi D, Lenzken SC et al (2016) Minor intron splicing is regulated by FUS and affected by ALS-associated FUS mutants. EMBO J 35(14):1504–1521

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Volonte C, Apolloni S, Parisi C (2015) MicroRNAs: newcomers into the ALS picture. CNS Neurol Disord Drug Targets 14(2):194–207

    Article  CAS  PubMed  Google Scholar 

  206. Goodall EF, Heath PR, Bandmann O, Kirby J, Shaw PJ (2013) Neuronal dark matter: the emerging role of microRNAs in neurodegeneration. Front Cell Neurosci 7:178

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  207. Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, Cooch N et al (2004) The microprocessor complex mediates the genesis of microRNAs. Nature 432(7014):235–240

    Article  CAS  PubMed  Google Scholar 

  208. Casafont I, Bengoechea R, Tapia O, Berciano MT, Lafarga M (2009) TDP-43 localizes in mRNA transcription and processing sites in mammalian neurons. J Struct Biol 167(3):235–241

    Article  CAS  PubMed  Google Scholar 

  209. Di Carlo V, Grossi E, Laneve P, Morlando M, Dini Modigliani S, Ballarino M et al (2013) TDP-43 regulates the microprocessor complex activity during in vitro neuronal differentiation. Mol Neurobiol 48(3):952–963

    Article  PubMed  CAS  Google Scholar 

  210. Buratti E, De Conti L, Stuani C, Romano M, Baralle M, Baralle F (2010) Nuclear factor TDP-43 can affect selected microRNA levels. FEBS J 277(10):2268–2281

    Article  CAS  PubMed  Google Scholar 

  211. King IN, Yartseva V, Salas D, Kumar A, Heidersbach A, Ando DM et al (2014) The RNA-binding protein TDP-43 selectively disrupts microRNA-1/206 incorporation into the RNA-induced silencing complex. J Biol Chem 289(20):14263–14271

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Zhang Z, Almeida S, Lu Y, Nishimura AL, Peng L, Sun D et al (2013) Downregulation of microRNA-9 in iPSC-derived neurons of FTD/ALS patients with TDP-43 mutations. PLoS One 8(10):e76055

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Park YY, Kim SB, Han HD, Sohn BH, Kim JH, Liang J et al (2013) Tat-activating regulatory DNA-binding protein regulates glycolysis in hepatocellular carcinoma by regulating the platelet isoform of phosphofructokinase through microRNA 520. Hepatology 58(1):182–191

    Article  CAS  PubMed  Google Scholar 

  214. Kawahara Y, Mieda-Sato A (2012) TDP-43 promotes microRNA biogenesis as a component of the Drosha and Dicer complexes. Proc Natl Acad Sci U S A 109(9):3347–3352

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Fan Z, Chen X, Chen R (2014) Transcriptome-wide analysis of TDP-43 binding small RNAs identifies miR-NID1 (miR-8485), a novel miRNA that represses NRXN1 expression. Genomics 103(1):76–82

    Article  CAS  PubMed  Google Scholar 

  216. Morlando M, Dini Modigliani S, Torrelli G, Rosa A, Di Carlo V, Caffarelli E et al (2012) FUS stimulates microRNA biogenesis by facilitating co-transcriptional Drosha recruitment. EMBO J 31(24):4502–4510

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Grammatikakis I, Panda AC, Abdelmohsen K, Gorospe M (2014) Long noncoding RNAs(lncRNAs) and the molecular hallmarks of aging. Aging (Albany NY) 6(12):992–1009

    Article  Google Scholar 

  218. Roberts TC, Morris KV, Wood MJ (2014) The role of long non-coding RNAs in neurodevelopment, brain function and neurological disease. Philos Trans R Soc Lond Ser B Biol Sci 369(1652):20130507.

    Google Scholar 

  219. Liu X, Li D, Zhang W, Guo M, Zhan Q (2012) Long non-coding RNA gadd7 interacts with TDP-43 and regulates Cdk6 mRNA decay. EMBO J 31(23):4415–4427

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Wu H, Yin QF, Luo Z, Yao RW, Zheng CC, Zhang J et al (2016) Unusual processing generates SPA LncRNAs that sequester multiple RNA binding proteins. Mol Cell 64(3):534–548

    Article  CAS  PubMed  Google Scholar 

  221. Guo F, Jiao F, Song Z, Li S, Liu B, Yang H et al (2015) Regulation of MALAT1 expression by TDP43 controls the migration and invasion of non-small cell lung cancer cells in vitro. Biochem Biophys Res Commun 465(2):293–298

    Article  CAS  PubMed  Google Scholar 

  222. Balas MM, Porman AM, Hansen KC, Johnson AM (2018) SILAC-MS profiling of reconstituted human chromatin platforms for the study of transcription and RNA regulation. J Proteome Res 17(10):3475–3484

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Militello G, Hosen MR, Ponomareva Y, Gellert P, Weirick T, John D et al (2018) A novel long non-coding RNA myolinc regulates myogenesis through TDP-43 and Filip1. J Mol Cell Biol 10(2):102–117

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Li P, Ruan X, Yang L, Kiesewetter K, Zhao Y, Luo H et al (2015) A liver-enriched long non-coding RNA, lncLSTR, regulates systemic lipid metabolism in mice. Cell Metab 21(3):455–467

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Zhao XS, Tao N, Zhang C, Gong CM, Dong CY (2019) Long noncoding RNA MIAT acts as an oncogene in Wilms’ tumor through regulation of DGCR8. Eur Rev Med Pharmacol Sci 23(23):10257–10263

    PubMed  Google Scholar 

  226. Lourenco GF, Janitz M, Huang Y, Halliday GM (2015) Long noncoding RNAs in TDP-43 and FUS/TLS-related frontotemporal lobar degeneration (FTLD). Neurobiol Dis 82:445–454

    Article  CAS  PubMed  Google Scholar 

  227. Biscarini S, Capauto D, Peruzzi G, Lu L, Colantoni A, Santini T et al (2018) Characterization of the lncRNA transcriptome in mESC-derived motor neurons: implications for FUS-ALS. Stem Cell Res 27:172–179

    Article  CAS  PubMed  Google Scholar 

  228. Lo Piccolo L, Jantrapirom S, Nagai Y, Yamaguchi M (2017) FUS toxicity is rescued by the modulation of lncRNA hsromega expression in Drosophila melanogaster. Sci Rep 7(1):15660

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  229. Gagliardi S, Pandini C, Garofalo M, Bordoni M, Pansarasa O, Cereda C (2018) Long non coding RNAs and ALS: still much to do. Noncoding RNA Res 3(4):226–231

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Cirillo D, Agostini F, Klus P, Marchese D, Rodriguez S, Bolognesi B et al (2013) Neurodegenerative diseases: quantitative predictions of protein-RNA interactions. RNA 19(2):129–140

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Li W, Jin Y, Prazak L, Hammell M, Dubnau J (2012) Transposable elements in TDP-43-mediated neurodegenerative disorders. PLoS One 7(9):e44099

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Dormann D, Haass C (2011) TDP-43 and FUS: a nuclear affair. Trends Neurosci 34(7):339–348

    Article  CAS  PubMed  Google Scholar 

  233. Dormann D, Rodde R, Edbauer D, Bentmann E, Fischer I, Hruscha A et al (2010) ALS-associated fused in sarcoma (FUS) mutations disrupt Transportin-mediated nuclear import. EMBO J 29(16):2841–2857

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Nishimura AL, Zupunski V, Troakes C, Kathe C, Fratta P, Howell M et al (2010) Nuclear import impairment causes cytoplasmic trans-activation response DNA-binding protein accumulation and is associated with frontotemporal lobar degeneration. Brain 133(Pt 6):1763–1771

    Article  PubMed  Google Scholar 

  235. Tradewell ML, Yu Z, Tibshirani M, Boulanger MC, Durham HD, Richard S (2012) Arginine methylation by PRMT1 regulates nuclear-cytoplasmic localization and toxicity of FUS/TLS harbouring ALS-linked mutations. Hum Mol Genet 21(1):136–149

    Article  PubMed  CAS  Google Scholar 

  236. Dormann D, Madl T, Valori CF, Bentmann E, Tahirovic S, Abou-Ajram C et al (2012) Arginine methylation next to the PY-NLS modulates Transportin binding and nuclear import of FUS. EMBO J 31(22):4258–4275

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Scaramuzzino C, Monaghan J, Milioto C, Lanson NA Jr, Maltare A, Aggarwal T et al (2013) Protein arginine methyltransferase 1 and 8 interact with FUS to modify its sub-cellular distribution and toxicity in vitro and in vivo. PLoS One 8(4):e61576

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Yamaguchi A, Kitajo K (2012) The effect of PRMT1-mediated arginine methylation on the subcellular localization, stress granules, and detergent-insoluble aggregates of FUS/TLS. PLoS One 7(11):e49267

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Zhang YJ, Gendron TF, Grima JC, Sasaguri H, Jansen-West K, Xu YF et al (2016) C9ORF72 poly(GA) aggregates sequester and impair HR23 and nucleocytoplasmic transport proteins. Nat Neurosci 19(5):668–677

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Chou CC, Zhang Y, Umoh ME, Vaughan SW, Lorenzini I, Liu F et al (2018) TDP-43 pathology disrupts nuclear pore complexes and nucleocytoplasmic transport in ALS/FTD. Nat Neurosci 21(2):228–239

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Strong MJ, Volkening K, Hammond R, Yang W, Strong W, Leystra-Lantz C et al (2007) TDP43 is a human low molecular weight neurofilament (hNFL) mRNA-binding protein. Mol Cell Neurosci 35(2):320–327

    Article  CAS  PubMed  Google Scholar 

  242. Kim SH, Shanware NP, Bowler MJ, Tibbetts RS (2010) Amyotrophic lateral sclerosis-associated proteins TDP-43 and FUS/TLS function in a common biochemical complex to co-regulate HDAC6 mRNA. J Biol Chem 285(44):34097–34105

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Fiesel FC, Schurr C, Weber SS, Kahle PJ (2011) TDP-43 knockdown impairs neurite outgrowth dependent on its target histone deacetylase 6. Mol Neurodegener 6:64

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Sephton CF, Cenik C, Kucukural A, Dammer EB, Cenik B, Han Y et al (2011) Identification of neuronal RNA targets of TDP-43-containing ribonucleoprotein complexes. J Biol Chem 286(2):1204–1215

    Article  CAS  PubMed  Google Scholar 

  245. Costessi L, Porro F, Iaconcig A, Muro AF (2014) TDP-43 regulates beta-adducin (Add2) transcript stability. RNA Biol 11(10):1280–1290

    Article  PubMed  Google Scholar 

  246. Lee S, Lee TA, Lee E, Kang S, Park A, Kim SW et al (2015) Identification of a subnuclear body involved in sequence-specific cytokine RNA processing. Nat Commun 6:5791

    Article  CAS  PubMed  Google Scholar 

  247. Stallings NR, Puttaparthi K, Dowling KJ, Luther CM, Burns DK, Davis K et al (2013) TDP-43, an ALS linked protein, regulates fat deposition and glucose homeostasis. PLoS One 8(8):e71793

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. McDonald KK, Aulas A, Destroismaisons L, Pickles S, Beleac E, Camu W et al (2011) TAR DNA-binding protein 43 (TDP-43) regulates stress granule dynamics via differential regulation of G3BP and TIA-1. Hum Mol Genet 20(7):1400–1410

    Article  CAS  PubMed  Google Scholar 

  249. Vanderweyde T, Youmans K, Liu-Yesucevitz L, Wolozin B (2013) Role of stress granules and RNA-binding proteins in neurodegeneration: a mini-review. Gerontology 59(6):524–533

    Article  CAS  PubMed  Google Scholar 

  250. Gu J, Wu F, Xu W, Shi J, Hu W, Jin N et al (2017) TDP-43 suppresses tau expression via promoting its mRNA instability. Nucleic Acids Res 45(10):6177–6193

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Udagawa T, Fujioka Y, Tanaka M, Honda D, Yokoi S, Riku Y et al (2015) FUS regulates AMPA receptor function and FTLD/ALS-associated behaviour via GluA1 mRNA stabilization. Nat Commun 6:7098

    Article  CAS  PubMed  Google Scholar 

  252. Swanger SA, Bassell GJ (2011) Making and breaking synapses through local mRNA regulation. Curr Opin Genet Dev 21(4):414–421

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Fallini C, Bassell GJ, Rossoll W (2012) The ALS disease protein TDP-43 is actively transported in motor neuron axons and regulates axon outgrowth. Hum Mol Genet 21(16):3703–3718

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Narayanan RK, Mangelsdorf M, Panwar A, Butler TJ, Noakes PG, Wallace RH (2013) Identification of RNA bound to the TDP-43 ribonucleoprotein complex in the adult mouse brain. Amyotroph Lateral Scler Frontotemporal Degener 14(4):252–260

    Article  CAS  PubMed  Google Scholar 

  255. Fujii R, Okabe S, Urushido T, Inoue K, Yoshimura A, Tachibana T et al (2005) The RNA binding protein TLS is translocated to dendritic spines by mGluR5 activation and regulates spine morphology. Curr Biol 15(6):587–593

    Article  CAS  PubMed  Google Scholar 

  256. Belly A, Moreau-Gachelin F, Sadoul R, Goldberg Y (2005) Delocalization of the multifunctional RNA splicing factor TLS/FUS in hippocampal neurones: exclusion from the nucleus and accumulation in dendritic granules and spine heads. Neurosci Lett 379(3):152–157

    Article  CAS  PubMed  Google Scholar 

  257. Fujii R, Takumi T (2005) TLS facilitates transport of mRNA encoding an actin-stabilizing protein to dendritic spines. J Cell Sci 118(Pt 24):5755–5765

    Article  CAS  PubMed  Google Scholar 

  258. Yoshimura A, Fujii R, Watanabe Y, Okabe S, Fukui K, Takumi T (2006) Myosin-Va facilitates the accumulation of mRNA/protein complex in dendritic spines. Curr Biol 16(23):2345–2351

    Article  CAS  PubMed  Google Scholar 

  259. Muresan V, Ladescu MZ (2016) Shared molecular mechanisms in Alzheimer’s disease and amyotrophic lateral sclerosis: neurofilament-dependent transport of sAPP, FUS, TDP-43 and SOD1, with endoplasmic reticulum-like tubules. Neurodegener Dis 16(1–2):55–61

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  261. Freibaum BD, Chitta RK, High AA, Taylor JP (2010) Global analysis of TDP-43 interacting proteins reveals strong association with RNA splicing and translation machinery. J Proteome Res 9(2):1104–1120

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Godena VK, Romano G, Romano M, Appocher C, Klima R, Buratti E et al (2011) TDP-43 regulates drosophila neuromuscular junctions growth by modulating Futsch/MAP1B levels and synaptic microtubules organization. PLoS One 6(3):e17808

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  263. Coyne AN, Siddegowda BB, Estes PS, Johannesmeyer J, Kovalik T, Daniel SG et al (2014) Futsch/MAP1B mRNA is a translational target of TDP-43 and is neuroprotective in a Drosophila model of amyotrophic lateral sclerosis. J Neurosci 34(48):15962–15974

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  264. Romano M, Feiguin F, Buratti E (2016) TBPH/TDP-43 modulates translation of Drosophila futsch mRNA through an UG-rich sequence within its 5’UTR. Brain Res 1647:50–56

    Google Scholar 

  265. Russo A, Scardigli R, La Regina F, Murray ME, Romano N, Dickson DW et al (2017) Increased cytoplasmic TDP-43 reduces global protein synthesis by interacting with RACK1 on polyribosomes. Hum Mol Genet 26(8):1407–1418

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. Yasuda K, Zhang H, Loiselle D, Haystead T, Macara IG, Mili S (2013) The RNA-binding protein Fus directs translation of localized mRNAs in APC-RNP granules. J Cell Biol 203(5):737–746

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  267. Kamelgarn M, Chen J, Kuang L, Jin H, Kasarskis EJ, Zhu H (2018) ALS mutations of FUS suppress protein translation and disrupt the regulation of nonsense-mediated decay. Proc Natl Acad Sci U S A 115(51):E11904–E11E13

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  268. Budini M, Buratti E, Morselli E, Criollo A (2017) Autophagy and Its impact on neurodegenerative diseases: new roles for TDP-43 and C9orf72. Front Mol Neurosci 10:170

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  269. Gotzl JK, Lang CM, Haass C, Capell A (2016) Impaired protein degradation in FTLD and related disorders. Ageing Res Rev 32:122–139

    Article  PubMed  CAS  Google Scholar 

  270. Renaud L, Picher-Martel V, Codron P, Julien JP (2019) Key role of UBQLN2 in pathogenesis of amyotrophic lateral sclerosis and frontotemporal dementia. Acta Neuropathol Commun 7(1):103

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  271. Rainero I, Rubino E, Michelerio A, D’Agata F, Gentile S, Pinessi L (2017) Recent advances in the molecular genetics of frontotemporal lobar degeneration. Funct Neurol 32(1):7–16

    Article  PubMed  PubMed Central  Google Scholar 

  272. Pensato V, Magri S, Bella ED, Tannorella P, Bersano E, Soraru G et al (2020) Sorting rare als genetic variants by targeted re-sequencing panel in Italian patients: OPTN, VCP, and SQSTM1 variants account for 3% of rare genetic forms. J Clin Med 9(2):412

    Article  CAS  PubMed Central  Google Scholar 

  273. Polymenidou M, Lagier-Tourenne C, Hutt KR, Bennett CF, Cleveland DW, Yeo GW (2012) Misregulated RNA processing in amyotrophic lateral sclerosis. Brain Res 1462:3–15

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Tank EM, Figueroa-Romero C, Hinder LM, Bedi K, Archbold HC, Li X et al (2018) Abnormal RNA stability in amyotrophic lateral sclerosis. Nat Commun 9(1):2845

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  275. Soo KY, Sultana J, King AE, Atkinson R, Warraich ST, Sundaramoorthy V et al (2015) ALS-associated mutant FUS inhibits macroautophagy which is restored by overexpression of Rab1. Cell Death Dis 1:15030

    Article  CAS  Google Scholar 

  276. Lin TW, Chen MT, Lin LT, Huang PI, Lo WL, Yang YP et al (2017) TDP-43/HDAC6 axis promoted tumor progression and regulated nutrient deprivation-induced autophagy in glioblastoma. Oncotarget 8(34):56612–56625

    Google Scholar 

  277. Ling SC, Dastidar SG, Tokunaga S, Ho WY, Lim K, Ilieva H et al (2019) Overriding FUS autoregulation in mice triggers gain-of-toxic dysfunctions in RNA metabolism and autophagy-lysosome axis. Elife 8:e40811

    Google Scholar 

  278. Xia Q, Wang H, Hao Z, Fu C, Hu Q, Gao F et al (2016) TDP-43 loss of function increases TFEB activity and blocks autophagosome-lysosome fusion. EMBO J 35(2):121–142

    Article  CAS  PubMed  Google Scholar 

  279. Wang Y, Liu FT, Wang YX, Guan RY, Chen C, Li DK et al (2018) Autophagic modulation by trehalose reduces accumulation of TDP-43 in a cell model of amyotrophic lateral sclerosis via TFEB activation. Neurotox Res 34(1):109–120

    Article  CAS  PubMed  Google Scholar 

  280. Wang IF, Guo BS, Liu YC, Wu CC, Yang CH, Tsai KJ et al (2012) Autophagy activators rescue and alleviate pathogenesis of a mouse model with proteinopathies of the TAR DNA-binding protein 43. Proc Natl Acad Sci U S A 109(37):15024–15029

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  281. Barmada SJ, Serio A, Arjun A, Bilican B, Daub A, Ando DM et al (2014) Autophagy induction enhances TDP43 turnover and survival in neuronal ALS models. Nat Chem Biol 10(8):677–685

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  282. Zhou F, Dong H, Liu Y, Yan L, Sun C, Hao P et al (2018) Raloxifene, a promising estrogen replacement, limits TDP-25 cell death by enhancing autophagy and suppressing apoptosis. Brain Res Bull 140:281–290

    Article  CAS  PubMed  Google Scholar 

  283. Marrone L, Drexler HCA, Wang J, Tripathi P, Distler T, Heisterkamp P et al (2019) FUS pathology in ALS is linked to alterations in multiple ALS-associated proteins and rescued by drugs stimulating autophagy. Acta Neuropathol 138(1):67–84

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  284. Strohm L, Behrends C (2020) Glia-specific autophagy dysfunction in ALS. Semin Cell Dev Biol 99:172–182

    Google Scholar 

  285. Koza P, Beroun A, Konopka A, Gorkiewicz T, Bijoch L, Torres JC et al (2019) Neuronal TDP-43 depletion affects activity-dependent plasticity. Neurobiol Dis 130:104499

    Article  CAS  PubMed  Google Scholar 

  286. Ling SC (2018) Synaptic paths to neurodegeneration: the emerging role of TDP-43 and FUS in synaptic functions. Neural Plast 2018:8413496

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  287. Heyburn L, Moussa CE (2016) TDP-43 overexpression impairs presynaptic integrity. Neural Regen Res 11(12):1910–1911

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. Gulino R, Forte S, Parenti R, Gulisano M (2015) TDP-43 as a modulator of synaptic plasticity in a mouse model of spinal motoneuron degeneration. CNS Neurol Disord Drug Targets 14(1):55–60

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  290. Chand KK, Lee KM, Lee JD, Qiu H, Willis EF, Lavidis NA et al (2018) Defects in synaptic transmission at the neuromuscular junction precede motor deficits in a TDP-43(Q331K) transgenic mouse model of amyotrophic lateral sclerosis. FASEB J 32(5):2676–2689

    Article  PubMed  Google Scholar 

  291. Romano G, Holodkov N, Klima R, Grilli F, Guarnaccia C, Nizzardo M et al (2018) Downregulation of glutamic acid decarboxylase in Drosophila TDP-43-null brains provokes paralysis by affecting the organization of the neuromuscular synapses. Sci Rep 8(1):1809

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  292. Petel Legare V, Harji ZA, Rampal CJ, Allard-Chamard X, Rodriguez EC, Armstrong GAB (2019) Augmentation of spinal cord glutamatergic synaptic currents in zebrafish primary motoneurons expressing mutant human TARDBP (TDP-43). Sci Rep 9(1):9122

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  293. Sephton CF, Tang AA, Kulkarni A, West J, Brooks M, Stubblefield JJ et al (2014) Activity-dependent FUS dysregulation disrupts synaptic homeostasis. Proc Natl Acad Sci U S A 111(44):E4769–E4778

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  294. Yokoi S, Udagawa T, Fujioka Y, Honda D, Okado H, Watanabe H et al (2017) 3’UTR length-dependent control of SynGAP isoform alpha2 mrna by fus and elav-like proteins promotes dendritic spine maturation and cognitive function. Cell Rep 20(13):3071–3084

    Article  CAS  PubMed  Google Scholar 

  295. Polymenidou M, Cleveland DW (2012) Prion-like spread of protein aggregates in neurodegeneration. J Exp Med 209(5):889–893

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  296. Lee S, Kim HJ (2015) Prion-like mechanism in amyotrophic lateral sclerosis: are protein aggregates the key? Exp Neurobiol 24(1):1–7

    Article  PubMed  Google Scholar 

  297. Pradat PF, Kabashi E, Desnuelle C (2015) Deciphering spreading mechanisms in amyotrophic lateral sclerosis: clinical evidence and potential molecular processes. Curr Opin Neurol 28(5):455–461

    Article  PubMed  Google Scholar 

  298. Ayers JI, Cashman NR (2018) Prion-like mechanisms in amyotrophic lateral sclerosis. Handb Clin Neurol 153:337–354

    Article  PubMed  Google Scholar 

  299. Harrison AF, Shorter J (2017) RNA-binding proteins with prion-like domains in health and disease. Biochem J 474(8):1417–1438

    Article  CAS  PubMed  Google Scholar 

  300. Cushman M, Johnson BS, King OD, Gitler AD, Shorter J (2010) Prion-like disorders: blurring the divide between transmissibility and infectivity. J Cell Sci 123(Pt 8):1191–1201

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  301. Furukawa Y, Kaneko K, Watanabe S, Yamanaka K, Nukina N (2011) A seeding reaction recapitulates intracellular formation of Sarkosyl-insoluble transactivation response element (TAR) DNA-binding protein-43 inclusions. J Biol Chem 286(21):18664–18672

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  302. Nonaka T, Masuda-Suzukake M, Arai T, Hasegawa Y, Akatsu H, Obi T et al (2013) Prion-like properties of pathological TDP-43 aggregates from diseased brains. Cell Rep 4(1):124–134

    Article  CAS  PubMed  Google Scholar 

  303. Feiler MS, Strobel B, Freischmidt A, Helferich AM, Kappel J, Brewer BM et al (2015) TDP-43 is intercellularly transmitted across axon terminals. J Cell Biol 211(4):897–911

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  304. Maggio S, Ceccaroli P, Polidori E, Cioccoloni A, Stocchi V, Guescini M (2019) Signal Exchange through Extracellular Vesicles in Neuromuscular Junction Establishment and Maintenance: From Physiology to Pathology. Int J Mol Sci 20(11):2804

    Article  CAS  PubMed Central  Google Scholar 

  305. Zeineddine R, Pundavela JF, Corcoran L, Stewart EM, Do-Ha D, Bax M et al (2015) SOD1 protein aggregates stimulate macropinocytosis in neurons to facilitate their propagation. Mol Neurodegener 10(1):57

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  306. Nomura T, Watanabe S, Kaneko K, Yamanaka K, Nukina N, Furukawa Y (2014) Intranuclear aggregation of mutant FUS/TLS as a molecular pathomechanism of amyotrophic lateral sclerosis. J Biol Chem 289(2):1192–1202

    Article  CAS  PubMed  Google Scholar 

  307. Lanson NA Jr, Maltare A, King H, Smith R, Kim JH, Taylor JP et al (2011) A Drosophila model of FUS-related neurodegeneration reveals genetic interaction between FUS and TDP-43. Hum Mol Genet 20(13):2510–2523

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  308. Wang JW, Brent JR, Tomlinson A, Shneider NA, McCabe BD (2011) The ALS-associated proteins FUS and TDP-43 function together to affect Drosophila locomotion and life span. J Clin Invest 121(10):4118–4126

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  309. Kabashi E, Bercier V, Lissouba A, Liao M, Brustein E, Rouleau GA et al (2011) FUS and TARDBP but not SOD1 interact in genetic models of amyotrophic lateral sclerosis. PLoS Genet 7(8):e1002214

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  310. Sun Z, Diaz Z, Fang X, Hart MP, Chesi A, Shorter J et al (2011) Molecular determinants and genetic modifiers of aggregation and toxicity for the ALS disease protein FUS/TLS. PLoS Biol 9(4):e1000614

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  311. Suzuki H, Shibagaki Y, Hattori S, Matsuoka M (2015) Nuclear TDP-43 causes neuronal toxicity by escaping from the inhibitory regulation by hnRNPs. Hum Mol Genet 24(6):1513–1527

    Article  CAS  PubMed  Google Scholar 

  312. Moujalled D, James JL, Yang S, Zhang K, Duncan C, Moujalled DM et al (2015) Phosphorylation of hnRNP K by cyclin-dependent kinase 2 controls cytosolic accumulation of TDP-43. Hum Mol Genet 24(6):1655–1669

    Google Scholar 

  313. Appocher C, Mohagheghi F, Cappelli S, Stuani C, Romano M, Feiguin F et al (2017) Major hnRNP proteins act as general TDP-43 functional modifiers both in Drosophila and human neuronal cells. Nucleic Acids Res 45(13):8026–8045

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  314. Hart MP, Gitler AD (2012) ALS-associated ataxin 2 polyQ expansions enhance stress-induced caspase 3 activation and increase TDP-43 pathological modifications. J Neurosci 32(27):9133–9142

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  315. Barmada SJ, Ju S, Arjun A, Batarse A, Archbold HC, Peisach D et al (2015) Amelioration of toxicity in neuronal models of amyotrophic lateral sclerosis by hUPF1. Proc Natl Acad Sci U S A 112(25):7821–7826

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  316. Manzo E, Lorenzini I, Barrameda D, O’Conner AG, Barrows JM, Starr A et al (2019) Glycolysis upregulation is neuroprotective as a compensatory mechanism in ALS. Elife 8:e45114

    Google Scholar 

  317. Donde A, Sun M, Jeong YH, Wen X, Ling J, Lin S et al (2020) Upregulation of ATG7 attenuates motor neuron dysfunction associated with depletion of TARDBP/TDP-43. Autophagy 16(4):672–682

    Google Scholar 

  318. Steyaert J, Scheveneels W, Vanneste J, Van Damme P, Robberecht W, Callaerts P et al (2018) FUS-induced neurotoxicity in Drosophila is prevented by downregulating nucleocytoplasmic transport proteins. Hum Mol Genet 27(23):4103–4116

    CAS  PubMed  PubMed Central  Google Scholar 

  319. Casci I, Krishnamurthy K, Kour S, Tripathy V, Ramesh N, Anderson EN et al (2019) Muscleblind acts as a modifier of FUS toxicity by modulating stress granule dynamics and SMN localization. Nat Commun 10(1):5583

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  320. Lu L, Zheng L, Si Y, Luo W, Dujardin G, Kwan T et al (2014) Hu antigen R (HuR) is a positive regulator of the RNA-binding proteins TDP-43 and FUS/TLS: implications for amyotrophic lateral sclerosis. J Biol Chem 289(46):31792–31804

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  321. Becker LA, Huang B, Bieri G, Ma R, Knowles DA, Jafar-Nejad P et al (2017) Therapeutic reduction of ataxin-2 extends lifespan and reduces pathology in TDP-43 mice. Nature 544(7650):367–371

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  322. Mackenzie IR, Rademakers R, Neumann M (2010) TDP-43 and FUS in amyotrophic lateral sclerosis and frontotemporal dementia. Lancet Neurol 9(10):995–1007

    Article  CAS  PubMed  Google Scholar 

  323. Neumann M, Rademakers R, Roeber S, Baker M, Kretzschmar HA, Mackenzie IR (2009) A new subtype of frontotemporal lobar degeneration with FUS pathology. Brain 132(Pt 11):2922–2931

    Article  PubMed  PubMed Central  Google Scholar 

  324. Baloh RH (2012) How do the RNA-binding proteins TDP-43 and FUS relate to amyotrophic lateral sclerosis and frontotemporal degeneration, and to each other? Curr Opin Neurol 25(6):701–707

    Article  CAS  PubMed  Google Scholar 

  325. Palomo V, Tosat-Bitrian C, Nozal V, Nagaraj S, Martin-Requero A, Martinez A (2019) TDP-43: A Key Therapeutic Target beyond Amyotrophic Lateral Sclerosis. ACS Chem Neurosci 10(3):1183–1196

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by AriSLA (Italy) grant (PathensTDP) and Beneficentia Stiftung (Liechtenstein).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Emanuele Buratti .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2021 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Buratti, E. (2021). Trends in Understanding the Pathological Roles of TDP-43 and FUS Proteins. In: Ghetti, B., Buratti, E., Boeve, B., Rademakers, R. (eds) Frontotemporal Dementias . Advances in Experimental Medicine and Biology, vol 1281. Springer, Cham. https://doi.org/10.1007/978-3-030-51140-1_15

Download citation

Publish with us

Policies and ethics