Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

The expanding biology of the C9orf72 nucleotide repeat expansion in neurodegenerative disease

Key Points

  • Since its discovery as the most common genetic abnormality in familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), researchers have sought to understand the pathophysiology that results from the chromosome 9 open reading frame 72 (C9orf72) hexanucleotide expansion. Several pathways have been proposed to contribute.

  • The nucleotide expansion may disrupt transcription, promoter activation and epigenetic regulation and may therefore drive a loss of C9orf72 function.

  • A body of evidence suggests that cellular dysfunction results from toxicity that occurs downstream of the nucleotide repeat.

  • There are several types of pathology associated with the nucleotide expansion that may drive this toxicity, including RNA foci, dipeptide repeat aggregates and nuclear pathology.

  • Nucleocytoplasmic transport defects are a key contributor to C9orf72-driven pathophysiology. Studies of human brains, induced pluripotent stem cell-derived neurons, flies and yeast models, converge to suggest that nuclear export and import are defective in cells carrying the mutation.

  • Several therapeutic strategies are being explored. For example, antisense oligonucleotides and small molecules may mitigate toxicity caused by the C9orf72 nucleotide repeat expansion.

Abstract

A nucleotide repeat expansion (NRE) within the chromosome 9 open reading frame 72 (C9orf72) gene was the first of this type of mutation to be linked to multiple neurological conditions, including amyotrophic lateral sclerosis and frontotemporal dementia. The pathogenic mechanisms through which the C9orf72 NRE contributes to these disorders include loss of C9orf72 function and gain-of-function mechanisms of C9orf72 driven by toxic RNA and protein species encoded by the NRE. These mechanisms have been linked to several cellular defects — including nucleocytoplasmic trafficking deficits and nuclear stress — that have been observed in both patients and animal models.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The C9orf72 nucleotide repeat expansion.
Figure 2: Cellular processes impaired by the C9orf72 nucleotide repeat expansion.
Figure 3: Identification of a diverse C9orf72•protein interactome.

Similar content being viewed by others

References

  1. Renton, A. E. et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72, 257–268 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. DeJesus-Hernandez, M. et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72, 245–256 (2011). References 1 and 2 are the seminal publications that defined the C9orf72 mutation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Majounie, E. et al. Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. Lancet Neurol. 11, 323–330 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhang, D., Iyer, L. M., He, F. & Aravind, L. Discovery of novel DENN proteins: implications for the evolution of eukaryotic intracellular membrane structures and human disease. Front. Genet. 3, 283 (2012).

    PubMed  PubMed Central  Google Scholar 

  5. Levine, T. P., Daniels, R. D., Gatta, A. T., Wong, L. H. & Hayes, M. J. The product of C9orf72, a gene strongly implicated in neurodegeneration, is structurally related to DENN Rab-GEFs. Bioinformatics 29, 499–503 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Allaire, P. D. et al. The Connecdenn DENN domain: a GEF for Rab35 mediating cargo-specific exit from early endosomes. Mol. Cell 37, 370–382 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Marat, A. L., Dokainish, H. & McPherson, P. S. DENN domain proteins: regulators of Rab GTPases. J. Biol. Chem. 286, 13791–13800 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ciura, S. et al. Loss of function of C9orf72 causes motor deficits in a zebrafish model of amyotrophic lateral sclerosis. Ann. Neurol. 74, 180–187 (2013).

    CAS  PubMed  Google Scholar 

  9. Suzuki, N. et al. The mouse C9ORF72 ortholog is enriched in neurons known to degenerate in ALS and FTD. Nat. Neurosci. 16, 1725–1727 (2013). This report shows that the expression of endogenous C9orf72 is elevated in neurons.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kim, M. S. et al. A draft map of the human proteome. Nature 509, 575–581 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Xiao, S. et al. Isoform-specific antibodies reveal distinct subcellular localizations of C9orf72 in amyotrophic lateral sclerosis. Ann. Neurol. 78, 568–583 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Farg, M. A. et al. C9ORF72, implicated in amytrophic lateral sclerosis and frontotemporal dementia, regulates endosomal trafficking. Hum. Mol. Genet. 23, 3579–3595 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Donnelly, C. J. et al. RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Neuron 80, 415–428 (2013). A study showing that human C9orf72 iPSC-derived motor neurons can model disease and that ASOs can mitigate neuronal injury.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Almeida, S. et al. Modeling key pathological features of frontotemporal dementia with C9ORF72 repeat expansion in iPSC-derived human neurons. Acta Neuropathol. 126, 385–399 (2013). This report shows that iPSC-derived cortical neurons exhibit RNA foci and are sensitive to autophagy-induced stress.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gijselinck, I. et al. A C9orf72 promoter repeat expansion in a Flanders-Belgian cohort with disorders of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis spectrum: a gene identification study. Lancet Neurol. 11, 54–65 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. Belzil, V. V. et al. Reduced C9orf72 gene expression in c9FTD/ALS is caused by histone trimethylation, an epigenetic event detectable in blood. Acta Neuropathol. 126, 895–905 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Waite, A. J. et al. Reduced C9orf72 protein levels in frontal cortex of amyotrophic lateral sclerosis and frontotemporal degeneration brain with the C9ORF72 hexanucleotide repeat expansion. Neurobiol. Aging 35, 1779.e5–1779.e13 (2014).

    Article  CAS  Google Scholar 

  18. Haeusler, A. R. et al. C9orf72 nucleotide repeat structures initiate molecular cascades of disease. Nature 507, 195–200 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Xi, Z. et al. Hypermethylation of the CpG island near the G4C2 repeat in ALS with a C9orf72 expansion. Am. J. Hum. Genet. 92, 981–989 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sareen, D. et al. Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion. Sci. Transl Med. 5, 208ra149 (2013). This paper describes altered transcription of C9orf72 in iPSC-derived motor neurons expressing the expanded allele and the correction of these defects with ASOs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. van Blitterswijk, M. et al. Novel clinical associations with specific C9ORF72 transcripts in patients with repeat expansions in C9ORF72. Acta Neuropathol. 130, 863–876 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. O'Rourke, J. G. et al. C9orf72 BAC transgenic mice display typical pathologic features of ALS/FTD. Neuron 88, 892–901 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Grabczyk, E. & Usdin, K. The GAA*TTC triplet repeat expanded in Friedreich's ataxia impedes transcription elongation by T7 RNA polymerase in a length and supercoil dependent manner. Nucleic Acids Res. 28, 2815–2822 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ohshima, K., Montermini, L., Wells, R. D. & Pandolfo, M. Inhibitory effects of expanded GAA. TTC triplet repeats from intron I of the Friedreich ataxia gene on transcription and replication in vivo. J. Biol. Chem. 273, 14588–14595 (1998).

    Article  CAS  PubMed  Google Scholar 

  25. Parsons, M. A., Sinden, R. R. & Izban, M. G. Transcriptional properties of RNA polymerase II within triplet repeat-containing DNA from the human myotonic dystrophy and fragile X loci. J. Biol. Chem. 273, 26998–27008 (1998).

    Article  CAS  PubMed  Google Scholar 

  26. Tornaletti, S., Park-Snyder, S. & Hanawalt, P. C. G4-forming sequences in the non-transcribed DNA strand pose blocks to T7 RNA polymerase and mammalian RNA polymerase II. J. Biol. Chem. 283, 12756–12762 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Li, Y. et al. Expanded GAA repeats impede transcription elongation through the FXN gene and induce transcriptional silencing that is restricted to the FXN locus. Hum. Mol. Genet. 24, 6932–6943 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Liu, C. R. et al. Spt4 is selectively required for transcription of extended trinucleotide repeats. Cell 148, 690–701 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Batra, R., Charizanis, K. & Swanson, M. S. Partners in crime: bidirectional transcription in unstable microsatellite disease. Hum. Mol. Genet. 19, R77–R82 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zu, T. et al. RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia. Proc. Natl Acad. Sci. USA 110, E4968–E4977 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mori, K. et al. Bidirectional transcripts of the expanded C9orf72 hexanucleotide repeat are translated into aggregating dipeptide repeat proteins. Acta Neuropathol. 126, 881–893 (2013). References 30 and 31 show that repeat-associated aggregates generated from sense and antisense C9orf72 transcripts are detected in the human brain.

    Article  CAS  PubMed  Google Scholar 

  32. Core, L. J., Waterfall, J. J. & Lis, J. T. Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters. Science 322, 1845–1848 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Seila, A. C. et al. Divergent transcription from active promoters. Science 322, 1849–1851 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bernstein, E. & Allis, C. D. RNA meets chromatin. Genes Dev. 19, 1635–1655 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Pelechano, V. & Steinmetz, L. M. Gene regulation by antisense transcription. Nat. Rev. Genet. 14, 880–893 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Chung, D. W., Rudnicki, D. D., Yu, L. & Margolis, R. L. A natural antisense transcript at the Huntington's disease repeat locus regulates HTT expression. Hum. Mol. Genet. 20, 3467–3477 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Sopher, B. L. et al. CTCF regulates ataxin-7 expression through promotion of a convergently transcribed, antisense noncoding RNA. Neuron 70, 1071–1084 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gascon, E. & Gao, F. B. The emerging roles of microRNAs in the pathogenesis of frontotemporal dementia-amyotrophic lateral sclerosis (FTD-ALS) spectrum disorders. J. Neurogenet. 28, 30–40 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Freischmidt, A. et al. Serum microRNAs in patients with genetic amyotrophic lateral sclerosis and pre-manifest mutation carriers. Brain 137, 2938–2950 (2014).

    Article  PubMed  Google Scholar 

  40. Therrien, M., Rouleau, G. A., Dion, P. A. & Parker, J. A. Deletion of C9ORF72 results in motor neuron degeneration and stress sensitivity in C. elegans. PLoS ONE 8, e83450 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wen, X. et al. Antisense proline-arginine RAN dipeptides linked to C9ORF72-ALS/FTD form toxic nuclear aggregates that initiate in vitro and in vivo neuronal death. Neuron 84, 1213–1225 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lagier-Tourenne, C. et al. Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration. Proc. Natl Acad. Sci. USA 110, E4530–E4539 (2013). This study reveals the presence of (C 4 G 2 ) RNA foci in ALS and FTD and shows that antisense knockdown of C9orf72 in the adult rodent does not induce neurodegeneration.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Koppers, M. et al. C9orf72 ablation in mice does not cause motor neuron degeneration or motor deficits. Ann. Neurol. 78, 426–438 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Harms, M. B. et al. Lack of C9ORF72 coding mutations supports a gain of function for repeat expansions in amyotrophic lateral sclerosis. Neurobiol. Aging 34, 2234.e13–2234.e19 (2013).

    Article  CAS  Google Scholar 

  45. Fratta, P. et al. Homozygosity for the C9orf72 GGGGCC repeat expansion in frontotemporal dementia. Acta Neuropathol. 126, 401–409 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Peters, O. M. et al. Human C9ORF72 hexanucleotide expansion reproduces RNA foci and dipeptide repeat proteins but not neurodegeneration in BAC transgenic mice. Neuron 88, 902–909 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Mizielinska, S. et al. C9orf72 repeat expansions cause neurodegeneration in Drosophila through arginine-rich proteins. Science 345, 1192–1194 (2014). This paper reports that selected RAN translation peptides, when overexpressed in fly, can be neurotoxic.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gendron, T. F. et al. Antisense transcripts of the expanded C9ORF72 hexanucleotide repeat form nuclear RNA foci and undergo repeat-associated non-ATG translation in c9FTD/ALS. Acta Neuropathol. 126, 829–844 (2013). A study showing the presence of RNA foci and RAN translation aggregates in a C9orf72 human brain.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Cooper-Knock, J. et al. Antisense RNA foci in the motor neurons of C9ORF72-ALS patients are associated with TDP-43 proteinopathy. Acta Neuropathol. 130, 63–75 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Mizielinska, S. et al. C9orf72 frontotemporal lobar degeneration is characterised by frequent neuronal sense and antisense RNA foci. Acta Neuropathol. 126, 845–857 (2013). A highly descriptive study quantifying RNA foci and RANT products in a brain of a patient with C9orf72 ALS.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gami, P. et al. A 30-unit hexanucleotide repeat expansion in C9orf72 induces pathological lesions with dipeptide-repeat proteins and RNA foci, but not TDP-43 inclusions and clinical disease. Acta Neuropathol. 130, 599–601 (2015).

    Article  PubMed  Google Scholar 

  52. Todd, P. K. & Paulson, H. L. RNA-mediated neurodegeneration in repeat expansion disorders. Ann. Neurol. 67, 291–300 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Miller, J. W. et al. Recruitment of human muscleblind proteins to (CUG)n expansions associated with myotonic dystrophy. EMBO J. 19, 4439–4448 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Cooper-Knock, J. et al. Sequestration of multiple RNA recognition motif-containing proteins by C9orf72 repeat expansions. Brain 137, 2040–2051 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Mori, K. et al. 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, 413–423 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Xu, Z. et al. Expanded GGGGCC repeat RNA associated with amyotrophic lateral sclerosis and frontotemporal dementia causes neurodegeneration. Proc. Natl Acad. Sci. USA 110, 7778–7783 (2013). The first study to show that (G 4 C 2 ) RNA expression in Drosophila is neurotoxic.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Prudencio, M. et al. Distinct brain transcriptome profiles in C9orf72-associated and sporadic ALS. Nat. Neurosci. 18, 1175–1182 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Lee, Y.-B. et al. Hexanucleotide repeats in ALS/FTD form length-dependent RNA foci, sequester RNA binding proteins, and are neurotoxic. Cell Rep. 5, 1178–1186 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Zhang, K. et al. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 525, 56–61 (2015). This study provides evidence of nucleocytoplasmic transport defects, including impaired nuclear protein import, in (G 4 C 2 ) RNA Drosophila and C9orf72 ALS/FTD iPSC-derived neurons.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zu, T. et al. Non-ATG-initiated translation directed by microsatellite expansions. Proc. Natl Acad. Sci. USA 108, 260–265 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Todd, P. K. et al. CGG repeat-associated translation mediates neurodegeneration in fragile X tremor ataxia syndrome. Neuron 78, 440–455 (2013).

    Article  CAS  PubMed  Google Scholar 

  62. Mori, K. et al. The C9orf72 GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in FTLD/ALS. Science 339, 1335–1338 (2013).

    Article  CAS  PubMed  Google Scholar 

  63. Ash, P. E. A. et al. Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron 77, 639–646 (2013). This paper describes the detection of RANT products in patients with C9orf72 ALS.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Mackenzie, I. R. et al. Dipeptide repeat protein pathology in C9ORF72 mutation cases: clinico-pathological correlations. Acta Neuropathol. 126, 859–879 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Mann, D. M. et al. Dipeptide repeat proteins are present in the p62 positive inclusions in patients with frontotemporal lobar degeneration and motor neurone disease associated with expansions in C9ORF72. Acta Neuropathol. Commun. 1, 68 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Cleary, J. D. & Ranum, L. P. Repeat associated non-ATG (RAN) translation: new starts in microsatellite expansion disorders. Curr. Opin. Genet. Dev. 26, 6–15 (2014).

    Article  CAS  PubMed  Google Scholar 

  67. Gomez-Deza, J. et al. Dipeptide repeat protein inclusions are rare in the spinal cord and almost absent from motor neurons in C9ORF72 mutant amyotrophic lateral sclerosis and are unlikely to cause their degeneration. Acta Neuropathol. Commun. 3, 38 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Schludi, M. H. et al. Distribution of dipeptide repeat proteins in cellular models and C9orf72 mutation cases suggests link to transcriptional silencing. Acta Neuropathol. 130, 537–555 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gendron, T. F. et al. Cerebellar c9RAN proteins associate with clinical and neuropathological characteristics of C9ORF72 repeat expansion carriers. Acta Neuropathol. 130, 559–573 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Cooper-Knock, J. et al. Clinico-pathological features in amyotrophic lateral sclerosis with expansions in C9ORF72. Brain 135, 751–764 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Baborie, A. et al. Accumulation of dipeptide repeat proteins predates that of TDP-43 in frontotemporal lobar degeneration associated with hexanucleotide repeat expansions in C9ORF72 gene. Neuropathol. Appl. Neurobiol. 41, 601–612 (2014).

    Article  CAS  Google Scholar 

  72. Chew, J. et al. Neurodegeneration. C9ORF72 repeat expansions in mice cause TDP-43 pathology, neuronal loss, and behavioral deficits. Science 348, 1151–1154 (2015). This report shows that adeno-associated virus-mediated overexpression of (G 4 C 2 ) RNA in rodent brain causes RNA foci, generates RANT products and induces neurodegeneration.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Su, Z. et al. Discovery of a biomarker and lead small molecules to target r(GGGGCC)-associated defects in c9FTD/ALS. Neuron 83, 1043–1050 (2014). This paper shows that small molecules that bind to (G 4 C 2 ) hairpins reduce RNA foci and RANT production, and that (GP) RANT products are present in the spinal cord of a patient with C9orf72 ALS.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Devlin, A.-C. et al. Human iPSC-derived motoneurons harbouring TARDBP or C9ORF72 ALS mutations are dysfunctional despite maintaining viability. Nat. Commun. 6, 5999 (2015).

    Article  CAS  PubMed  Google Scholar 

  75. Wainger, B. J. et al. Intrinsic membrane hyperexcitability of amyotrophic lateral sclerosis patient-derived motor neurons. Cell Rep. 7, 1–11 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Wainger, B. J. & Cudkowicz, M. E. Cortical hyperexcitability in amyotrophic lateral sclerosis: C9orf72 repeats. JAMA Neurol. 72, 1235–1236 (2015).

    Article  PubMed  Google Scholar 

  77. Kwon, I. et al. Poly-dipeptides encoded by the C9orf72 repeats bind nucleoli, impede RNA biogenesis, and kill cells. Science 345, 1139–1145 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. May, S. et al. C9orf72 FTLD/ALS-associated Gly-Ala dipeptide repeat proteins cause neuronal toxicity and Unc119 sequestration. Acta Neuropathol. 128, 485–503 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Zhang, Y.-J. et al. Aggregation-prone c9FTD/ALS poly(GA) RAN-translated proteins cause neurotoxicity by inducing ER stress. Acta Neuropathol. 128, 505–524 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Yang, D. et al. FTD/ALS-associated poly(GR) protein impairs the Notch pathway and is recruited by poly(GA) into cytoplasmic inclusions. Acta Neuropathol. 130, 525–535 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Tran, H. et al. Differential toxicity of nuclear RNA foci versus dipeptide repeat proteins in a Drosophila model of C9ORF72 FTD/ALS. Neuron 87, 1207–1214 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Yamakawa, M. et al. Characterization of the dipeptide repeat protein in the molecular pathogenesis of c9FTD/ALS. Hum. Mol. Genet. 24, 1630–1645 (2015).

    Article  CAS  PubMed  Google Scholar 

  83. Tsoi, H., Lau, T. C.-K., Tsang, S.-Y., Lau, K.-F. & Chan, H. Y. E. CAG expansion induces nucleolar stress in polyglutamine diseases. Proc. Natl Acad. Sci. USA 109, 13428–13433 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Tao, Z. et al. Nucleolar stress and impaired stress granule formation contribute to C9orf72 RAN translation-induced cytotoxicity. Hum. Mol. Genet. 24, 2426–2441 (2015).

    Article  CAS  PubMed  Google Scholar 

  85. Freibaum, B. D. et al. GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature 525, 129–133 (2015). An unbiased genetic screen of novel (G 4 C 2) Drosophila model reveals several genetic modifiers involved in nucleocytoplasmic transport and components of the NPC.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Jovicic, A. et al. Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. Nat. Neurosci. 18, 1226–1229 (2015). A genetic screen of (PR)-expressing yeast reveals many genetic modifiers involved in nucleocytoplasmic transport.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Woerner, A. C. et al. Cytoplasmic protein aggregates interfere with nucleocytoplasmic transport of protein and RNA. Science 351, 173–176 (2016).

    Article  CAS  PubMed  Google Scholar 

  88. Ling, J. P., Pletnikova, O., Troncoso, J. C. & Wong, P. C. TDP-43 repression of nonconserved cryptic exons is compromised in ALS-FTD. Science 349, 650–655 (2015). This paper reports that loss of TDP43 enhances the presence of cryptic exons.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Wheeler, T. M. et al. Targeting nuclear RNA for in vivo correction of myotonic dystrophy. Nature 488, 111–115 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Miller, T. M. et al. An antisense oligonucleotide against SOD1 delivered intrathecally for patients with SOD1 familial amyotrophic lateral sclerosis: a Phase 1, randomised, first-in-man study. Lancet Neurol. 12, 435–442 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Keil, J. M. et al. A short antisense oligonucleotide ameliorates symptoms of severe mouse models of spinal muscular atrophy. Mol. Ther. Nucleic Acids 3, e174 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Yang, W. Y., Wilson, H. D., Velagapudi, S. P. & Disney, M. D. Inhibition of Non-ATG translational events in cells via covalent small molecules targeting RNA. J. Am. Chem. Soc. 137, 5336–5345 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zamiri, B., Reddy, K., Macgregor, R. B. Jr & Pearson, C. E. TMPyP4 porphyrin distorts RNA G-quadruplex structures of the disease-associated r(GGGGCC)n repeat of the C9orf72 gene and blocks interaction of RNA-binding proteins. J. Biol. Chem. 289, 4653–4659 (2014).

    Article  CAS  PubMed  Google Scholar 

  94. Haines, J. D. et al. Nuclear export inhibitors avert progression in preclinical models of inflammatory demyelination. Nat. Neurosci. 18, 511–520 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Winer, L. et al. SOD1 in cerebral spinal fluid as a pharmacodynamic marker for antisense oligonucleotide therapy. JAMA Neurol. 70, 201–207 (2013).

    Article  PubMed  Google Scholar 

  96. Liang, P. et al. CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes. Protein Cell 6, 363–372 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Xi, Z. et al. Jump from pre-mutation to pathologic expansion in C9orf72. Am. J. Hum. Genet. 96, 962–970 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Xi, Z. et al. The C9orf72 repeat expansion itself is methylated in ALS and FTLD patients. Acta Neuropathol. 129, 715–727 (2015).

    Article  CAS  PubMed  Google Scholar 

  99. McMurray, C. T. Mechanisms of trinucleotide repeat instability during human development. Nat. Rev. Genet. 11, 786–799 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Suh, E. et al. Semi-automated quantification of C9orf72 expansion size reveals inverse correlation between hexanucleotide repeat number and disease duration in frontotemporal degeneration. Acta Neuropathol. 130, 363–372 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Gijselinck, I. et al. The C9orf72 repeat size correlates with onset age of disease, DNA methylation and transcriptional downregulation of the promoter. Mol. Psychiatry http://dx.doi.org/10.1038/mp.2015.159 (2015).

  102. Gallagher, M. D. et al. TMEM106B is a genetic modifier of frontotemporal lobar degeneration with C9orf72 hexanucleotide repeat expansions. Acta Neuropathol. 127, 407–418 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. van Blitterswijk, M. et al. TMEM106B protects C9ORF72 expansion carriers against frontotemporal dementia. Acta Neuropathol. 127, 397–406 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Mirkin, S. M. Expandable DNA repeats and human disease. Nature 447, 932–940 (2007).

    Article  CAS  PubMed  Google Scholar 

  105. Orr, H. T. & Zoghbi, H. Y. Trinucleotide repeat disorders. Annu. Rev. Neurosci. 30, 575–621 (2007).

    Article  CAS  PubMed  Google Scholar 

  106. La Spada, A. R. & Taylor, J. P. Repeat expansion disease: progress and puzzles in disease pathogenesis. Nat. Rev. Genet. 11, 247–258 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Pearson, C. E., Nichol Edamura, K. & Cleary, J. D. Repeat instability: mechanisms of dynamic mutations. Nat. Rev. Genet. 6, 729–742 (2005).

    Article  CAS  PubMed  Google Scholar 

  108. Dols-Icardo, O. et al. Characterization of the repeat expansion size in C9orf72 in amyotrophic lateral sclerosis and frontotemporal dementia. Hum. Mol. Genet. 23, 749–754 (2014).

    Article  CAS  PubMed  Google Scholar 

  109. Nordin, A. et al. Extensive size variability of the GGGGCC expansion in C9orf72 in both neuronal and non-neuronal tissues in 18 patients with ALS or FTD. Hum. Mol. Genet. 24, 3133–3142 (2015).

    Article  CAS  PubMed  Google Scholar 

  110. Aguilera, A. & Garcia-Muse, T. Causes of genome instability. Annu. Rev. Genet. 47, 1–32 (2013).

    Article  CAS  PubMed  Google Scholar 

  111. Lopez Castel, A., Cleary, J. D. & Pearson, C. E. Repeat instability as the basis for human diseases and as a potential target for therapy. Nat. Rev. Mol. Cell Biol. 11, 165–170 (2010).

    Article  CAS  PubMed  Google Scholar 

  112. Kruisselbrink, E. et al. Mutagenic capacity of endogenous G4 DNA underlies genome instability in FANCJ-defective C. elegans. Curr. Biol. 18, 900–905 (2008).

    Article  CAS  PubMed  Google Scholar 

  113. Ribeyre, C. et al. The yeast Pif1 helicase prevents genomic instability caused by G-quadruplex-forming CEB1 sequences in vivo. PLoS Genet. 5, e1000475 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Piazza, A. et al. Genetic instability triggered by G-quadruplex interacting Phen-DC compounds in Saccharomyces cerevisiae. Nucleic Acids Res. 38, 4337–4348 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. De, S. & Michor, F. DNA secondary structures and epigenetic determinants of cancer genome evolution. Nat. Struct. Mol. Biol. 18, 950–955 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Aghili, L., Foo, J., DeGregori, J. & De, S. Patterns of somatically acquired amplifications and deletions in apparently normal tissues of ovarian cancer patients. Cell Rep. 7, 1310–1319 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Koole, W. et al. A polymerase Theta-dependent repair pathway suppresses extensive genomic instability at endogenous G4 DNA sites. Nat. Commun. 5, 3216 (2014).

    Article  CAS  PubMed  Google Scholar 

  118. van Kregten, M. & Tijsterman, M. The repair of G-quadruplex-induced DNA damage. Exp. Cell Res. 329, 178–183 (2014).

    Article  CAS  PubMed  Google Scholar 

  119. Aguilera, A. The connection between transcription and genomic instability. EMBO J. 21, 195–201 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Li, X. & Manley, J. L. Cotranscriptional processes and their influence on genome stability. Genes Dev. 20, 1838–1847 (2006).

    Article  CAS  PubMed  Google Scholar 

  121. Aguilera, A. & Garcia-Muse, T. R loops: from transcription byproducts to threats to genome stability. Mol. Cell 46, 115–124 (2012).

    Article  CAS  PubMed  Google Scholar 

  122. Bhatia, V. et al. BRCA2 prevents R-loop accumulation and associates with TREX-2 mRNA export factor PCID2. Nature 511, 362–365 (2014).

    Article  CAS  PubMed  Google Scholar 

  123. Skourti-Stathaki, K. & Proudfoot, N. J. A double-edged sword: R loops as threats to genome integrity and powerful regulators of gene expression. Genes Dev. 28, 1384–1396 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Lin, Y., Dent, S. Y., Wilson, J. H., Wells, R. D. & Napierala, M. R loops stimulate genetic instability of CTG. CAG repeats. Proc. Natl Acad. Sci. USA 107, 692–697 (2010).

    Article  CAS  PubMed  Google Scholar 

  125. Nakamori, M., Pearson, C. E. & Thornton, C. A. Bidirectional transcription stimulates expansion and contraction of expanded (CTG)*(CAG) repeats. Hum. Mol. Genet. 20, 580–588 (2011).

    Article  CAS  PubMed  Google Scholar 

  126. Reddy, K. et al. Processing of double-R-loops in (CAG).(CTG) and C9orf72 (GGGGCC).(GGCCCC) repeats causes instability. Nucleic Acids Res. 42, 10473–10487 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Moreira, M. C. et al. Senataxin, the ortholog of a yeast RNA helicase, is mutant in ataxia-ocular apraxia 2. Nat. Genet. 36, 225–227 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Martin-Tumasz, S. & Brow, D. A. S. cerevisiae Sen1 helicase domain exhibits 5′ to 3′ helicase activity with a preference for translocation on DNA rather than RNA. J. Biol. Chem. 290, 22880–22889 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Belzil, V. V. et al. Characterization of DNA hypermethylation in the cerebellum of c9FTD/ALS patients. Brain Res. 1584, 15–21 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Liu, E. Y. et al. C9orf72 hypermethylation protects against repeat expansion-associated pathology in ALS/FTD. Acta Neuropathol. 128, 525–541 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. McMillan, C. T. et al. C9orf72 promoter hypermethylation is neuroprotective: neuroimaging and neuropathologic evidence. Neurology 84, 1622–1630 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Russ, J. et al. Hypermethylation of repeat expanded C9orf72 is a clinical and molecular disease modifier. Acta Neuropathol. 129, 39–52 (2015).

    Article  CAS  PubMed  Google Scholar 

  134. Naumann, A., Kraus, C., Hoogeveen, A., Ramirez, C. M. & Doerfler, W. Stable DNA methylation boundaries and expanded trinucleotide repeats: role of DNA insertions. J. Mol. Biol. 426, 2554–2566 (2014).

    Article  CAS  PubMed  Google Scholar 

  135. Simone, R., Fratta, P., Neidle, S., Parkinson, G. N. & Isaacs, A. M. G-quadruplexes: emerging roles in neurodegenerative diseases and the non-coding transcriptome. FEBS Lett. 589, 1653–1668 (2015).

    Article  CAS  PubMed  Google Scholar 

  136. Wang, J., Haeusler, A. R. & Simko, E. A. Emerging role of RNA*DNA hybrids in C9orf72-linked neurodegeneration. Cell Cycle 14, 526–532 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Lin, J. et al. Stabilization of G-quadruplex DNA by C-5-methyl-cytosine in bcl-2 promoter: implications for epigenetic regulation. Biochem. Biophys. Res. Commun. 433, 368–373 (2013).

    Article  CAS  PubMed  Google Scholar 

  138. Brcic, J. & Plavec, J. Solution structure of a DNA quadruplex containing ALS and FTD related GGGGCC repeat stabilized by 8-bromodeoxyguanosine substitution. Nucleic Acids Res. 43, 8590–8600 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Wojciechowska, M., Olejniczak, M., Galka-Marciniak, P., Jazurek, M. & Krzyzosiak, W. J. RAN translation and frameshifting as translational challenges at simple repeats of human neurodegenerative disorders. Nucleic Acids Res. 42, 11849–11864 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Colussi, T. M. et al. Initiation of translation in bacteria by a structured eukaryotic IRES RNA. Nature 519, 110–113 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Ren, Q. et al. Alternative reading frame selection mediated by a tRNA-like domain of an internal ribosome entry site. Proc. Natl Acad. Sci. USA 109, E630–E639 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Toulouse, A. et al. Ribosomal frameshifting on MJD-1 transcripts with long CAG tracts. Hum. Mol. Genet. 14, 2649–2660 (2005).

    Article  CAS  PubMed  Google Scholar 

  143. Gaspar, C. et al. CAG tract of MJD-1 may be prone to frameshifts causing polyalanine accumulation. Hum. Mol. Genet. 9, 1957–1966 (2000).

    Article  CAS  PubMed  Google Scholar 

  144. Davies, J. E. & Rubinsztein, D. C. Polyalanine and polyserine frameshift products in Huntington's disease. J. Med. Genet. 43, 893–896 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Stochmanski, S. J. et al. Expanded ATXN3 frameshifting events are toxic in Drosophila and mammalian neuron models. Hum. Mol. Genet. 21, 2211–2218 (2012).

    CAS  PubMed  Google Scholar 

  146. Girstmair, H. et al. Depletion of cognate charged transfer RNA causes translational frameshifting within the expanded CAG stretch in huntingtin. Cell Rep. 3, 148–159 (2013).

    Article  CAS  PubMed  Google Scholar 

  147. Yu, C. H., Teulade-Fichou, M. P. & Olsthoorn, R. C. Stimulation of ribosomal frameshifting by RNA G-quadruplex structures. Nucleic Acids Res. 42, 1887–1892 (2014).

    Article  CAS  PubMed  Google Scholar 

  148. Cammas, A. et al. Stabilization of the G-quadruplex at the VEGF IRES represses cap-independent translation. RNA Biol. 12, 320–329 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank L. Ranum, A. Gitler, L. Petrucelli, P. Taylor, D. Cleveland, F. Bennet, F. Rigo, J. Wang, P. Wong and T. Lloyd for discussions that helped provide a foundation for this Review. Financial support was provided from the following institutes: the National Institutes of Health-National Institute of Neurological Disorders and Stroke (NIH-NINDS); Target ALS; the Muscular Dystrophy Association; the Robert Packard Center for ALS Research; the Live Like Lou at the Pittsburgh Foundation; and the Brain Institute at the University of Pittsburgh School of Medicine, USA.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Christopher J. Donnelly or Jeffrey D. Rothstein.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information S1 (table)

Mechanisms of C9orf72 Pathophysiology (PDF 156 kb)

Supplementary information S2 (table)

Author-provided unique (G4C2)n RNA•protein interactors lists* (XLSX 44 kb)

PowerPoint slides

Glossary

GDP–GTP nucleotide exchange factor

(GEF). A protein that functions to stimulate the release of guanosine diphosphate (GDP) from a GTPase to allow subsequent binding of the guanosine triphosphate (GTP) to the active site.

Epigenetic silencing

Covalent DNA or histone modifications, such as DNA or histone methylation, that act to repress or silence a genomic region by reducing access of the transcriptional machinery to the DNA.

Polymerase processivity

A relative measure of the functional efficiency and rate of polymerase to process (replicate or transcribe) nucleic acid information.

Bidirectional transcription

Transcription that occurs simultaneously on both the positive and the negative strands of DNA, where the direction of RNA polymerase progression along each strand is either convergent or divergent.

Haplotype

A combination of alleles at different loci in the genome that tend to be inherited together because they show high linkage disequilibrium (often because they are physically close).

Induced pluripotent stem cells

(iPSCs). Cells that are created from differentiated cell types — for example, fibroblasts — that are reprogrammed by a cocktail of transcription factors (or other approaches) back to a pluripotent state. These cells can then be differentiated into cells of distinct lineages — for example, neurons.

R loops

A nucleic acid structure that occurs when a single strand of RNA invades a double-stranded DNA, forming an RNA:DNA hybrid that is stabilized through Watson–Crick base pairs, and leaves a displaced single-stranded DNA molecule.

dsDNA melting

The separation of base-paired DNA strands through the disruption of Watson–Crick pairing by physical or thermodynamic means.

Interactomes

Sets of physical interactions occurring between two or more components.

G-quadruplex

A structure composed of G-quartets, four planar guanine molecules that are stabilized by Hoogsteen base pairing, that are stacked on top of each other and stabilized by a central cation and π-stacking interactions between the G-quartets.

Hairpin

A nucleic acid molecule that contains a stem region, which can be composed of complimentary base pairing and be interspersed with single-stranded or loop elements, and contains a tight looped region (hairpin turn) at the end of the base-paired stem region.

Repeat-associated non-ATG-dependent translation

(RANT). The non-canonical translation of a repetitive RNA sequence initiated by an AUG start codon.

Nucleocytoplasmic trafficking

The bidirectional protein transport between the cytoplasm and the nuclear matrix through the nuclear pore complex.

Genetic modifier

A genetic variation in (cis) or outside (trans) a gene or genetic locus that alters the phenotypic expression of the gene.

CRISPR/Cas technology

The essential components of the bacterial adaptive immunity, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR-associated (Cas) genes, that are currently being utilized and developed for precise editing, regulating and targeting of genes in model systems and organisms.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Haeusler, A., Donnelly, C. & Rothstein, J. The expanding biology of the C9orf72 nucleotide repeat expansion in neurodegenerative disease. Nat Rev Neurosci 17, 383–395 (2016). https://doi.org/10.1038/nrn.2016.38

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn.2016.38

This article is cited by

Search

Quick links

Nature Briefing

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

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