Elsevier

The Lancet Neurology

Volume 16, Issue 1, January 2017, Pages 88-96
The Lancet Neurology

Rapid Review
DNA repair in the trinucleotide repeat disorders

https://doi.org/10.1016/S1474-4422(16)30350-7Get rights and content

Summary

Background

Inherited diseases caused by unstable repeated DNA sequences are rare, but together represent a substantial cause of morbidity. Trinucleotide repeat disorders are severe, usually life-shortening, neurological disorders caused by nucleotide expansions, and most have no disease-modifying treatments. Longer repeat expansions are associated with genetic anticipation (ie, earlier disease onset in successive generations), although the differences in age at onset are not entirely accounted for by repeat length. Such phenotypic variation within disorders implies the existence of additional modifying factors in pathways that can potentially be modulated to treat disease.

Recent developments

A genome-wide association study detected genetic modifiers of age at onset in Huntington's disease. Similar findings were seen in the spinocerebellar ataxias, indicating an association between DNA damage-response and repair pathways and the age at onset of disease. These studies also suggest that a common genetic mechanism modulates age at onset across polyglutamine diseases and could extend to other repeat expansion disorders. Genetic defects in DNA repair underlie other neurodegenerative disorders (eg, ataxia-telangiectasia), and DNA double-strand breaks are crucial to the modulation of early gene expression, which provides a mechanistic link between DNA repair and neurodegeneration. Mismatch and base-excision repair are important in the somatic expansion of repeated sequences in mouse models of trinucleotide repeat disorders, and somatic expansion of the expanded CAG tract in HTT correlates with age at onset of Huntington's disease and other trinucleotide repeat disorders.

Where next?

To understand the common genetic architecture of trinucleotide repeat disorders and any further genetic susceptibilities in individual disorders, genetic analysis with increased numbers of variants and sample sizes is needed, followed by sequencing approaches to define the phenotype-modifying variants. The findings must then be translated into cell biology analyses to elucidate the mechanisms through which the genetic variants operate. Genes that have roles in the DNA damage response could underpin a common DNA repeat-based mechanism and provide new therapeutic targets (and hence therapeutics) in multiple trinucleotide repeat disorders.

Introduction

Trinucleotide repeat disorders—inherited diseases caused by unstable repeated DNA sequences—were first characterised in the 1990s and are individually rare.1 Fragile X syndrome is the most common, with a prevalence of about one per 4000 boys. Myotonic dystrophy and Huntington's disease affect around one per 10 000 people, and most spinocerebellar ataxias affect around one per 100 000 people, although the prevalence of Huntington's disease and the spinocerebellar ataxias varies widely across geographical regions. Only a few cases have been identified for other such disorders. Together, however, trinucleotide repeat disorders represent a substantial source of morbidity. Most are life-shortening and have debilitating symptoms, and no disease-modifying treatments are available. Although the mutational mechanisms are similar, the repeated DNA sequences occur in different genomic contexts, and even in the polyglutamine diseases, where the repeated codon is translated to glutamine, the proteins are functionally unrelated. The nature and expression pattern of the repeat-containing proteins probably leads to the clinical differences between these diseases,1 but the substantial phenotypic variation seen within each disease remains only partly explained. This variability can be exploited to gain insights into disease mechanisms though genetics.2

The trinucleotide repeat disorders fall into two main categories: those in which the repeated sequence is translated into a protein product and those in which the repeat lies outside the coding sequence (table 1), but the non-coding disease-associated repeat sequences are usually longer than the coding sequences. All the trinucleotide repeat disorders are associated with genetic anticipation—earlier onset of disease in successive generations of families—caused by germline expansion of the repeat.16 Repeat expansion occurs in dividing and non-dividing cells, and is tissue specific, cell specific, and disease specific.1 Expansion of the repeat is ameliorated if the repeated sequence is interrupted by other codons.

Although associations between repeats and specific loci have been known since the 1990s, the mechanistic cascade from repeat to clinical phenotype remains unclear in trinucleotide repeat disorders, which has hindered the development of new treatments. Some pathogenic mechanisms are common to multiple diseases. For instance, a repeat that prevents gene expression is seen in fragile X syndrome and Friedreich's ataxia.17 Pathogenic RNA foci in myotonic dystrophy and myotonic dystrophy-like 2 give rise to characteristic splicing deficits,18 and have been noted in other trinucleotide repeat disorders, such as myotonic dystrophy 1 and 2.19 Repeat-associated non-ATG translation, which was first identified in myotonic dystrophy and spinocerebellar ataxia 8,20 has since been found in Huntington's disease,21 frontotemporal dementia and amyotrophic lateral sclerosis (which are caused by the C9ORF72 hexanucleotide repeat), and other trinucleotide repeat disorders.22 In frontotemporal dementia and amyotrophic lateral sclerosis, the C9ORF72 disease-associated repeat dipeptides are neurotoxic,23 although whether such dipeptides have any pathogenic role in trinucleotide repeat disorders is unknown.

Similar mechanisms might be seen in the polyglutamine (CAG repeat) diseases;22 however, the proteins that contain expanded polyglutamine tracts aggregate and form characteristic insoluble protein inclusions in neural and other cells in these disorders. Such insoluble inclusions are also widely seen in other neurodegenerative disorders,24 which has led to the hypothesis that they or their soluble oligomers are pathogenic. So far, the notion that preventing aggregation can prevent disease in human beings has not been proven.25 Nevertheless, some findings hint at this possibility. For example, in trials of aducanumab, an antibody that binds and reduces deposition of amyloid β, cognitive benefits were recorded in mouse models and in clinical studies of people early in the course of Alzheimer's disease.26 Ataxin 1 (ATXN1) oligomers drive the degenerative effects in spinocerebellar ataxia 1 and induce local spread of pathology.27 These effects were partly inhibited with immunotherapy.28 The biological consequences of expanded polyglutamine have been extensively studied and a wide range of potentially harmful outcomes detected,29, 30 but which of these are important in the manifestation of disease remains unclear. Genetic evidence indicates that the DNA damage response and DNA repair affect the clinical presentation of Huntington's disease and multiple spinocerebellar ataxias,31, 32, 33 which suggests that there are common modifiers that act on the mutated repeat itself. Together with evidence implicating these processes in trinucleotide repeat disorder biology, these findings shed light on specific mechanisms and highlight new targets for therapeutic intervention.

Section snippets

The DNA damage response and neurological disease

The DNA damage response (table 2) can be both harmful and protective in people with neurological diseases. Mutations in genes involved in the DNA damage response were first noted to cause neurological disease in ataxia-telangiectasia, a rare recessive childhood neurodegenerative disease. Mutations in the ataxia-telangiectasia mutant serine/threonine kinase gene (ATM) cause ataxia-telangiectasia. This gene controls cell-cycle arrest after DNA double-strand breaks, often leading to apoptosis and,

Genetic modifiers in the trinucleotide repeat disorders

One way of overcoming the difficulties of interpreting the cell biology findings is to return to the study of people carrying repeat expansions. In natural experiments,2 where conditions are not controlled by the researchers, it is possible to search for genetic loci that modify disease in a beneficial or deleterious way, and to reveal the biology that is likely to be relevant to disease manifestation. For instance, if genetic variation affects the timing of disease onset or the progression or

DNA repeat expansions and the DNA damage response

Repeat expansions in DNA are affected directly by activities of the DNA damage response (table 2).8 The repeats undergo expansion on transmission through the germline, in dividing and terminally differentiated somatic cells, and the repeat size increases with age.8 Strand breakage in the repeat is repaired, and it is at this point the repeat sequences are thought to expand.64, 68 The length of the repeat expansion in Huntington's disease correlates positively with the propensity for further

Conclusions and future directions

Showing that genetic modifiers exist in Mendelian trinucleotide repeat disorders demonstrates that finding genetic modifiers in rare genetic diseases is possible, and highlights areas of biology that modulate disease in people, such as specific aspects of the DNA damage response: mismatch repair, base-excision repair, and the Fanconi anaemia pathway. The findings raise two important questions. The first is whether genetic association with DNA repair processes occurs across all the trinucleotide

Search strategy and selection criteria

We searched PubMed for papers published from Jan 1, 2012, to Sept 30, 2016, using combinations of the terms “huntingt*”, “spinocerebellar ataxia”, “trinucleotide repeat”, “triplet repeat”, “repeat”, or “repeat disease”, and “DNA integrity”, “DNA repair”, “genome integrity”, or “genome repair”. Our search was limited to publications that included search terms in the title, abstract, or both. We identified further relevant papers by searching the reference lists of retrieved papers and through

References (99)

  • H Tran et al.

    Differential toxicity of nuclear RNA foci versus dipeptide repeat proteins in a Drosophila model of C9ORF72 FTD/ALS

    Neuron

    (2015)
  • J Bras et al.

    Mutations in PNKP cause recessive ataxia with oculomotor apraxia type 4

    Am J Hum Genet

    (2015)
  • M Weinfeld et al.

    Tidying up loose ends: the role of polynucleotide kinase/phosphatase in DNA strand break repair

    Trends Biochem Sci

    (2011)
  • E Hatchi et al.

    BRCA1 recruitment to transcriptional pause sites is required for R-loop-driven DNA damage repair

    Mol Cell

    (2015)
  • R Madabhushi et al.

    Activity-induced DNA breaks govern the expression of neuronal early-response genes

    Cell

    (2015)
  • H Jacobi et al.

    Long-term disease progression in spinocerebellar ataxia types 1, 2, 3, and 6: a longitudinal cohort study

    Lancet Neurol

    (2015)
  • V Ezzatizadeh et al.

    The mismatch repair system protects against intergenerational GAA repeat instability in a Friedreich ataxia mouse model

    Neurobiol Dis

    (2012)
  • F Morales et al.

    A polymorphism in the MSH3 mismatch repair gene is associated with the levels of somatic instability of the expanded CTG repeat in the blood DNA of myotonic dystrophy type 1 patients

    DNA Repair (Amst)

    (2016)
  • NJ Veitch et al.

    Inherited CAG·CTG allele length is a major modifier of somatic mutation length variability in Huntington disease

    DNA Repair (Amst)

    (2007)
  • KA Shah et al.

    The hidden side of unstable DNA repeats: Mutagenesis at a distance

    DNA Repair (Amst)

    (2015)
  • T Iyama et al.

    DNA repair mechanisms in dividing and non-dividing cells

    DNA Repair (Amst)

    (2013)
  • E Dragileva et al.

    Intergenerational and striatal CAG repeat instability in Huntington's disease knock-in mice involve different DNA repair genes

    Neurobiol Dis

    (2009)
  • H Xu et al.

    DNA repair/replication transcripts are down regulated in patients with fragile X syndrome

    BMC Res Notes

    (2013)
  • N Chatterjee et al.

    Mismatch repair enhances convergent transcription-induced cell death at trinucleotide repeats by activating ATR

    DNA Repair (Amst)

    (2016)
  • Y Liu et al.

    DNA base excision repair: a mechanism of trinucleotide repeat expansion

    Trends Biochem Sci

    (2012)
  • N Segui et al.

    Germline mutations in FAN1 cause hereditary colorectal cancer by impairing DNA repair

    Gastroenterology

    (2015)
  • S Pennell et al.

    FAN1 activity on asymmetric repair intermediates is mediated by an atypical monomeric virus-type replication-repair nuclease domain

    Cell Rep

    (2014)
  • D Viterbo et al.

    Replication stalling and heteroduplex formation within CAG/CTG trinucleotide repeats by mismatch repair

    DNA Repair (Amst)

    (2016)
  • HT Orr et al.

    Trinucleotide repeat disorders

    Annu Rev Neurosci

    (2007)
  • JF Gusella et al.

    Genetic modifiers of Huntington's disease

    Mov Disord

    (2014)
  • S Metsu et al.

    FRA2A is a CGG repeat expansion associated with silencing of AFF3

    PLoS Genet

    (2014)
  • RA Lokanga et al.

    Somatic expansion in mouse and human carriers of fragile X premutation alleles

    Hum Mutat

    (2013)
  • PK Todd et al.

    RNA-mediated neurodegeneration in repeat expansion disorders

    Ann Neurol

    (2010)
  • S Tezenas du Montcel et al.

    Modulation of the age at onset in spinocerebellar ataxia by CAG tracts in various genes

    Brain

    (2014)
  • V Evans-Galea M et al.

    Beyond loss of frataxin: the complex molecular pathology of Friedreich ataxia

    Discov Med

    (2014)
  • S Martins et al.

    Haplotype diversity and somatic instability in normal and expanded SCA8 alleles

    Am J Med Genet Part B Neuropsychiatr Genet

    (2005)
  • C Shoubridge et al.

    Polyalanine tract disorders and neurocognitive phenotypes

  • H Trang et al.

    Massive CAG repeat expansion and somatic instability in maternally transmitted infantile spinocerebellar ataxia type 7

    JAMA Neurol

    (2015)
  • S Nageshwaran et al.

    Epigenetics and triplet repeat neurological diseases

    Front Neurol

    (2015)
  • T Zu et al.

    Non-ATG-initiated translation directed by microsatellite expansions

    Proc Natl Acad Sci USA

    (2011)
  • TPJ Knowles et al.

    The amyloid state and its association with protein misfolding diseases

    Nat Rev Mol Cell Biol

    (2014)
  • JR Harrison et al.

    Alzheimer's disease: the amyloid hypothesis on trial

    Br J Psychiatry

    (2016)
  • J Sevigny et al.

    The antibody aducanumab reduces Abeta plaques in Alzheimer's disease

    Nature

    (2016)
  • CA Lasagna-Reeves et al.

    A native interactor scaffolds and stabilizes toxic ATAXIN-1 oligomers in SCA1

    Elife

    (2015)
  • CA Lasagna-Reeves et al.

    Ataxin-1 oligomers induce local spread of pathology and decreasing them by passive immunization slows spinocerebellar ataxia type 1 phenotypes

    Elife

    (2015)
  • GP Bates et al.

    Huntington disease

    Nat Rev Dis Prim

    (2015)
  • JL Guo et al.

    Cell-to-cell transmission of pathogenic proteins in neurodegenerative diseases

    Nat Med

    (2014)
  • Identification of genetic factors that modify clinical onset of Huntington's disease

    Cell

    (2015)
  • C Bettencourt et al.

    DNA repair pathways underlie a common genetic mechanism modulating onset in polyglutamine diseases

    Ann Neurol

    (2016)
  • Cited by (69)

    View all citing articles on Scopus
    View full text