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:

Molecular mechanisms underlying nucleotide repeat expansion disorders

An Author Correction to this article was published on 06 July 2021

This article has been updated

Abstract

The human genome contains over one million short tandem repeats. Expansion of a subset of these repeat tracts underlies over fifty human disorders, including common genetic causes of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (C9orf72), polyglutamine-associated ataxias and Huntington disease, myotonic dystrophy, and intellectual disability disorders such as Fragile X syndrome. In this Review, we discuss the four major mechanisms by which expansion of short tandem repeats causes disease: loss of function through transcription repression, RNA-mediated gain of function through gelation and sequestration of RNA-binding proteins, gain of function of canonically translated repeat-harbouring proteins, and repeat-associated non-AUG translation of toxic repeat peptides. Somatic repeat instability amplifies these mechanisms and influences both disease age of onset and tissue specificity of pathogenic features. We focus on the crosstalk between these disease mechanisms, and argue that they often synergize to drive pathogenesis. We also discuss the emerging native functions of repeat elements and how their dynamics might contribute to disease at a larger scale than currently appreciated. Lastly, we propose that lynchpins tying these disease mechanisms and native functions together offer promising therapeutic targets with potential shared applications across this class of human disorders.

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

Access options

Buy this article

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

Fig. 1: Molecular mechanisms of nucleotide repeat expansion pathogenesis.
Fig. 2: Repeat-induced transcriptional gene silencing, R-loops and somatic instability.
Fig. 3: Mechanisms of RNA toxicity in repeat expansion diseases.
Fig. 4: Mechanisms of repeat-associated non-AUG translation.
Fig. 5: Synergy across pathogenic mechanisms of repeat expansion diseases.
Fig. 6: Roles of repeats in human disease and neuronal function.

Similar content being viewed by others

Change history

References

  1. Lander, E. S. et al. Initial sequencing and analysis of the human genome. Nature 409, 860–921 (2001).

    Article  CAS  PubMed  Google Scholar 

  2. Kruglyak, S., Durrett, R. T., Schug, M. D. & Aquadro, C. F. Equilibrium distributions of microsatellite repeat length resulting from a balance between slippage events and point mutations. Proc. Natl Acad. Sci. USA 95, 10774–10778 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Quilez, J. et al. Polymorphic tandem repeats within gene promoters act as modifiers of gene expression and DNA methylation in humans. Nucleic Acids Res. 44, 3750–3762 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Fotsing, S. F. et al. The impact of short tandem repeat variation on gene expression. Nat. Genet. 51, 1652–1659 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Fu, Y. H. et al. Variation of the CGG repeat at the fragile X site results in genetic instability: resolution of the Sherman paradox. Cell 67, 1047–1058 (1991).

    Article  CAS  PubMed  Google Scholar 

  6. Oberlé, I. et al. Instability of a 550-base pair DNA segment and abnormal methylation in fragile X syndrome. Science 252, 1097–1102 (1991).

    Article  PubMed  Google Scholar 

  7. Verkerk, A. J. et al. Identification of a gene (FMR-1) containing a CGG repeat coincident with a breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell 65, 905–914 (1991).

    Article  CAS  PubMed  Google Scholar 

  8. Kremer, E. J. et al. Mapping of DNA instability at the fragile X to a trinucleotide repeat sequence p(CCG)n. Science 252, 1711–1714 (1991).

    Article  CAS  PubMed  Google Scholar 

  9. La Spada, A. R., Wilson, E. M., Lubahn, D. B., Harding, A. E. & Fischbeck, K. H. Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy. Nature 352, 77–79 (1991).

    Article  PubMed  Google Scholar 

  10. Hannan, A. J. Tandem repeats mediating genetic plasticity in health and disease. Nat. Rev. Genet. 19, 286–298 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Gymrek, M. A genomic view of short tandem repeats. Curr. Opin. Genet. Dev. 44, 9–16 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Balzano, E., Pelliccia, F. & Giunta, S. Genome (in)stability at tandem repeats. Semin. Cell Dev. Biol. https://doi.org/10.1016/j.semcdb.2020.10.003 (2020).

    Article  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Nussbacher, J. K., Tabet, R., Yeo, G. W. & Lagier-Tourenne, C. Disruption of RNA metabolism in neurological diseases and emerging therapeutic interventions. Neuron 102, 294–320 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Khristich, A. N. & Mirkin, S. M. On the wrong DNA track: molecular mechanisms of repeat-mediated genome instability. J. Biol. Chem. 295, 4134–4170 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Paulson, H. Repeat expansion diseases. Handb. Clin. Neurol. 147, 105–123 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Wells, R. D. & Ashizawa, T. Genetic Instabilities and Neurological Diseases. (Elsevier, 2006).

  18. Martin, J. P. & Bell, J. A pedigree of mental defect showing sex-linkage. J. Neurol. Psychiatry 6, 154–157 (1943).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lubs, H. A. A marker X chromosome. Am. J. Hum. Genet. 21, 231–244 (1969).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Eichler, E. E. et al. Evolution of the cryptic FMR1 CGG repeat. Nat. Genet. 11, 301–308 (1995).

    Article  CAS  PubMed  Google Scholar 

  21. Sutcliffe, J. S. et al. DNA methylation represses FMR-1 transcription in fragile X syndrome. Hum. Mol. Genet. 1, 397–400 (1992).

    Article  CAS  PubMed  Google Scholar 

  22. Coffee, B., Zhang, F., Ceman, S., Warren, S. T. & Reines, D. Histone modifications depict an aberrantly heterochromatinized FMR1 gene in fragile X syndrome. Am. J. Hum. Genet. 71, 923–932 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Gedeon, A. K. et al. Fragile X syndrome without CCG amplification has an FMR1 deletion. Nat. Genet. 1, 341–344 (1992).

    Article  CAS  PubMed  Google Scholar 

  24. De Boulle, K. et al. A point mutation in the FMR-1 gene associated with fragile X mental retardation. Nat. Genet. 3, 31–35 (1993).

    Article  PubMed  Google Scholar 

  25. Santoro, M. R., Bray, S. M. & Warren, S. T. Molecular mechanisms of fragile X syndrome: a twenty-year perspective. Annu. Rev. Pathol. 7, 219–245 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Usdin, K. & Kumari, D. Repeat-mediated epigenetic dysregulation of the FMR1 gene in the fragile X-related disorders. Front. Genet. 6, 192 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Colak, D. et al. Promoter-bound trinucleotide repeat mRNA drives epigenetic silencing in fragile X syndrome. Science 343, 1002–1005 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kumari, D. & Usdin, K. Polycomb group complexes are recruited to reactivated FMR1 alleles in Fragile X syndrome in response to FMR1 transcription. Hum. Mol. Genet. 23, 6575–6583 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Eiges, R. et al. Developmental study of fragile X syndrome using human embryonic stem cells derived from preimplantation genetically diagnosed embryos. Cell Stem Cell 1, 568–577 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Kumari, D., Sciascia, N. & Usdin, K. Small molecules targeting H3K9 methylation prevent silencing of reactivated FMR1 alleles in fragile X syndrome patient derived cells. Genes 11, (2020).

  31. Liu, X. S. et al. Rescue of fragile X syndrome neurons by DNA methylation editing of the FMR1 gene. Cell 172, 979–992.e6 (2018). Targeting TET1–dCas9 to the CGG repeat of the FMR1 locus drives demethylation and gene reactivation in iPS cell-derived neurons, resulting in phenotypic correction.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Chiurazzi, P., Pomponi, M. G., Willemsen, R., Oostra, B. A. & Neri, G. In vitro reactivation of the FMR1 gene involved in fragile X syndrome. Hum. Mol. Genet. 7, 109–113 (1998).

    Article  CAS  PubMed  Google Scholar 

  33. Godde, J. S., Kass, S. U., Hirst, M. C. & Wolffe, A. P. Nucleosome assembly on methylated CGG triplet repeats in the fragile X mental retardation gene 1 promoter. J. Biol. Chem. 271, 24325–24328 (1996).

    Article  CAS  PubMed  Google Scholar 

  34. 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 

  35. 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 

  36. 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 

  37. 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 

  38. Gijselinck, I. et al. The C9orf72 repeat size correlates with onset age of disease, DNA methylation and transcriptional downregulation of the promoter. Mol. Psychiatry 21, 1112–1124 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Gu, Y., Shen, Y., Gibbs, R. A. & Nelson, D. L. Identification of FMR2, a novel gene associated with the FRAXE CCG repeat and CpG island. Nat. Genet. 13, 109–113 (1996).

    Article  CAS  PubMed  Google Scholar 

  40. Gecz, J., Gedeon, A. K., Sutherland, G. R. & Mulley, J. C. Identification of the gene FMR2, associated with FRAXE mental retardation. Nat. Genet. 13, 105–108 (1996).

    Article  CAS  PubMed  Google Scholar 

  41. Campuzano, V. et al. Friedreich’s ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion. Science 271, 1423–1427 (1996).

    Article  CAS  PubMed  Google Scholar 

  42. Gottesfeld, J. M. Molecular mechanisms and therapeutics for the GAA·TTC expansion disease Friedreich ataxia. Neurother. J. Am. Soc. Exp. Neurother. 16, 1032–1049 (2019).

    Google Scholar 

  43. Bidichandani, S. I., Ashizawa, T. & Patel, P. I. The GAA triplet-repeat expansion in Friedreich ataxia interferes with transcription and may be associated with an unusual DNA structure. Am. J. Hum. Genet. 62, 111–121 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Rodden, L. N. et al. Methylated and unmethylated epialleles support variegated epigenetic silencing in Friedreich ataxia. Hum. Mol. Genet. 29, 3818–3829 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Sakamoto, N. et al. Sticky DNA: self-association properties of long GAA.TTC repeats in R.R.Y triplex structures from Friedreich’s ataxia. Mol. Cell 3, 465–475 (1999).

    Article  CAS  PubMed  Google Scholar 

  46. De Biase, I., Chutake, Y. K., Rindler, P. M. & Bidichandani, S. I. Epigenetic silencing in Friedreich ataxia is associated with depletion of CTCF (CCCTC-binding factor) and antisense transcription. PLoS ONE 4, e7914 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Mikaeili, H., Sandi, M., Bayot, A., Al-Mahdawi, S. & Pook, M. A. FAST-1 antisense RNA epigenetically alters FXN expression. Sci. Rep. 8, 17217 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Eimer, H. et al. RNA-dependent epigenetic silencing directs transcriptional downregulation caused by intronic repeat expansions. Cell 174, 1095–1105.e11 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Li, L., Matsui, M. & Corey, D. R. Activating frataxin expression by repeat-targeted nucleic acids. Nat. Commun. 7, 10606 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Punga, T. & Bühler, M. Long intronic GAA repeats causing Friedreich ataxia impede transcription elongation. EMBO Mol. Med. 2, 120–129 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kim, E., Napierala, M. & Dent, S. Y. R. Hyperexpansion of GAA repeats affects post-initiation steps of FXN transcription in Friedreich’s ataxia. Nucleic Acids Res. 39, 8366–8377 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kumari, D., Biacsi, R. E. & Usdin, K. Repeat expansion affects both transcription initiation and elongation in Friedreich ataxia cells. J. Biol. Chem. 286, 4209–4215 (2011).

    Article  CAS  PubMed  Google Scholar 

  53. Reddy, K. et al. Determinants of R-loop formation at convergent bidirectionally transcribed trinucleotide repeats. Nucleic Acids Res. 39, 1749–1762 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Loomis, E. W., Sanz, L. A., Chédin, F. & Hagerman, P. J. Transcription-associated R-loop formation across the human FMR1 CGG-repeat region. PLOS Genet. 10, e1004294 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Abu Diab, M. et al. The G-rich repeats in FMR1 and C9orf72 loci are hotspots for local unpairing of DNA. Genetics 210, 1239–1252 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Robin, G. et al. Calcium dysregulation and Cdk5-ATM pathway involved in a mouse model of fragile X-associated tremor/ataxia syndrome. Hum. Mol. Genet. 26, 2649–2666 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Farg, M. A., Konopka, A., Soo, K. Y., Ito, D. & Atkin, J. D. The DNA damage response (DDR) is induced by the C9orf72 repeat expansion in amyotrophic lateral sclerosis. Hum. Mol. Genet. 26, 2882–2896 (2017).

    Article  CAS  PubMed  Google Scholar 

  58. 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 

  59. Lin, Y., Dent, S. Y. R., 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 

  60. Massey, T. H. & Jones, L. The central role of DNA damage and repair in CAG repeat diseases. Dis. Model. Mech. 11, dmm031930 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Askeland, G. et al. Increased nuclear DNA damage precedes mitochondrial dysfunction in peripheral blood mononuclear cells from Huntington’s disease patients. Sci. Rep. 8, 9817 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Moshell, A. N., Tarone, R. E., Barrett, S. F. & Robbins, J. H. Radiosensitivity in Huntington’s disease: implications for pathogenesis and presymptomatic diagnosis. Lancet Lond. Engl. 1, 9–11 (1980).

    Article  CAS  Google Scholar 

  63. Xiao, H. et al. A polyglutamine expansion disease protein sequesters PTIP to attenuate DNA repair and increase genomic instability. Hum. Mol. Genet. 21, 4225–4236 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. López 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  PubMed  CAS  Google Scholar 

  65. 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  PubMed  PubMed Central  CAS  Google Scholar 

  66. Pearson, C. E., Ewel, A., Acharya, S., Fishel, R. A. & Sinden, R. R. Human MSH2 binds to trinucleotide repeat DNA structures associated with neurodegenerative diseases. Hum. Mol. Genet. 6, 1117–1123 (1997).

    Article  CAS  PubMed  Google Scholar 

  67. Keogh, N., Chan, K. Y., Li, G.-M. & Lahue, R. S. MutSβ abundance and Msh3 ATP hydrolysis activity are important drivers of CTG•CAG repeat expansions. Nucleic Acids Res. 45, 10068–10078 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Neil, A. J. et al. Replication-independent instability of Friedreich’s ataxia GAA repeats during chronological aging. Proc. Natl Acad. Sci. USA 118, e2013080118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gonitel, R. et al. DNA instability in postmitotic neurons. Proc. Natl Acad. Sci. USA 105, 3467–3472 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Genetic Modifiers of Huntington’s Disease (GeM-HD) Consortium. Identification of genetic factors that modify clinical onset of Huntington’s disease. Cell 162, 516–526 (2015).

    Article  CAS  Google Scholar 

  71. Lee, J.-M. et al. A modifier of Huntington’s disease onset at the MLH1 locus. Hum. Mol. Genet. 26, 3859–3867 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Bettencourt, C. et al. DNA repair pathways underlie a common genetic mechanism modulating onset in polyglutamine diseases: DNA repair pathways modify polyQ disease onset. Ann. Neurol. 79, 983–990 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Kovalenko, M. et al. Msh2 acts in medium-spiny striatal neurons as an enhancer of CAG instability and mutant huntingtin phenotypes in Huntington’s disease knock-in mice. PLoS ONE 7, e44273 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Pinto, R. M. et al. Mismatch repair genes Mlh1 and Mlh3 modify CAG instability in Huntington’s disease mice: genome-wide and candidate approaches. PLoS Genet. 9, e1003930 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Nakamori, M. et al. A slipped-CAG DNA-binding small molecule induces trinucleotide-repeat contractions in vivo. Nat. Genet. 52, 146–159 (2020). Identification of small-molecule binders to CTG/CAG slip-outs that form during repeat transcription and replication reveals that the small molecules favoured MMR-dependent repeat contraction.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Taneja, K. L., McCurrach, M., Schalling, M., Housman, D. & Singer, R. H. Foci of trinucleotide repeat transcripts in nuclei of myotonic dystrophy cells and tissues. J. Cell Biol. 128, 995–1002 (1995).

    Article  CAS  PubMed  Google Scholar 

  77. Zhang, N. & Ashizawa, T. RNA toxicity and foci formation in microsatellite expansion diseases. Curr. Opin. Genet. Dev. 44, 17–29 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. White, M. C. et al. Inactivation of hnRNP K by expanded intronic AUUCU repeat induces apoptosis via translocation of PKCδ to mitochondria in spinocerebellar ataxia 10. PLOS Genet. 6, e1000984 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Mizielinska, S. et al. C9orf72 frontotemporal lobar degeneration is characterised by frequent neuronal sense and antisense RNA foci. Acta Neuropathol. 126, 845–857 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. 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 

  81. Burguete, A. S. et al. GGGGCC microsatellite RNA is neuritically localized, induces branching defects, and perturbs transport granule function. eLife 4, e08881 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  82. 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).

    Article  CAS  PubMed  Google Scholar 

  83. Botta, A. et al. The CTG repeat expansion size correlates with the splicing defects observed in muscles from myotonic dystrophy type 1 patients. J. Med. Genet. 45, 639–646 (2008).

    Article  CAS  PubMed  Google Scholar 

  84. Wojciechowska, M. & Krzyzosiak, W. J. Cellular toxicity of expanded RNA repeats: focus on RNA foci. Hum. Mol. Genet. 20, 3811–3821 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Krzyzosiak, W. J. et al. Triplet repeat RNA structure and its role as pathogenic agent and therapeutic target. Nucleic Acids Res. 40, 11–26 (2012).

    Article  CAS  PubMed  Google Scholar 

  86. Fratta, P. et al. C9orf72 hexanucleotide repeat associated with amyotrophic lateral sclerosis and frontotemporal dementia forms RNA G-quadruplexes. Sci. Rep. 2, 1016 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Reddy, K., Zamiri, B., Stanley, S. Y. R., Macgregor, R. B. & Pearson, C. E. The disease-associated r(GGGGCC)n repeat from the C9orf72 gene forms tract length-dependent uni- and multimolecular RNA G-quadruplex structures. J. Biol. Chem. 288, 9860–9866 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Conlon, E. G. et al. The C9ORF72 GGGGCC expansion forms RNA G-quadruplex inclusions and sequesters hnRNP H to disrupt splicing in ALS brains. eLife 5, e17820 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Mooers, B. H. M., Logue, J. S. & Berglund, J. A. The structural basis of myotonic dystrophy from the crystal structure of CUG repeats. Proc. Natl Acad. Sci. USA 102, 16626–16631 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Tian, B. et al. Expanded CUG repeat RNAs form hairpins that activate the double-stranded RNA-dependent protein kinase PKR. RNA 6, 79–87 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. van Cruchten, R. T. P., Wieringa, B. & Wansink, D. G. Expanded CUG repeats in DMPK transcripts adopt diverse hairpin conformations without influencing the structure of the flanking sequences. RNA 25, 481–495 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Jain, A. & Vale, R. D. RNA phase transitions in repeat expansion disorders. Nature 546, 243–247 (2017). Repeat RNAs form phase-separated droplets through gelation in vitro and in nuclei, with variable properties depending on repeat length and sequence.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Querido, E., Gallardo, F., Beaudoin, M., Ménard, C. & Chartrand, P. Stochastic and reversible aggregation of mRNA with expanded CUG-triplet repeats. J. Cell Sci. 124, 1703–1714 (2011).

    Article  CAS  PubMed  Google Scholar 

  94. 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 

  95. Van Treeck, B. & Parker, R. Emerging roles for intermolecular RNA-RNA interactions in RNP assemblies. Cell 174, 791–802 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Li, P. et al. Phase transitions in the assembly of multivalent signalling proteins. Nature 483, 336–340 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Banjade, S. & Rosen, M. K. Phase transitions of multivalent proteins can promote clustering of membrane receptors. eLife 3, e04123 (2014).

    Article  PubMed Central  Google Scholar 

  98. Lin, Y., Protter, D. S. W., Rosen, M. K. & Parker, R. Formation and maturation of phase-separated liquid droplets by RNA-binding proteins. Mol. Cell 60, 208–219 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Loughlin, F. E. et al. Tandem RNA binding sites induce self-association of the stress granule marker protein TIA-1. Nucleic Acids Res. 49, 2403–2417 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  100. Rhine, K., Vidaurre, V. & Myong, S. RNA droplets. Annu. Rev. Biophys. 49, 247–265 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Wang, E. T. et al. Transcriptome alterations in myotonic dystrophy skeletal muscle and heart. Hum. Mol. Genet. 28, 1312–1321 (2019).

    Article  CAS  PubMed  Google Scholar 

  102. Kanadia, R. N. et al. A muscleblind knockout model for myotonic dystrophy. Science 302, 1978–1980 (2003).

    Article  CAS  PubMed  Google Scholar 

  103. Mankodi, A. et al. Myotonic dystrophy in transgenic mice expressing an expanded CUG repeat. Science 289, 1769–1773 (2000).

    Article  CAS  PubMed  Google Scholar 

  104. Kanadia, R. N. et al. Reversal of RNA missplicing and myotonia after muscleblind overexpression in a mouse poly(CUG) model for myotonic dystrophy. Proc. Natl Acad. Sci. USA 103, 11748–11753 (2006). Kanadia et al. (2003) and Kanadia et al. (2006) establish in mice that sequestration of MBNL protein by CUG repeat RNA is sufficient to explain most of the muscle phenotypes observed in DM1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  Google Scholar 

  106. 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 

  107. Zhang, K. et al. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 525, 56–61 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. 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 

  109. Sellier, C. et al. Sequestration of DROSHA and DGCR8 by expanded CGG RNA repeats alters microRNA processing in fragile X-associated tremor/ataxia syndrome. Cell Rep. 3, 869–880 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Sofola, O. A. et al. RNA-binding proteins hnRNP A2/B1 and CUGBP1 suppress fragile X CGG premutation repeat-induced neurodegeneration in a Drosophila model of FXTAS. Neuron 55, 565–571 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Jin, P. et al. Pur alpha binds to rCGG repeats and modulates repeat-mediated neurodegeneration in a Drosophila model of fragile X tremor/ataxia syndrome. Neuron 55, 556–564 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Batra, R. et al. Loss of MBNL leads to disruption of developmentally regulated alternative polyadenylation in RNA-mediated disease. Mol. Cell 56, 311–322 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. 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 

  114. Masuda, A. et al. CUGBP1 and MBNL1 preferentially bind to 3′ UTRs and facilitate mRNA decay. Sci. Rep. 2, 209 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Farina, K. L., Huttelmaier, S., Musunuru, K., Darnell, R. & Singer, R. H. Two ZBP1 KH domains facilitate beta-actin mRNA localization, granule formation, and cytoskeletal attachment. J. Cell Biol. 160, 77–87 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Wang, E. T. et al. Transcriptome-wide regulation of pre-mRNA splicing and mRNA localization by muscleblind proteins. Cell 150, 710–724 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Taliaferro, J. M. et al. Distal alternative last exons localize mRNAs to neural projections. Mol. Cell 61, 821–833 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Müller-McNicoll, M., Rossbach, O., Hui, J. & Medenbach, J. Auto-regulatory feedback by RNA-binding proteins. J. Mol. Cell Biol. 11, 930–939 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Konieczny, P., Stepniak-Konieczna, E. & Sobczak, K. MBNL expression in autoregulatory feedback loops. RNA Biol. 15, 1–8 (2018).

    Article  PubMed  Google Scholar 

  120. de Mezer, M., Wojciechowska, M., Napierala, M., Sobczak, K. & Krzyzosiak, W. J. Mutant CAG repeats of huntingtin transcript fold into hairpins, form nuclear foci and are targets for RNA interference. Nucleic Acids Res. 39, 3852–3863 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Mykowska, A., Sobczak, K., Wojciechowska, M., Kozlowski, P. & Krzyzosiak, W. J. CAG repeats mimic CUG repeats in the misregulation of alternative splicing. Nucleic Acids Res. 39, 8938–8951 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Kino, Y. et al. Nuclear localization of MBNL1: splicing-mediated autoregulation and repression of repeat-derived aberrant proteins. Hum. Mol. Genet. 24, 740–756 (2015).

    Article  CAS  PubMed  Google Scholar 

  124. 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 

  125. Thornton, C. A., Johnson, K. & Moxley, R. T. Myotonic dystrophy patients have larger CTG expansions in skeletal muscle than in leukocytes. Ann. Neurol. 35, 104–107 (1994).

    Article  CAS  PubMed  Google Scholar 

  126. Otero, B. A. et al. Transcriptome alterations in myotonic dystrophy frontal cortex. Cell Rep. 34, (2021).

  127. Sellier, C. et al. rbFOX1/MBNL1 competition for CCUG RNA repeats binding contributes to myotonic dystrophy type 1/type 2 differences. Nat. Commun. 9, 2009 (2018). RBFOX proteins compete with MBNL proteins for binding sites on CCUG repeat RNAs, offering a novel mechanism by which RBP sequestration and toxicity can be attenuated through competition between proteins.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Carrell, S. T. et al. Dmpk gene deletion or antisense knockdown does not compromise cardiac or skeletal muscle function in mice. Hum. Mol. Genet. 25, 4328–4338 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Hsu, T.-C. et al. Deactivation of TBP contributes to SCA17 pathogenesis. Hum. Mol. Genet. 23, 6878–6893 (2014).

    Article  PubMed  CAS  Google Scholar 

  130. Lieberman, A. P., Shakkottai, V. G. & Albin, R. L. Polyglutamine repeats in neurodegenerative diseases. Annu. Rev. Pathol. 14, 1–27 (2019).

    Article  CAS  PubMed  Google Scholar 

  131. Bäuerlein, F. J. B. et al. In situ architecture and cellular interactions of polyQ inclusions. Cell 171, 179–187.e10 (2017).

    Article  PubMed  CAS  Google Scholar 

  132. Peskett, T. R. et al. A liquid to solid phase transition underlying pathological huntingtin exon1 aggregation. Mol. Cell 70, 588–601.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Scherzinger, E. et al. Huntingtin-encoded polyglutamine expansions form amyloid-like protein aggregates in vitro and in vivo. Cell 90, 549–558 (1997).

    Article  CAS  PubMed  Google Scholar 

  134. Paulson, H. L. et al. Intranuclear inclusions of expanded polyglutamine protein in spinocerebellar ataxia type 3. Neuron 19, 333–344 (1997).

    Article  CAS  PubMed  Google Scholar 

  135. Ordway, J. M. et al. Ectopically expressed CAG repeats cause intranuclear inclusions and a progressive late onset neurological phenotype in the mouse. Cell 91, 753–763 (1997).

    Article  CAS  PubMed  Google Scholar 

  136. Mangiarini, L. et al. Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 87, 493–506 (1996).

    Article  CAS  PubMed  Google Scholar 

  137. Schilling, G. et al. Intranuclear inclusions and neuritic aggregates in transgenic mice expressing a mutant N-terminal fragment of huntingtin. Hum. Mol. Genet. 8, 397–407 (1999).

    Article  CAS  PubMed  Google Scholar 

  138. Yamamoto, A., Lucas, J. J. & Hen, R. Reversal of neuropathology and motor dysfunction in a conditional model of Huntington’s disease. Cell 101, 57–66 (2000).

    Article  CAS  PubMed  Google Scholar 

  139. Riley, B. E. & Orr, H. T. Polyglutamine neurodegenerative diseases and regulation of transcription: assembling the puzzle. Genes Dev. 20, 2183–2192 (2006).

    Article  CAS  PubMed  Google Scholar 

  140. Katsuno, M. et al. Reversible disruption of dynactin 1-mediated retrograde axonal transport in polyglutamine-induced motor neuron degeneration. J. Neurosci. 26, 12106–12117 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Katsuno, M. et al. Testosterone reduction prevents phenotypic expression in a transgenic mouse model of spinal and bulbar muscular atrophy. Neuron 35, 843–854 (2002).

    Article  CAS  PubMed  Google Scholar 

  142. Montie, H. L. et al. Cytoplasmic retention of polyglutamine-expanded androgen receptor ameliorates disease via autophagy in a mouse model of spinal and bulbar muscular atrophy. Hum. Mol. Genet. 18, 1937–1950 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Palazzolo, I. et al. Akt blocks ligand binding and protects against expanded polyglutamine androgen receptor toxicity. Hum. Mol. Genet. 16, 1593–1603 (2007).

    Article  CAS  PubMed  Google Scholar 

  144. Irwin, S. et al. RNA association and nucleocytoplasmic shuttling by ataxin-1. J. Cell Sci. 118, 233–242 (2005).

    Article  CAS  PubMed  Google Scholar 

  145. Lam, Y. C. et al. ATAXIN-1 interacts with the repressor Capicua in its native complex to cause SCA1 neuropathology. Cell 127, 1335–1347 (2006).

    Article  CAS  PubMed  Google Scholar 

  146. Klement, I. A. et al. Ataxin-1 nuclear localization and aggregation: role in polyglutamine-induced disease in SCA1 transgenic mice. Cell 95, 41–53 (1998).

    Article  CAS  PubMed  Google Scholar 

  147. Lai, S., O’Callaghan, B., Zoghbi, H. Y. & Orr, H. T. 14-3-3 binding to ataxin-1(ATXN1) regulates its dephosphorylation at Ser-776 and transport to the nucleus. J. Biol. Chem. 286, 34606–34616 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Emamian, E. S. et al. Serine 776 of ataxin-1 is Critical for polyglutamine-induced disease in SCA1 transgenic mice. Neuron 38, 375–387 (2003).

    Article  CAS  PubMed  Google Scholar 

  149. Duvick, L. et al. SCA1-like disease in mice expressing wild-type ataxin-1 with a serine to aspartic acid replacement at residue 776. Neuron 67, 929–935 (2010). Altering the phosphorylation state of ATXN1is sufficient to elicit toxicity even in the absence of polyQ expansion, confirming that aberrant native-protein function has a role in pathogenesis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Williams, A. J. & Paulson, H. L. Polyglutamine neurodegeneration: protein misfolding revisited. Trends Neurosci. 31, 521–528 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Klockgether, T., Mariotti, C. & Paulson, H. L. Spinocerebellar ataxia. Nat. Rev. Dis. Prim. 5, 24 (2019).

    Article  PubMed  Google Scholar 

  152. Zu, T. et al. Non-ATG-initiated translation directed by microsatellite expansions. Proc. Natl Acad. Sci. USA 108, 260–265 (2011). Repeat RNAs can be translated in the absence of an AUG start codon in many repeat expansion disorders.

    Article  CAS  PubMed  Google Scholar 

  153. 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 

  154. Ash, P. E. A. et al. Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron 77, 639–646 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. 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 

  156. Bañez-Coronel, M. et al. RAN translation in Huntington disease. Neuron 88, 667–677 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Zu, T. et al. RAN translation regulated by muscleblind proteins in myotonic dystrophy type 2. Neuron 95, 1292–1305.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Soragni, E. et al. Repeat-associated non-ATG (RAN) translation in Fuchs’ endothelial corneal dystrophy. Invest. Ophthalmol. Vis. Sci. 59, 1888–1896 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Ishiguro, T. et al. Regulatory role of RNA chaperone TDP-43 for RNA misfolding and repeat-associated translation in SCA31. Neuron 94, 108–124.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. 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 

  161. Krans, A., Kearse, M. G. & Todd, P. K. Repeat-associated non-AUG translation from antisense CCG repeats in fragile X tremor/ataxia syndrome. Ann. Neurol. 80, 871–881 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Jackson, R. J., Hellen, C. U. T. & Pestova, T. V. The mechanism of eukaryotic translation initiation and principles of its regulation. Nat. Rev. Mol. Cell Biol. 11, 113–127 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Kearse, M. G. et al. CGG repeat-associated non-AUG translation utilizes a cap-dependent scanning mechanism of initiation to produce toxic proteins. Mol. Cell 62, 314–322 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Sellier, C. et al. Translation of expanded CGG repeats into FMRpolyG is pathogenic and may contribute to fragile X tremor ataxia syndrome. Neuron 93, 331–347 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Linsalata, A. E. et al. DDX3X and specific initiation factors modulate FMR1 repeat-associated non-AUG-initiated translation. EMBO Rep. 20, e47498 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  166. Kearse, M. G. & Wilusz, J. E. Non-AUG translation: a new start for protein synthesis in eukaryotes. Genes Dev. 31, 1717–1731 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Green, K. M. et al. RAN translation at C9orf72-associated repeat expansions is selectively enhanced by the integrated stress response. Nat. Commun. 8, 2005 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  168. Tabet, R. et al. CUG initiation and frameshifting enable production of dipeptide repeat proteins from ALS/FTD C9ORF72 transcripts. Nat. Commun. 9, 152 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  169. Sonobe, Y. et al. Translation of dipeptide repeat proteins from the C9ORF72 expanded repeat is associated with cellular stress. Neurobiol. Dis. 116, 155–165 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Westergard, T. et al. Repeat-associated non-AUG translation in C9orf72-ALS/FTD is driven by neuronal excitation and stress. EMBO Mol. Med. 11, (2019).

  171. Cheng, W. et al. C9ORF72 GGGGCC repeat-associated non-AUG translation is upregulated by stress through eIF2α phosphorylation. Nat. Commun. 9, 51 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  172. Kwan, T. & Thompson, S. R. Noncanonical translation initiation in éukaryotes. Cold Spring Harb. Perspect. Biol. 11, (2019).

  173. Yamada, S. B. et al. RPS25 is required for efficient RAN translation of C9orf72 and other neurodegenerative disease-associated nucleotide repeats. Nat. Neurosci. 22, 1383–1388 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. 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 

  175. 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 

  176. Mizielinska, S. et al. C9orf72 repeat expansions cause neurodegeneration in Drosophila through arginine-rich proteins. Science 345, 1192–1194 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Oh, S. Y. et al. RAN translation at CGG repeats induces ubiquitin proteasome system impairment in models of fragile X-associated tremor ataxia syndrome. Hum. Mol. Genet. 24, 4317–4326 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Hukema, R. K. et al. Reversibility of neuropathology and motor deficits in an inducible mouse model for FXTAS. Hum. Mol. Genet. 24, 4948–4957 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Castro, H. et al. Selective rescue of heightened anxiety but not gait ataxia in a premutation 90CGG mouse model of Fragile X-associated tremor/ataxia syndrome. Hum. Mol. Genet. 26, 2133–2145 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Rodriguez, C. M. et al. A native function for RAN translation and CGG repeats in regulating fragile X protein synthesis. Nat. Neurosci. 23, 386–397 (2020). Selective targeting of RAN translation initiation with non-cleaving ASOs suppresses repeat toxicity while boosting FMRP production in human neurons, establishing a native role for RAN translation in regulation of neuronal protein synthesis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Jovičić, A. et al. Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. Nat. Neurosci. 18, 1226–1229 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Lee, K.-H. et al. C9orf72 dipeptide repeats impair the assembly, dynamics, and function of membrane-less organelles. Cell 167, 774–788.e17 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. 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 

  184. 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 

  185. 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 

  186. 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 

  187. Kanekura, K. et al. Poly-dipeptides encoded by the C9ORF72 repeats block global protein translation. Hum. Mol. Genet. 25, 1803–1813 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Boeynaems, S. et al. Drosophila screen connects nuclear transport genes to DPR pathology in c9ALS/FTD. Sci. Rep. 6, 20877 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Zhang, Y.-J. et al. Poly(GR) impairs protein translation and stress granule dynamics in C9orf72-associated frontotemporal dementia and amyotrophic lateral sclerosis. Nat. Med. 24, 1136–1142 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Hao, Z. et al. Motor dysfunction and neurodegeneration in a C9orf72 mouse line expressing poly-PR. Nat. Commun. 10, 2906 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  191. Zhang, Y.-J. et al. Heterochromatin anomalies and double-stranded RNA accumulation underlie C9orf72 poly(PR) toxicity. Science 363, eaav2606 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Flores, B. N. et al. Distinct C9orf72-associated dipeptide repeat structures correlate with neuronal toxicity. PLoS ONE 11, e0165084 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  193. Ohki, Y. et al. Glycine-alanine dipeptide repeat protein contributes to toxicity in a zebrafish model of C9orf72 associated neurodegeneration. Mol. Neurodegener. 12, 6 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  194. Guo, Q. et al. In situ structure of neuronal C9orf72 poly-GA aggregates reveals proteasome recruitment. Cell 172, 696–705.e12 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Freibaum, B. D. & Taylor, J. P. The role of dipeptide repeats in C9ORF72-related ALS-FTD. Front. Mol. Neurosci. 10, 35 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  196. Nguyen, L., Cleary, J. D. & Ranum, L. P. W. Repeat-associated non-ATG translation: molecular mechanisms and contribution to neurological disease. Annu. Rev. Neurosci. 42, 227–247 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Odeh, H. M. & Shorter, J. Arginine-rich dipeptide-repeat proteins as phase disruptors in C9-ALS/FTD. Emerg. Top. Life Sci. 4, 293–305 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Mackenzie, I. R. A. et al. Quantitative analysis and clinico-pathological correlations of different dipeptide repeat protein pathologies in C9ORF72 mutation carriers. Acta Neuropathol. 130, 845–861 (2015).

    Article  CAS  PubMed  Google Scholar 

  199. Davidson, Y. S. et al. Brain distribution of dipeptide repeat proteins in frontotemporal lobar degeneration and motor neurone disease associated with expansions in C9ORF72. Acta Neuropathol. Commun. 2, 70 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  200. Davidson, Y. et al. Neurodegeneration in frontotemporal lobar degeneration and motor neurone disease associated with expansions in C9orf72 is linked to TDP-43 pathology and not associated with aggregated forms of dipeptide repeat proteins. Neuropathol. Appl. Neurobiol. 42, 242–254 (2016).

    Article  CAS  PubMed  Google Scholar 

  201. Quaegebeur, A., Glaria, I., Lashley, T. & Isaacs, A. M. Soluble and insoluble dipeptide repeat protein measurements in C9orf72-frontotemporal dementia brains show regional differential solubility and correlation of poly-GR with clinical severity. Acta Neuropathol. Commun. 8, 184 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Goodman, L. D. et al. eIF4B and eIF4H mediate GR production from expanded G4C2 in a Drosophila model for C9orf72-associated ALS. Acta Neuropathol. Commun. 7, 62 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  203. Ayhan, F. et al. SCA8 RAN polySer protein preferentially accumulates in white matter regions and is regulated by eIF3F. EMBO J. 37, e99023 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Cheng, W. et al. CRISPR-Cas9 screens identify the RNA helicase DDX3X as a repressor of C9ORF72 (GGGGCC)n repeat-associated non-AUG translation. Neuron 104, 885–898.e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Harding, H. P. et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol. Cell 11, 619–633 (2003).

    Article  CAS  PubMed  Google Scholar 

  206. Pakos-Zebrucka, K. et al. The integrated stress response. EMBO Rep. 17, 1374–1395 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Harding, H. P. et al. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol. Cell 6, 1099–1108 (2000).

    Article  CAS  PubMed  Google Scholar 

  208. Zu, T. et al. Metformin inhibits RAN translation through PKR pathway and mitigates disease in C9orf72 ALS/FTD mice. Proc. Natl Acad. Sci. USA 117, 18591–18599 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Tiscornia, G. & Mahadevan, M. S. Myotonic dystrophy: the role of the CUG triplet repeats in splicing of a novel DMPK exon and altered cytoplasmic DMPK mRNA isoform ratios. Mol. Cell 5, 959–967 (2000).

    Article  CAS  PubMed  Google Scholar 

  210. Sznajder, Ł. J. et al. Intron retention induced by microsatellite expansions as a disease biomarker. Proc. Natl Acad. Sci. USA 115, 4234–4239 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Sirp, A. et al. The Fuchs corneal dystrophy-associated CTG repeat expansion in the TCF4 gene affects transcription from its alternative promoters. Sci. Rep. 10, 18424 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  213. Neueder, A. et al. The pathogenic exon 1 HTT protein is produced by incomplete splicing in Huntington’s disease patients. Sci. Rep. 7, 1307 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  214. Shi, Y. et al. Haploinsufficiency leads to neurodegeneration in C9ORF72 ALS/FTD human induced motor neurons. Nat. Med. 24, 313–325 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Shao, Q. et al. C9orf72 deficiency promotes motor deficits of a C9ALS/FTD mouse model in a dose-dependent manner. Acta Neuropathol. Commun. 7, 32 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  216. Zhu, Q. et al. Reduced C9ORF72 function exacerbates gain of toxicity from ALS/FTD-causing repeat expansion in C9orf72. Nat. Neurosci. 23, 615–624 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Gray, M. et al. Full-length human mutant huntingtin with a stable polyglutamine repeat can elicit progressive and selective neuropathogenesis in BACHD mice. J. Neurosci. Off. J. Soc. Neurosci. 28, 6182–6195 (2008).

    Article  CAS  Google Scholar 

  218. Li, L.-B., Yu, Z., Teng, X. & Bonini, N. M. RNA toxicity is a component of ataxin-3 degeneration in Drosophila. Nature 453, 1107–1111 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Rué, L. et al. Targeting CAG repeat RNAs reduces Huntington’s disease phenotype independently of huntingtin levels. J. Clin. Invest. 126, 4319–4330 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  220. Wang, Q., Conlon, E. G., Manley, J. L. & Rio, D. C. Widespread intron retention impairs protein homeostasis in C9orf72 ALS brains. Genome Res. 30, 1705–1715 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Hautbergue, G. M. et al. SRSF1-dependent nuclear export inhibition of C9ORF72 repeat transcripts prevents neurodegeneration and associated motor deficits. Nat. Commun. 8, 16063 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Cleary, J. D., Pattamatta, A. & Ranum, L. P. W. Repeat-associated non-ATG (RAN) translation. J. Biol. Chem. 293, 16127–16141 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. McEachin, Z. T. et al. Chimeric peptide species contribute to divergent dipeptide repeat pathology in c9ALS/FTD and SCA36. Neuron 107, 292–305.e6 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. 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 

  225. Wills, N. M. & Atkins, J. F. The potential role of ribosomal frameshifting in generating aberrant proteins implicated in neurodegenerative diseases. RNA 12, 1149–1153 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  227. Saffert, P., Adamla, F., Schieweck, R., Atkins, J. F. & Ignatova, Z. An expanded CAG repeat in huntingtin causes +1 frameshifting. J. Biol. Chem. 291, 18505–18513 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Ma, L. et al. Composition of the intranuclear inclusions of fragile X-associated tremor/ataxia syndrome. Acta Neuropathol. Commun. 7, 143 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  229. Haify, S. N. et al. Lack of a clear behavioral phenotype in an inducible FXTAS mouse model despite the presence of neuronal FMRpolyG-positive aggregates. Front. Mol. Biosci. 7, 599101 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Asamitsu, S. et al. CGG repeat RNA G-quadruplexes interact with FMRpolyG to cause neuronal dysfunction in fragile X-related tremor/ataxia syndrome. Sci. Adv. 7, eabd9440 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Heatwole, C. et al. Patient-reported impact of symptoms in myotonic dystrophy type 1 (PRISM-1). Neurology 79, 348 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  232. Ashizawa, T., Dubel, J. R. & Harati, Y. Somatic instability of CTG repeat in myotonic dystrophy. Neurology 43, 2674–2678 (1993).

    Article  CAS  PubMed  Google Scholar 

  233. Sznajder, Ł. J. et al. Loss of MBNL1 induces RNA misprocessing in the thymus and peripheral blood. Nat. Commun. 11, 2022 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Ishiura, H. et al. Expansions of intronic TTTCA and TTTTA repeats in benign adult familial myoclonic epilepsy. Nat. Genet. 50, 581–590 (2018).

    Article  CAS  PubMed  Google Scholar 

  235. Corbett, M. A. et al. Intronic ATTTC repeat expansions in STARD7 in familial adult myoclonic epilepsy linked to chromosome 2. Nat. Commun. 10, 4920 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  236. Florian, R. T. et al. Unstable TTTTA/TTTCA expansions in MARCH6 are associated with familial adult myoclonic epilepsy type 3. Nat. Commun. 10, 4919 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  237. Yeetong, P. et al. TTTCA repeat insertions in an intron of YEATS2 in benign adult familial myoclonic epilepsy type 4. Brain J. Neurol. 142, 3360–3366 (2019).

    Article  Google Scholar 

  238. Seixas, A. I. et al. A pentanucleotide ATTTC repeat insertion in the non-coding region of DAB1, mapping to SCA37, causes spinocerebellar ataxia. Am. J. Hum. Genet. 101, 87–103 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Saudou, F. & Humbert, S. The biology of huntingtin. Neuron 89, 910–926 (2016).

    Article  CAS  PubMed  Google Scholar 

  240. Rüb, U. et al. Degeneration of the cerebellum in Huntington’s disease (HD): possible relevance for the clinical picture and potential gateway to pathological mechanisms of the disease process. Brain Pathol. 23, 165–177 (2013).

    Article  PubMed  CAS  Google Scholar 

  241. Shortt, J. A., Ruggiero, R. P., Cox, C., Wacholder, A. C. & Pollock, D. D. Finding and extending ancient simple sequence repeat-derived regions in the human genome. Mob. DNA 11, 11 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Pheasant, M. & Mattick, J. S. Raising the estimate of functional human sequences. Genome Res. 17, 1245–1253 (2007).

    Article  CAS  PubMed  Google Scholar 

  243. Elden, A. C. et al. Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 466, 1069–1075 (2010). Ataxin 2 can modify TDP43 toxicity in yeast and flies, and intermediate repeats in ataxinК2 serve as a common risk allele in ALS.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Tazelaar, G. H. P. et al. ATXN1 repeat expansions confer risk for amyotrophic lateral sclerosis and contribute to TDP-43 mislocalization. Brain Commun. 2, fcaa064 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  245. Conforti, F. L. et al. Ataxin-1 and ataxin-2 intermediate-length polyQ expansions in amyotrophic lateral sclerosis. Neurology 79, 2315–2320 (2012).

    Article  CAS  PubMed  Google Scholar 

  246. Lattante, S. et al. ATXN1 intermediate-length polyglutamine expansions are associated with amyotrophic lateral sclerosis. Neurobiol. Aging 64, 157.e1–157.e5 (2018).

    Article  CAS  Google Scholar 

  247. R, D. et al. Pathogenic huntingtin repeat expansions in patients with frontotemporal dementia and amyotrophic lateral sclerosis. Neuron 109, 448–460 (2021).

    Article  CAS  Google Scholar 

  248. Blauw, H. M. et al. NIPA1 polyalanine repeat expansions are associated with amyotrophic lateral sclerosis. Hum. Mol. Genet. 21, 2497–2502 (2012).

    Article  CAS  PubMed  Google Scholar 

  249. Course, M. M. et al. Evolution of a human-specific tandem repeat associated with ALS. Am. J. Hum. Genet. 107, 445–460 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Yuan, Y. et al. Identification of GGC repeat expansion in the NOTCH2NLC gene in amyotrophic lateral sclerosis. Neurology 95, e3394–e3405 (2020).

    Article  CAS  PubMed  Google Scholar 

  251. Willems, T. et al. Genome-wide profiling of heritable and de novo STR variations. Nat. Methods 14, 590–592 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Trost, B. et al. Genome-wide detection of tandem DNA repeats that are expanded in autism. Nature 586, 80–86 (2020). Repeat expansions are a common feature in autism genomes and occur at a much higher frequency than previously appreciated.

    Article  CAS  PubMed  Google Scholar 

  253. Mitra, I. et al. Patterns of de novo tandem repeat mutations and their role in autism. Nature 589, 246–250 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Jansen, A., Gemayel, R. & Verstrepen, K. J. Unstable microsatellite repeats facilitate rapid evolution of coding and regulatory sequences. Genome Dyn. 7, 108–125 (2012).

    Article  CAS  PubMed  Google Scholar 

  255. Vinces, M. D., Legendre, M., Caldara, M., Hagihara, M. & Verstrepen, K. J. Unstable tandem repeats in promoters confer transcriptional evolvability. Science 324, 1213–1216 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Verstrepen, K. J., Jansen, A., Lewitter, F. & Fink, G. R. Intragenic tandem repeats generate functional variability. Nat. Genet. 37, 986–990 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Caron, N. S., Desmond, C. R., Xia, J. & Truant, R. Polyglutamine domain flexibility mediates the proximity between flanking sequences in huntingtin. Proc. Natl. Acad. Sci. USA 110, 14610–14615 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Ashkenazi, A. et al. Polyglutamine tracts regulate beclin 1-dependent autophagy. Nature 545, 108–111 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Liquori, C. L. et al. Myotonic dystrophy type 2 caused by a CCTG expansion in intron 1 of ZNF9. Science 293, 864–867 (2001).

    Article  CAS  PubMed  Google Scholar 

  260. Ranum, L. P. W. & Day, J. W. Myotonic dystrophy: clinical and molecular parallels between myotonic dystrophy type 1 and type 2. Curr. Neurol. Neurosci. Rep. 2, 465–470 (2002).

    Article  PubMed  Google Scholar 

  261. Kim, G., Gautier, O., Tassoni-Tsuchida, E., Ma, X. R. & Gitler, A. D. ALS genetics: gains, losses, and implications for future therapies. Neuron 108, 822–842 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Ishiura, H. et al. Noncoding CGG repeat expansions in neuronal intranuclear inclusion disease, oculopharyngodistal myopathy and an overlapping disease. Nat. Genet. 51, 1222–1232 (2019).

    Article  CAS  PubMed  Google Scholar 

  263. Sone, J. et al. Long-read sequencing identifies GGC repeat expansions in NOTCH2NLC associated with neuronal intranuclear inclusion disease. Nat. Genet. 51, 1215–1221 (2019).

    Article  CAS  PubMed  Google Scholar 

  264. Tian, Y. et al. Expansion of human-specific GGC repeat in neuronal intranuclear inclusion disease-related disorders. Am. J. Hum. Genet. 105, 166–176 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. Gelpi, E. et al. Neuronal intranuclear (hyaline) inclusion disease and fragile X-associated tremor/ataxia syndrome: a morphological and molecular dilemma. Brain J. Neurol. 140, e51 (2017).

    Article  Google Scholar 

  266. Boivin, M. et al. Translation of GGC repeat expansions into a toxic polyglycine protein in NIID defines a novel class of human genetic disorders: the polyG diseases. Neuron https://doi.org/10.1016/j.neuron.2021.03.038 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  267. Cortese, A. et al. Biallelic expansion of an intronic repeat in RFC1 is a common cause of late-onset ataxia. Nat. Genet. 51, 649–658 (2019). A new recessively inherited repeat expansion, whose generation and pathogenesis mechanisms remain a mystery, is discovered.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  268. Rafehi, H. et al. Bioinformatics-based identification of expanded repeats: a non-reference intronic pentamer expansion in RFC1 causes CANVAS. Am. J. Hum. Genet. 105, 151–165 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Scriba, C. K. et al. A novel RFC1 repeat motif (ACAGG) in two Asia-Pacific CANVAS families. Brain J. Neurol. 143, 2904–2910 (2020).

    Article  Google Scholar 

  270. Tsuchiya, M. et al. RFC1 repeat expansion in Japanese patients with late-onset cerebellar ataxia. J. Hum. Genet. 65, 1143–1147 (2020).

    Article  CAS  PubMed  Google Scholar 

  271. Beecroft, S. J. et al. A Māori specific RFC1 pathogenic repeat configuration in CANVAS, likely due to a founder allele. Brain J. Neurol. 143, 2673–2680 (2020).

    Article  Google Scholar 

  272. Jones, C. et al. Association of a chromosome deletion syndrome with a fragile site within the proto-oncogene CBL2. Nature 376, 145–149 (1995).

    Article  CAS  PubMed  Google Scholar 

  273. Parodi, S. et al. Parental origin and somatic mosaicism of PHOX2B mutations in congenital central hypoventilation syndrome. Hum. Mutat. 29, 206–206 (2008).

    Article  PubMed  Google Scholar 

  274. Owens, K. M. et al. Analysis of de novo HOXA13 polyalanine expansions supports replication slippage without repair in their generation. Am. J. Med. Genet. A. 161, 1019–1027 (2013).

    Article  CAS  Google Scholar 

  275. Brown, L. Y. et al. Holoprosencephaly due to mutations in ZIC2: alanine tract expansion mutations may be caused by parental somatic recombination. Hum. Mol. Genet. 10, 791–796 (2001).

    Article  CAS  PubMed  Google Scholar 

  276. Poirier, K. et al. Maternal mosaicism for mutations in the ARX gene in a family with X linked mental retardation. Hum. Genet. 118, 45–48 (2005).

    Article  CAS  PubMed  Google Scholar 

  277. Friocourt, G. & Parnavelas, J. G. Mutations in ARX result in several defects involving GABAergic neurons. Front. Cell. Neurosci. 4, (2010).

Download references

Acknowledgements

This work was supported by US National Institutes of Health grants NS099280, NS086810 and P50HD104463 and VA grant BLRD BX004842 to P.K.T., and US National Institutes of Health grants AG058636, R01NS112291 and R01NS114253 to E.T.W. I.M. was supported by the Alzheimer’s Association Research Fellowship (AARF-20-684648). C.P.K. is supported by the US National Science Foundation Graduate Research Fellowship Program.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to Eric T. Wang or Peter K. Todd.

Ethics declarations

Competing interests

P.K.T. holds a shared patent with Ionis Pharmaceuticals on ASO technologies for use against RAN translation. He also serves as a paid consultant for Denali Therapeutics, holds stock options in this company and has received licensing fees for use of experimental tools developed by his research group. The other authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Molecular Cell Biology thanks Clotilde Lagier-Tourenne and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

Non-canonical translation

Translation initiation that does not require one or more canonical RNA features, such as a 5′ cap or an AUG start codon.

Somatic instability

DNA repair-mediated differences in the number of short tandem repeats between cells of an individual which develop over their lifetime.

Ribonucleoprotein

(RNP). A transient or stable complex formed by multivalent interaction of proteins and RNAs.

Repeat-associated non-AUG (RAN) translation

A form of non-canonical translation initiated at an expanded-repeat RNA in the absence of an AUG start codon.

Transcriptional gene silencing

The process of inhibiting RNA polymerase-mediated RNA synthesis, which results in abolition of or partial decrease in RNA production.

R-loop

A three-stranded RNA–DNA hybrid structure containing two strands of DNA and one strand of RNA, the latter of which forms Watson–Crick base pairs with the complementary DNA strand.

Rare fragile site

Refers to rare heritable expanded repeat loci that tend to break when cells are exposed to replication stress, such as growth in a folate-free medium.

DNA mismatch repair

(MMR). Repair pathway of erroneous insertions, deletions and base misincorporations during DNA replication and transcription. MMR has a role in resolving R-loops and repairing DNA damage caused by oxidative stress.

RNA foci

RNA clusters formed by accumulation of expanded-repeat RNAs or ribonucleoprotein complexes, primarily in the nucleus and sometimes in the cytoplasm.

Secondary structures

The patterns of intramolecular base pairing of nucleotides within an RNA molecule.

G-quadruplexes

Stable nucleic acid secondary structures formed by the stacking of several planar guanine quadruplets.

A-form double helix

A right-handed, compact helical structure formed by RNA–DNA and RNA–RNA duplexes.

Phase separation

The thermodynamic process by which a homogeneous mixture of components separates spontaneously into different coexisting phases. In aqueous solutions of biomolecules, phase separation is often driven by intrinsically disordered proteins and/or multivalent interactions between proteins and RNAs.

Phase diagram

A graphical depiction of the phases accessible to a system as a function of multiple thermodynamic properties.

Near-cognate start codons

Also known as near-AUG codons, these codons differ from the canonical AUG start codon by a single base, for example, a CUG codon.

Upstream open reading frames

(uORFs). Small open reading frames in the 5′ untranslated region of an mRNA capable of undergoing translation.

Internal ribosomal entry site

(IRES). A structured RNA element that allows non-canonical translation initiation from within the mRNA without requiring a 5′ cap.

Stress granules

Membraneless ribonucleoprotein compartments that appear in the cytosol in response to various cellular stresses.

Frameshifting

Shifts or slips of translating ribosomes along the mRNA which change the open reading frame and the sequence of the translated protein.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Malik, I., Kelley, C.P., Wang, E.T. et al. Molecular mechanisms underlying nucleotide repeat expansion disorders. Nat Rev Mol Cell Biol 22, 589–607 (2021). https://doi.org/10.1038/s41580-021-00382-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-021-00382-6

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