Skip to main content

Advertisement

Log in

Senataxin mutations elicit motor neuron degeneration phenotypes and yield TDP-43 mislocalization in ALS4 mice and human patients

  • Original Paper
  • Published:
Acta Neuropathologica Aims and scope Submit manuscript

Abstract

Amyotrophic lateral sclerosis type 4 (ALS4) is a rare, early-onset, autosomal dominant form of ALS, characterized by slow disease progression and sparing of respiratory musculature. Dominant, gain-of-function mutations in the senataxin gene (SETX) cause ALS4, but the mechanistic basis for motor neuron toxicity is unknown. SETX is a RNA-binding protein with a highly conserved helicase domain, but does not possess a low-complexity domain, making it unique among ALS-linked disease proteins. We derived ALS4 mouse models by expressing two different senataxin gene mutations (R2136H and L389S) via transgenesis and knock-in gene targeting. Both approaches yielded SETX mutant mice that develop neuromuscular phenotypes and motor neuron degeneration. Neuropathological characterization of SETX mice revealed nuclear clearing of TDP-43, accompanied by TDP-43 cytosolic mislocalization, consistent with the hallmark pathology observed in human ALS patients. Postmortem material from ALS4 patients exhibited TDP-43 mislocalization in spinal cord motor neurons, and motor neurons from SETX ALS4 mice displayed enhanced stress granule formation. Immunostaining analysis for nucleocytoplasmic transport proteins Ran and RanGAP1 uncovered nuclear membrane abnormalities in the motor neurons of SETX ALS4 mice, and nuclear import was delayed in SETX ALS4 cortical neurons, indicative of impaired nucleocytoplasmic trafficking. SETX ALS4 mice thus recapitulated ALS disease phenotypes in association with TDP-43 mislocalization and provided insight into the basis for TDP-43 histopathology, linking SETX dysfunction to common pathways of ALS motor neuron degeneration.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Al-Chalabi A, Fang F, Hanby MF, Leigh PN, Shaw CE, Ye W, Rijsdijk F (2010) An estimate of amyotrophic lateral sclerosis heritability using twin data. J Neurol Neurosurg Psychiatry 81:1324–1326. https://doi.org/10.1136/jnnp.2010.207464

    Article  PubMed  CAS  Google Scholar 

  2. Al-Sarraj S, King A, Troakes C, Smith B, Maekawa S, Bodi I, Rogelj B, Al-Chalabi A, Hortobagyi T, Shaw CE (2011) p62 positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the cerebellum and hippocampus define the pathology of C9orf72-linked FTLD and MND/ALS. Acta Neuropathol 122:691–702. https://doi.org/10.1007/s00401-011-0911-2

    Article  PubMed  CAS  Google Scholar 

  3. Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, Mori H, Mann D, Tsuchiya K, Yoshida M, Hashizume Y 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 351:602–611

    Article  PubMed  CAS  Google Scholar 

  4. Avemaria F, Lunetta C, Tarlarini C, Mosca L, Maestri E, Marocchi A, Melazzini M, Penco S, Corbo M (2011) Mutation in the senataxin gene found in a patient affected by familial ALS with juvenile onset and slow progression. Amyotroph Lateral Scler 12:228–230. https://doi.org/10.3109/17482968.2011.566930

    Article  PubMed  CAS  Google Scholar 

  5. Ayala YM, De Conti L, Avendano-Vazquez SE, Dhir A, Romano M, D’Ambrogio A, Tollervey J, Ule J, Baralle M, Buratti E et al (2011) TDP-43 regulates its mRNA levels through a negative feedback loop. EMBO J 30:277–288. https://doi.org/10.1038/emboj.2010.310

    Article  PubMed  CAS  Google Scholar 

  6. Ayala YM, Zago P, D’Ambrogio A, Xu YF, Petrucelli L, Buratti E, Baralle FE (2008) Structural determinants of the cellular localization and shuttling of TDP-43. J Cell Sci 121:3778–3785. https://doi.org/10.1242/jcs.038950

    Article  PubMed  CAS  Google Scholar 

  7. Beck M, Schmidt A, Malmstroem J, Claassen M, Ori A, Szymborska A, Herzog F, Rinner O, Ellenberg J, Aebersold R (2011) The quantitative proteome of a human cell line. Mol Syst Biol 7:549. https://doi.org/10.1038/msb.2011.82

    Article  PubMed  PubMed Central  Google Scholar 

  8. Becker LA, Huang B, Bieri G, Ma R, Knowles DA, Jafar-Nejad P, Messing J, Kim HJ, Soriano A, Auburger G et al (2017) Therapeutic reduction of ataxin-2 extends lifespan and reduces pathology in TDP-43 mice. Nature 544:367–371. https://doi.org/10.1038/nature22038

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Bennett CL, Chen Y, Vignali M, Lo RS, Mason AG, Unal A, Huq Saifee NP, Fields S, La Spada AR (2013) Protein interaction analysis of senataxin and the ALS4 L389S mutant yields insights into senataxin post-translational modification and uncovers mutant-specific binding with a brain cytoplasmic RNA-encoded peptide. PLoS One 8:e78837. https://doi.org/10.1371/journal.pone.0078837

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Blokhuis AM, Koppers M, Groen EJ, van den Heuvel DM, Dini Modigliani S, Anink JJ, Fumoto K, van Diggelen F, Snelting A, Sodaar P et al (2016) Comparative interactomics analysis of different ALS-associated proteins identifies converging molecular pathways. Acta Neuropathol 132:175–196. https://doi.org/10.1007/s00401-016-1575-8

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Bozzoni V, Pansarasa O, Diamanti L, Nosari G, Cereda C, Ceroni M (2016) Amyotrophic lateral sclerosis and environmental factors. Funct Neurol 31:7–19

    PubMed  PubMed Central  Google Scholar 

  12. 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 20:1774–1784. https://doi.org/10.1093/emboj/20.7.1774

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Chance PF, Rabin BA, Ryan SG, Ding Y, Scavina M, Crain B, Griffin JW, Cornblath DR (1998) Linkage of the gene for an autosomal dominant form of juvenile amyotrophic lateral sclerosis to chromosome 9q34. Am J Hum Genet 62:633–640

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Chen YZ, Bennett CL, Huynh HM, Blair IP, Puls I, Irobi J, Dierick I, Abel A, Kennerson ML, Rabin BA et al (2004) DNA/RNA helicase gene mutations in a form of juvenile amyotrophic lateral sclerosis (ALS4). Am J Hum Genet 74:1128–1135. https://doi.org/10.1086/421054

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Chen YZ, Hashemi SH, Anderson SK, Huang Y, Moreira MC, Lynch DR, Glass IA, Chance PF, Bennett CL (2006) Senataxin, the yeast Sen1p orthologue: characterization of a unique protein in which recessive mutations cause ataxia and dominant mutations cause motor neuron disease. Neurobiol Dis 23:97–108. https://doi.org/10.1016/j.nbd.2006.02.007

    Article  PubMed  CAS  Google Scholar 

  16. Corcia P, Couratier P, Blasco H, Andres CR, Beltran S, Meininger V, Vourc’h P (2017) Genetics of amyotrophic lateral sclerosis. Rev Neurol (Paris). https://doi.org/10.1016/j.neurol.2017.03.030

    Article  Google Scholar 

  17. Custer SK, Garden GA, Gill N, Rueb U, Libby RT, Schultz C, Guyenet SJ, Deller T, Westrum LE, Sopher BL et al (2006) Bergmann glia expression of polyglutamine-expanded ataxin-7 produces neurodegeneration by impairing glutamate transport. Nat Neurosci 9:1302–1311

    Article  PubMed  CAS  Google Scholar 

  18. Dastidar SG, Landrieu PM, D’Mello SR (2011) FoxG1 promotes the survival of postmitotic neurons. J Neurosci 31:402–413. https://doi.org/10.1523/JNEUROSCI.2897-10.2011

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. de Planell-Saguer M, Schroeder DG, Rodicio MC, Cox GA, Mourelatos Z (2009) Biochemical and genetic evidence for a role of IGHMBP2 in the translational machinery. Hum Mol Genet 18:2115–2126. https://doi.org/10.1093/hmg/ddp134

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. DeMarini DJ, Papa FR, Swaminathan S, Ursic D, Rasmussen TP, Culbertson MR, Hochstrasser M (1995) The yeast SEN3 gene encodes a regulatory subunit of the 26S proteasome complex required for ubiquitin-dependent protein degradation in vivo. Mol Cell Biol 15:6311–6321

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Elden AC, Kim HJ, Hart MP, Chen-Plotkin AS, Johnson BS, Fang X, Armakola M, Geser F, Greene R, Lu MM et al (2010) Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 466:1069–1075. https://doi.org/10.1038/nature09320

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. 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 9:1104–1120. https://doi.org/10.1021/pr901076y

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Freibaum BD, Lu Y, Lopez-Gonzalez R, Kim NC, Almeida S, Lee KH, Badders N, Valentine M, Miller BL, Wong PC et al (2015) GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature 525:129–133. https://doi.org/10.1038/nature14974

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Gabaldon T, Koonin EV (2013) Functional and evolutionary implications of gene orthology. Nat Rev Genet 14:360–366. https://doi.org/10.1038/nrg3456

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Grohmann K, Schuelke M, Diers A, Hoffmann K, Lucke B, Adams C, Bertini E, Leonhardt-Horti H, Muntoni F, Ouvrier R et al (2001) Mutations in the gene encoding immunoglobulin mu-binding protein 2 cause spinal muscular atrophy with respiratory distress type 1. Nat Genet 29:75–77

    Article  PubMed  CAS  Google Scholar 

  26. Grunseich C, Wang IX, Watts JA, Burdick JT, Guber RD, Zhu Z, Bruzel A, Lanman T, Chen K, Schindler AB et al (2018) Senataxin mutation reveals how R-loops promote transcription by blocking DNA methylation at gene promoters. Mol Cell 69(426–437):e427. https://doi.org/10.1016/j.molcel.2017.12.030

    Article  CAS  Google Scholar 

  27. Gurney ME, Pu H, Chiu AY, Dal Canto MC, Polchow CY, Alexander DD, Caliendo J, Hentati A, Kwon YW, Deng HX et al (1994) Motor neuron degeneration in mice that express a human Cu, Zn superoxide dismutase mutation [see comments] [published erratum appears in Science 1995 Jul 14;269(5221):149]. Science 264:1772–1775

    Article  PubMed  CAS  Google Scholar 

  28. Guyenet SJ, Furrer SA, Damian VM, Baughan TD, La Spada AR, Garden GA (2010) A simple composite phenotype scoring system for evaluating mouse models of cerebellar ataxia. J Vis Exp. https://doi.org/10.3791/1787

    Article  PubMed  PubMed Central  Google Scholar 

  29. Kabashi E, Valdmanis PN, Dion P, Spiegelman D, McConkey BJ, Vande Velde C, Bouchard JP, Lacomblez L, Pochigaeva K, Salachas F et al (2008) TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat Genet 40:572–574

    Article  PubMed  CAS  Google Scholar 

  30. Kato M, Han TW, Xie S, Shi K, Du X, Wu LC, Mirzaei H, Goldsmith EJ, Longgood J, Pei J et al (2012) Cell-free formation of RNA granules: low complexity sequence domains form dynamic fibers within hydrogels. Cell 149:753–767. https://doi.org/10.1016/j.cell.2012.04.017

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  31. Kiernan MC, Vucic S, Cheah BC, Turner MR, Eisen A, Hardiman O, Burrell JR, Zoing MC (2011) Amyotrophic lateral sclerosis. Lancet 377:942–955. https://doi.org/10.1016/S0140-6736(10)61156-7

    Article  PubMed  CAS  Google Scholar 

  32. Kuehner JN, Pearson EL, Moore C (2011) Unravelling the means to an end: RNA polymerase II transcription termination. Nat Rev Mol Cell Biol 12:283–294

    Article  PubMed  CAS  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Lavin MF, Shiloh Y (1997) The genetic defect in ataxia-telangiectasia. Annu Rev Immunol 15:177–202

    Article  PubMed  CAS  Google Scholar 

  35. Le NT, Chang L, Kovlyagina I, Georgiou P, Safren N, Braunstein KE, Kvarta MD, Van Dyke AM, LeGates TA, Philips T et al (2016) Motor neuron disease, TDP-43 pathology, and memory deficits in mice expressing ALS-FTD-linked UBQLN2 mutations. Proc Natl Acad Sci USA 113:E7580–E7589. https://doi.org/10.1073/pnas.1608432113

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  36. Lee KH, Zhang P, Kim HJ, Mitrea DM, Sarkar M, Freibaum BD, Cika J, Coughlin M, Messing J, Molliex A et al (2016) c9orf72 dipeptide repeats impair the assembly, dynamics, and function of membrane-less organelles. Cell 167(774–788):e717. https://doi.org/10.1016/j.cell.2016.10.002

    Article  CAS  Google Scholar 

  37. Ling JP, Pletnikova O, Troncoso JC, Wong PC (2015) TDP-43 repression of nonconserved cryptic exons is compromised in ALS-FTD. Science 349:650–655. https://doi.org/10.1126/science.aab0983

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25:402–408

    Article  PubMed  CAS  Google Scholar 

  39. Lynch DR, Braastad CD, Nagan N (2007) Ovarian failure in ataxia with oculomotor apraxia type 2. Am J Med Genet A 143:1775–1777

    Article  CAS  Google Scholar 

  40. Malik B, Nirmalananthan N, Bilsland LG, La Spada AR, Hanna MG, Schiavo G, Gallo JM, Greensmith L (2011) Absence of disturbed axonal transport in spinal and bulbar muscular atrophy. Hum Mol Genet 20:1776–1786

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. McGoldrick P, Joyce PI, Fisher EM, Greensmith L (2013) Rodent models of amyotrophic lateral sclerosis. Biochim Biophys Acta 1832:1421–1436. https://doi.org/10.1016/j.bbadis.2013.03.012

    Article  PubMed  CAS  Google Scholar 

  42. Medghalchi SM, Frischmeyer PA, Mendell JT, Kelly AG, Lawler AM, Dietz HC (2001) Rent1, a trans-effector of nonsense-mediated mRNA decay, is essential for mammalian embryonic viability. Hum Mol Genet 10:99–105

    Article  PubMed  CAS  Google Scholar 

  43. Molliex A, Temirov J, Lee J, Coughlin M, Kanagaraj AP, Kim HJ, Mittag T, Taylor JP (2015) Phase separation by low complexity domains promotes stress granule assembly and drives pathological fibrillization. Cell 163:123–133. https://doi.org/10.1016/j.cell.2015.09.015

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Moreira MC, Klur S, Watanabe M, Nemeth AH, Le Ber I, Moniz JC, Tranchant C, Aubourg P, Tazir M, Schols L et al (2004) Senataxin, the ortholog of a yeast RNA helicase, is mutant in ataxia-ocular apraxia 2. Nat Genet 36:225–227. https://doi.org/10.1038/ng1303

    Article  PubMed  CAS  Google Scholar 

  45. Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM et al (2006) Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314:130–133

    Article  PubMed  CAS  Google Scholar 

  46. Padmanabhan K, Robles MS, Westerling T, Weitz CJ (2012) Feedback regulation of transcriptional termination by the mammalian circadian clock PERIOD complex. Science 337:599–602. https://doi.org/10.1126/science.1221592

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Rabin BA, Griffin JW, Crain BJ, Scavina M, Chance PF, Cornblath DR (1999) Autosomal dominant juvenile amyotrophic lateral sclerosis. Brain 122:1539–1550

    Article  PubMed  Google Scholar 

  49. Richard P, Feng S, Manley JL (2013) A SUMO-dependent interaction between Senataxin and the exosome, disrupted in the neurodegenerative disease AOA2, targets the exosome to sites of transcription-induced DNA damage. Genes Dev 27:2227–2232. https://doi.org/10.1101/gad.224923.113

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Richard P, Manley JL (2009) Transcription termination by nuclear RNA polymerases. Genes Dev 23:1247–1269

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Rudnik-Schoneborn S, Arning L, Epplen JT, Zerres K (2012) SETX gene mutation in a family diagnosed autosomal dominant proximal spinal muscular atrophy. Neuromuscul Disord 22:258–262. https://doi.org/10.1016/j.nmd.2011.09.006

    Article  PubMed  Google Scholar 

  52. Skourti-Stathaki K, Proudfoot NJ, Gromak N (2011) Human senataxin resolves RNA/DNA hybrids formed at transcriptional pause sites to promote Xrn2-dependent termination. Mol Cell 42:794–805

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Sopher BL, Thomas PS Jr, LaFevre-Bernt MA, Holm IE, Wilke SA, Ware CB, Jin LW, Libby RT, Ellerby LM, La Spada AR (2004) Androgen receptor YAC transgenic mice recapitulate SBMA motor neuronopathy and implicate VEGF164 in the motor neuron degeneration. Neuron 41:687–699

    Article  PubMed  CAS  Google Scholar 

  54. Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, Ackerley S, Durnall JC, Williams KL, Buratti E et al (2008) TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 319:1668–1672

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  55. Suraweera A, Lim Y, Woods R, Birrell GW, Nasim T, Becherel OJ, Lavin MF (2009) Functional role for senataxin, defective in ataxia oculomotor apraxia type 2, in transcriptional regulation. Hum Mol Genet 18:3384–3396. https://doi.org/10.1093/hmg/ddp278

    Article  PubMed  CAS  Google Scholar 

  56. Swarup V, Julien JP (2011) ALS pathogenesis: recent insights from genetics and mouse models. Prog Neuropsychopharmacol Biol Psychiatry 35:363–369

    Article  PubMed  CAS  Google Scholar 

  57. Taylor JP, Brown RH Jr, Cleveland DW (2016) Decoding ALS: from genes to mechanism. Nature 539:197–206. https://doi.org/10.1038/nature20413

    Article  PubMed  PubMed Central  Google Scholar 

  58. Tripolszki K, Torok D, Goudenege D, Farkas K, Sulak A, Torok N, Engelhardt JI, Klivenyi P, Procaccio V, Nagy N et al (2017) High-throughput sequencing revealed a novel SETX mutation in a Hungarian patient with amyotrophic lateral sclerosis. Brain Behav 7:e00669. https://doi.org/10.1002/brb3.669

    Article  PubMed  PubMed Central  Google Scholar 

  59. Van Den Bosch L (2011) Genetic rodent models of amyotrophic lateral sclerosis. J Biomed Biotechnol 2011:348765. https://doi.org/10.1155/2011/348765

    Article  CAS  Google Scholar 

  60. Wang IF, Chang HY, Hou SC, Liou GG, Way TD, James Shen CK (2012) The self-interaction of native TDP-43 C terminus inhibits its degradation and contributes to early proteinopathies. Nat Commun 3:766. https://doi.org/10.1038/ncomms1766

    Article  PubMed  CAS  Google Scholar 

  61. Wang J, Xu G, Slunt HH, Gonzales V, Coonfield M, Fromholt D, Copeland NG, Jenkins NA, Borchelt DR (2005) Coincident thresholds of mutant protein for paralytic disease and protein aggregation caused by restrictively expressed superoxide dismutase cDNA. Neurobiol Dis 20:943–952

    Article  PubMed  CAS  Google Scholar 

  62. Weng Y, Czaplinski K, Peltz SW (1996) Genetic and biochemical characterization of mutations in the ATPase and helicase regions of the Upf1 protein. Mol Cell Biol 16:5477–5490

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Wingo TS, Cutler DJ, Yarab N, Kelly CM, Glass JD (2011) The heritability of amyotrophic lateral sclerosis in a clinically ascertained United States research registry. PLoS One 6:e27985. https://doi.org/10.1371/journal.pone.0027985

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Woerner AC, Frottin F, Hornburg D, Feng LR, Meissner F, Patra M, Tatzelt J, Mann M, Winklhofer KF, Hartl FU et al (2016) Cytoplasmic protein aggregates interfere with nucleocytoplasmic transport of protein and RNA. Science 351:173–176. https://doi.org/10.1126/science.aad2033

    Article  PubMed  CAS  Google Scholar 

  65. Yeo AJ, Becherel OJ, Luff JE, Cullen JK, Wongsurawat T, Jenjaroenpoon P, Kuznetsov VA, McKinnon PJ, Lavin MF (2014) R-loops in proliferating cells but not in the brain: implications for AOA2 and other autosomal recessive ataxias. PLoS One 9:e90219. https://doi.org/10.1371/journal.pone.0090219

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Young JE, Garden GA, Martinez RA, Tanaka F, Sandoval CM, Smith AC, Sopher BL, Lin A, Fischbeck KH, Ellerby LM et al (2009) Polyglutamine-expanded androgen receptor truncation fragments activate a Bax-dependent apoptotic cascade mediated by DP5/Hrk. J Neurosci 29:1987–1997

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Yuce O, West SC (2013) Senataxin, defective in the neurodegenerative disorder ataxia with oculomotor apraxia 2, lies at the interface of transcription and the DNA damage response. Mol Cell Biol 33:406–417

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Zhang K, Donnelly CJ, Haeusler AR, Grima JC, Machamer JB, Steinwald P, Daley EL, Miller SJ, Cunningham KM, Vidensky S et al (2015) The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 525:56–61. https://doi.org/10.1038/nature14973

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to R. Palmiter (University of Washington) for providing the ES cell targeting vector (4317G9), B. Crain and L. Ostrow (Johns Hopkins Medical College) for providing ALS4 patient tissue samples and A. Wörner (Max-Planck Institute of Biochemistry) for providing the NLS-NES-eGFP shuttling reporter construct. We wish to thank J.F. Ervin for technical assistance. This work was supported by funding from the US National Institutes of Health (R01 GM094384 [C.L.B.]), the Robert Packard Center for ALS Research at Johns Hopkins School of Medicine [A.R.L.S.] and the Motor Neurone Disease Association (MNDA) [B.M. & L.G.].

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Albert R. La Spada.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bennett, C.L., Dastidar, S.G., Ling, SC. et al. Senataxin mutations elicit motor neuron degeneration phenotypes and yield TDP-43 mislocalization in ALS4 mice and human patients. Acta Neuropathol 136, 425–443 (2018). https://doi.org/10.1007/s00401-018-1852-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00401-018-1852-9

Keywords

Navigation