Sei sulla pagina 1di 9

Rapid Review

DNA repair in the trinucleotide repeat disorders


Lesley Jones, Henry Houlden, Sarah J Tabrizi

Summary
Lancet Neurol 2017; 16: 8896 Background Inherited diseases caused by unstable repeated DNA sequences are rare, but together represent a
MRC Centre for substantial cause of morbidity. Trinucleotide repeat disorders are severe, usually life-shortening, neurological
Neuropsychiatric Genetics and disorders caused by nucleotide expansions, and most have no disease-modifying treatments. Longer repeat expansions
Genomics, Institute of
are associated with genetic anticipation (ie, earlier disease onset in successive generations), although the dierences
Psychological Medicine and
Clinical Neurosciences, Cardiff in age at onset are not entirely accounted for by repeat length. Such phenotypic variation within disorders implies the
University, Cardiff, UK existence of additional modifying factors in pathways that can potentially be modulated to treat disease.
(Prof L Jones PhD); Department
of Molecular Neuroscience and
Recent developments A genome-wide association study detected genetic modiers of age at onset in Huntingtons
MRC Centre for Neuromuscular
Diseases, Institute of disease. Similar ndings were seen in the spinocerebellar ataxias, indicating an association between DNA damage-
Neurology, Queen Square, response and repair pathways and the age at onset of disease. These studies also suggest that a common genetic
London, UK mechanism modulates age at onset across polyglutamine diseases and could extend to other repeat expansion
(Prof H Houlden MD); and UCL
Huntingtons Disease Centre,
disorders. Genetic defects in DNA repair underlie other neurodegenerative disorders (eg, ataxia-telangiectasia), and
Department of DNA double-strand breaks are crucial to the modulation of early gene expression, which provides a mechanistic link
Neurodegenerative Disease, between DNA repair and neurodegeneration. Mismatch and base-excision repair are important in the somatic
Institute of Neurology, expansion of repeated sequences in mouse models of trinucleotide repeat disorders, and somatic expansion of the
University College London,
London, UK (Prof S J Tabrizi PhD)
expanded CAG tract in HTT correlates with age at onset of Huntingtons disease and other trinucleotide repeat
disorders.
Correspondence to:
Prof Lesley Jones, MRC Centre for
Neuropsychiatric Genetics and Where next? To understand the common genetic architecture of trinucleotide repeat disorders and any further genetic
Genomics, Haydn Ellis Building, susceptibilities in individual disorders, genetic analysis with increased numbers of variants and sample sizes is
Maindy Road, Cardiff University,
Cardiff CF24 4HQ, UK
needed, followed by sequencing approaches to dene the phenotype-modifying variants. The ndings must then be
jonesl1@cf.ac.uk 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 The trinucleotide repeat disorders fall into two main


Trinucleotide repeat disordersinherited diseases categories: those in which the repeated sequence is
caused by unstable repeated DNA sequenceswere rst translated into a protein product and those in which the
characterised in the 1990s and are individually rare.1 repeat lies outside the coding sequence (table 1), but the
Fragile X syndrome is the most common, with a non-coding disease-associated repeat sequences are
prevalence of about one per 4000 boys. Myotonic usually longer than the coding sequences. All the
dystrophy and Huntingtons disease aect around one trinucleotide repeat disorders are associated with genetic
per 10 000 people, and most spinocerebellar ataxias aect anticipationearlier onset of disease in successive
around one per 100 000 people, although the prevalence generations of familiescaused by germline expansion
of Huntingtons disease and the spinocerebellar ataxias of the repeat.16 Repeat expansion occurs in dividing and
varies widely across geographical regions. Only a few non-dividing cells, and is tissue specic, cell specic, and
cases have been identied for other such disorders. disease specic.1 Expansion of the repeat is ameliorated if
Together, however, trinucleotide repeat disorders the repeated sequence is interrupted by other codons.
represent a substantial source of morbidity. Most are Although associations between repeats and specic loci
life-shortening and have debilitating symptoms, and no have been known since the 1990s, the mechanistic cascade
disease-modifying treatments are available. Although from repeat to clinical phenotype remains unclear in
the mutational mechanisms are similar, the repeated trinucleotide repeat disorders, which has hindered the
DNA sequences occur in dierent genomic contexts, development of new treatments. Some pathogenic
and even in the polyglutamine diseases, where the mechanisms are common to multiple diseases. For
repeated codon is translated to glutamine, the proteins instance, a repeat that prevents gene expression is seen in
are functionally unrelated. The nature and expression fragile X syndrome and Friedreichs ataxia.17 Pathogenic
pattern of the repeat-containing proteins probably leads RNA foci in myotonic dystrophy and myotonic
to the clinical dierences between these diseases,1 but dystrophy-like 2 give rise to characteristic splicing decits,18
the substantial phenotypic variation seen within each and have been noted in other trinucleotide repeat
disease remains only partly explained. This variability disorders, such as myotonic dystrophy 1 and 2.19 Repeat-
can be exploited to gain insights into disease associated non-ATG translation, which was rst identied
mechanisms though genetics.2 in myotonic dystrophy and spinocerebellar ataxia 8,20 has

88 www.thelancet.com/neurology Vol 16 January 2017


Rapid Review

Gene Repeated Non-expanded Expanded (bp) Somatic expansion Comments


sequence (bp)

5 UTR
Fragile X syndrome3,4 FMR1 CGG <50 >200 Yes (M, H) FRAXE and other rare
fragile sites all CCG
Fragile X-associated tremor/ FMR1 CGG <50 50200 Yes (H, M) ..
ataxia syndrome4
Spinocerebellar ataxia 125 ATXN12 CAG 728 4678 Rare Might also be exonic
Frontotemporal dementia and C9ORF72 GGGGCC 217 1050 kb Yes (H) ..
amyotrophic lateral sclerosis6
Exon
Dentatorubral-pallidoluysian ATN1 CAG <48 48 Yes (H, M) ..
atrophy7
Huntingtons disease8 HTT CAG 635 36250 Yes (H, M) ..
Huntingtons disease-like 29 JPH3 CAG 628 4059 Yes (H) Intronic or 3 UTR also
Spinal and bulbar muscular AR CAG 936 3862 Yes (H, M) ..
atrophy10
Spinocerebellar ataxia 111 ATXN1 CAG 2637 3982 Yes (H, M) ..
Spinocerebellar ataxia 211 ATXN2 CAG 1529 3363 Yes (H, M) Also ALS13*
Spinocerebellar ataxia 311 ATXN3 CAG 1235 4784 Yes (H, M) ..
Spinocerebellar ataxia 611 CACNA1A CAG 716 2029 Rare meiotic ..
Spinocerebellar ataxia 711 ATXN7 CAG 814 3662 Yes (H, M) ..
Spinocerebellar ataxia 1711 TBP CAG 2942 4363 Meiotic ..
Intron
Friedreichs ataxia12 FXN GAA 630 661700 Yes (H, M) ..
Myotonic dystrophy 213 CNBP CCTG <30 5511 000 Yes (H, M) Complex repeat
structure
3 UTR
Myotonic dystrophy 18,13 DMPK CTG 537 100 to 2000 Yes (H, M) ..
Spinocerebellar ataxia 814 ATXN8 (CTA)n (CTG)n <33 80 Yes (H) Might also be exonic;
instability in
non-expanded and
expanded repeats

Only disease-causing repeat loci mentioned in the text are included, so many rare diseases caused by expansion of repeat codons, including those giving rise to shorter
polyalanine tracts,15 are not shown. H=seen in human tissues. M=seen in mouse model tissues. FRAXE=fragile site, folic acid type, rare, Fra(X)(Q28) E genetic locus. *ALS13 is a
disease caused by intermediate repeat lengths in ATXN2.

Table 1: Characteristics of selected disease-causing repeat loci

since been found in Huntingtons disease,21 frontotemporal possibility. For example, in trials of aducanumab, an
dementia and amyotrophic lateral sclerosis (which are antibody that binds and reduces deposition of amyloid ,
caused by the C9ORF72 hexanucleotide repeat), and other cognitive benets were recorded in mouse models and in
trinucleotide repeat disorders.22 In frontotemporal clinical studies of people early in the course of
dementia and amyotrophic lateral sclerosis, the C9ORF72 Alzheimers disease.26 Ataxin 1 (ATXN1) oligomers drive
disease-associated repeat dipeptides are neurotoxic,23 the degenerative eects in spinocerebellar ataxia 1 and
although whether such dipeptides have any pathogenic induce local spread of pathology.27 These eects were
role in trinucleotide repeat disorders is unknown. partly inhibited with immunotherapy.28 The biological
Similar mechanisms might be seen in the consequences of expanded polyglutamine have been
polyglutamine (CAG repeat) diseases;22 however, the extensively studied and a wide range of potentially
proteins that contain expanded polyglutamine tracts harmful outcomes detected,29,30 but which of these are
aggregate and form characteristic insoluble protein important in the manifestation of disease remains
inclusions in neural and other cells in these disorders. unclear. Genetic evidence indicates that the DNA damage
Such insoluble inclusions are also widely seen in other response and DNA repair aect the clinical presentation
neurodegenerative disorders,24 which has led to the of Huntingtons disease and multiple spinocerebellar
hypothesis that they or their soluble oligomers are ataxias,3133 which suggests that there are common
pathogenic. So far, the notion that preventing aggregation modiers that act on the mutated repeat itself. Together
can prevent disease in human beings has not been with evidence implicating these processes in trinucleotide
proven.25 Nevertheless, some ndings hint at this repeat disorder biology, these ndings shed light on

www.thelancet.com/neurology Vol 16 January 2017 89


Rapid Review

specic mechanisms and highlight new targets for in the spinocerebellar ataxias caused by CAG repeat
therapeutic intervention. expansions. Why this outcome should be so remains an
outstanding question. The nervous system is vulnerable
The DNA damage response and neurological to DNA damage because of its dependence on and high
disease levels of oxidative metabolism. This process generates
The DNA damage response (table 2) can be both harmful free radicals, which have the potential to cause single-
and protective in people with neurological diseases. strand breaks in DNA.44,49 Variation in genes in the DNA
Mutations in genes involved in the DNA damage response repair machinery can reduce the capacity to repair
were rst noted to cause neurological disease in ataxia- single-strand breaks through the modulation of
telangiectasia, a rare recessive childhood neuro- functional activity, and might, therefore, lead to
degenerative disease. Mutations in the ataxia-telangiectasia neuronal susceptibility. Madabhushi and colleagues50
mutant serine/threonine kinase gene (ATM) cause ataxia- showed that DNA damage and repair can directly aect
telangiectasia. This gene controls cell-cycle arrest after neuronal gene expression: activity-dependent trans-
DNA double-strand breaks, often leading to apoptosis cription of early-response genes in neurons triggered
and, thus, neurodegeneration.43 Mutations in other genes the formation of topoisomerase-II-mediated DNA
that cause incorrect resolution of DNA double-strand double-strand breaks in the promoters of these genes.
breaks also lead to severe developmental disorders of the The products of these early-response genes, such as
nervous system, such as ataxia-telangiectasia-like disease, c-Fos, regulate multiple downstream pathways and
Seckel syndrome involving the ataxia-telangiectasia and aect synapses to exert downstream eects on functions
Rad3-related protein gene (ATR), and Nijmegen breakage such as cognition, learning, and memory.44 Subtle
syndrome.42,44 These disorders also have widespread variation in these DNA repair proteins could alter the
extraneural eects, in contrast to those resulting from timing or repair of DNA double-strand breaks. Notably,
mutations in genes involved in the repair of DNA single- individuals carrying mutations in the tyrosyl-DNA
strand breaks, which usually have eects limited to the phosphodiesterase 2 gene (TDP2) have intellectual
nervous system, although still with harmful clinical disability, epilepsy, and ataxia, and deciency of TDP2
outcomes.43 Spinocerebellar ataxia with axonal neuropathy (which repairs topoisomerase-induced DNA breaks)
is caused by mutations in the tyrosyl-DNA leads to hypersensitivity to topoisomerase-II-mediated
phosphodiesterase 1 gene (TDP1) and the recessive DNA double-strand breaks.51
ataxias with oculomotor apraxia 1, 2, and 4 are caused by Conversely, DNA damage-response factors can
mutations in the aprataxin (APTX), senataxin (SETX), maintain appropriate neurological function and be
and polynucleotide kinase 3-phosphatase (PNKP) genes,45 neuroprotective. Increased DNA double-strand breaks
respectively. TDP1 repairs stalled topoisomerase IDNA have been linked to ageing and pathogenesis in
complexes, APTX and PNKP46 operate on nucleotides, neurodegenerative disorders such as Alzheimers
and SETX encodes a helicase involved in transcriptional disease.52 The breast cancer type 1 susceptibility protein
termination.47,48 (BRCA1), which resolves DNA double-strand breaks
Most of these recessive diseases result in ataxia with during homologous recombination,42 is neuroprotective
prominent cerebellar degeneration, which is also seen in mouse models of Alzheimers disease.53 This nding
complements those from earlier studies. For example,
DNA break type Mode of repair cell models expressing mutant huntingtin (HTT)
Fanconi anaemia Double strand Inter-strand cross-links, possibly other mechanisms34 accumulate DNA single-strand and double-strand
Homologous recombination Double strand Template-directed end joining from other chromatid35 breaks and display concomitant activation of the DNA
Non-homologous end joining Double strand Ligates double-strand breaks without a template36 damage response.54 Mutant HTT binds Ku70, a core
Base-excision repair Single strand Removes damaged bases, lls and ligates the strand37 component of non-homologous end joining,42 and
Direct repair Single strand Direct repair of damaged bases without removal38 overexpression can rescue the phenotype in the
Mismatch repair Single strand Corrects mismatches in replication and short insertions R6/2 model of Huntingtons disease.55 BRCA1 is
and deletions39 recruited to sites of DNA damage by -H2AX. In
Nucleotide-excision repair Single strand Removes DNA modications that cause structural Huntingtons disease cell lines, the amount of BRCA1
distortions40 recruited and the nuclear distribution of -H2AX were
Translesion synthesis .. Synthesises DNA at sites of damage during replication41 reduced,55 and this eect was rescued by overexpression
Lesions in DNA result from exogenous and endogenous processes. Repair, irrespective of cause, is fundamental to
of BRCA1.56 Mutant HTT and ATXN1 bind high-mobility
genome integrity. Mutations might be induced by damaged bases, structural modications of DNA through group protein B family proteins that are components of
supercoiling, looping out of strands, or inter-strand cross-linking; unrepaired lesions can lead to cell death or base-excision repair.42,56 In y and mouse models of
uncontrolled division. Clinically, inherited lesions in the genes involved in the DNA damage response confer
spinocerebellar ataxia 1 with expanded repeats in Atxn1,
susceptibility to cancers.42 These pathways are distinct, but many of the proteins within them operate in multiple
pathways, which must be borne in mind when considering their potential eects in mediating and modulating neuronal pathology was rescued by expression of high-
neuropathology. mobility group protein B1 (HMGB1), which acted to
reverse mitochondrial DNA damage repair in Atxn1
Table 2: Repair pathways of the DNA damage response
knockin mouse brains.57,58

90 www.thelancet.com/neurology Vol 16 January 2017


Rapid Review

Genetic modiers in the trinucleotide repeat genetic signal in the same direction.32 Thus, at least some
disorders genetic modiers in the DNA repair pathways seem to
One way of overcoming the diculties of interpreting act at the level of the mutation type (ie, the repeated
the cell biology ndings is to return to the study of people sequence itself) rather than aecting the functions of
carrying repeat expansions. In natural experiments,2 individual trinucleotide repeat disorder proteins.
where conditions are not controlled by the researchers, it Modication might arise through the somatic expansion
is possible to search for genetic loci that modify disease seen in many trinucleotide repeat disorders. In myotonic
in a benecial or deleterious way, and to reveal the dystrophy type 1, somatic expansions have been detected
biology that is likely to be relevant to disease in blood and other tissues. This discovery enabled
manifestation. For instance, if genetic variation aects Morales and colleagues67 to identify polymorphisms in
the timing of disease onset or the progression or severity the mutS homolog 3 gene (MSH3) in a cohort in Costa
of disease, drug manipulation of a specic biological Rica, which they found were associated with variation in
pathway might have similar eects on the disease somatic instability in blood, although they did not
phenotype. These types of studies in Mendelian disease identify any association with age at onset. Nevertheless,
have practical issues that should be considered. By their this nding indicates an intriguing direct link between
nature, trinucleotide repeat disorders are rare and, DNA repair gene polymorphisms and somatic instability
therefore, achieving samples large enough to provide of repeats, and supports the idea that mismatch repair is
sucient power is challenging. This diculty is being central to repeat expansion.
overcome through networks and consortia that aim to
collect data from large international cohorts of patients, DNA repeat expansions and the DNA damage
including DNA and, crucially, systematically collected response
clinical information. Two examples are the Enroll study Repeat expansions in DNA are aected directly by For the Enroll study in
in Huntingtons disease and the SPATAX consortium in activities of the DNA damage response (table 2).8 The Huntingtons disease see
https://www.enroll-hd.org/
the spinocerebellar ataxias.59 Even so, for many repeats undergo expansion on transmission through the
trinucleotide repeat disorders the sample sizes will be germline, in dividing and terminally dierentiated
small, and the approach of collecting larger and larger somatic cells, and the repeat size increases with age.8
samples to identify relevant genetic loci, as is possible Strand breakage in the repeat is repaired, and it is at this
with common diseases,60 might never be feasible. The point the repeat sequences are thought to expand.64,68 The
successful search for loci that modify age at onset in length of the repeat expansion in Huntingtons disease
Huntingtons disease, however, suggest that it is.31 correlates positively with the propensity for further
Genetic variation that modies rare Mendelian diseases somatic expansion,69 with the greatest expansions
might be caused by common genetic variants and have occurring in the striatum, which might explain why the
substantial eect sizes. As such, variants might not be striatum is particularly susceptible to degeneration in
under population selection pressure, as is seen in many this disorder.70 Proteins containing polyglutamine
common diseases, and might, therefore, be easier to expansions can bind nuclear proteins that operate in
nd. Huntingtons disease is relatively common among DNA repair, such as valosin-containing protein (VCP) or
rare diseases,61,62 and the collection of DNA and transitional endoplasmic reticulum ATPase (TERA),71
systematically collected clinical information, such as in which raises the possibility that accumulation of the
the Registry study of the European Huntingtons Disease expanded polyglutamine proteins induces DNA damage.
Network,63 has allowed an appropriately powered If true, disease pathogenesis might be exacerbated in a
genome-wide association study to be done. Three vicious cycle.
independent genome-wide loci have been signicantly The structure of the repeats aects the likelihood of
associated with age at motor onset, one on chromosome 8 expansion. Trinucleotide repeats can adopt multiple
and two close together on chromosome 15, along with a incorrectly paired structures, including hairpins, loops,
substantial enrichment of signal in the network of DNA triplet helices, and G-quadruplexes (gure).68,72 Bulky
repair genes.31 non-B DNA structures might be stable at large sizes73 and,
Mechanisms related to DNA repair have been therefore, act as substrates for the DNA damage response.
implicated as modulators of somatic expansion of the The CTGCAG repeat sequence adopts multiple transient
disease-associated repeated sequences in mouse models slipped-DNA junctions, leading to unpaired bases that
of Huntingtons disease, myotonic dystrophy,44,64 fragile X could be targets of DNA repair. In support of this
syndrome,65 and Friedreichs ataxia.66 Somatic expansion possibility, the prevalence of slipped-strand features
and the inverse correlation between CAG repeat length correlates with the degree of instability in tissues from
and age at onset are both widely seen in trinucleotide patients with myotonic dystrophy type 1.74 RNADNA
repeat disorders (table 1). In the spinocerebellar ataxias hybrids (R-loops) that are formed during transcription-
associated with CAG repeats, testing for single-nucleotide coupled nucleotide-excision repair prevent repeat
polymorphisms in the DNA repair pathway genes contraction and knock down of SETX, which controls
implicated in Huntingtons disease31 revealed a signicant resolution of R-loops and increases repeat instability in

www.thelancet.com/neurology Vol 16 January 2017 91


Rapid Review

human cell models.75 Damage to individual bases also In mammalian cells, two complexes are involved in
requires repair, and oxidative damage from the generation mismatch repair: MutS, which contains mutS
of reactive oxygen species through mitochondrial homolog 2 (MSH2) and mutS homolog 6 (MSH6) and
dysfunction and excitotoxicity, as is seen in Huntingtons preferentially targets mismatched bases, and MutS,
disease, can lead to the formation of aberrant DNA which contains MSH2 and MSH3 and preferentially
adducts and induction of the DNA damage response.29 targets small insertion-deletions.42,64 The MutS complexes
Such damage might also be potentiated by the expanded recruit the endonuclease MutLa, which is a complex of
repeats themselves through repeat-induced mutagenesis.76 mutL homolog 1 (MLH1) and mismatch repair system
The mechanisms of germline and somatic expansion component PMS1 homolog 2 (PMS2), and cleaves the
might involve dierent pathways, because replication is DNA of the lesioned strand;64 this action can also be
also associated with repeat expansion.77 In non-dividing performed by the MutL complex of MLH1 and
cells, such as neurons, DNA repair activities and mismatch repair system component PMS1, and the
expansion are thought to be associated with DNA damage, MutL complex of MLH1 and MLH3. MutS can cause
transcription, and chromatin dynamics, although some somatic and intergenerational CAGCTG repeat
evidence suggests that environmental stress induces instability, but the evidence for MutS is less consistent.64
DNA re-replication and promotes repeat expansion.78 Knock out of repair genes prevents somatic expansion
and ameliorates the phenotype of Huntingtons disease
Mismatch repair in mice.79,80 Susceptibility to somatic expansion was
Mismatch repair activity on DNA modulates somatic mapped to Mlh1 and Mlh3 in mouse chromosome
expansion of repeat tracts (gure),8 and elements of the substitution experiments,81 and Huntingtons disease
classic mismatch repair pathways might also act as mouse crosses in dierent background strains showed
downstream eectors of other DNA repair mechanisms. that increased concentrations of MSH3 were associated

A Mismatch repair of slipped strands B Transcription-coupled nucleotide-excision C Bulky structures detected by the Fanconi
repair of RNA-DNA hybrids anaemia pathway (hypothetical)
DNA Repeat DNA
Stalled
Hairpins
transcription G-quadruplexes, triple helices
Cruciforms
RNA
Loops TOPOII-mediated
promotor cleavage R-loops
MutS cleavage
FAN1 binding and cleavage by MLH1
MutS-directed and MutS-directed cleavage might be involved in repair

D Base-excision repair of damaged bases

Damage
DNA synthesis across repeat
RIM

OGG1/FEN1 cleavage

Damage to bases might be induced by environmental


and excitotoxic eects and oxidative stress

Figure: DNA repair and repeat expansion mechanisms potentially contributing to repeat instability
DNA mismatch repair (A), transcription-coupled nucleotide-excision repair (B), the Fanconi anaemia pathway (C), and base-excision repair (D) might all have roles
in repeat instability. Slipped strands of dierent sizes can occur in CAG repeats, in which CG bases are Watson-Crick paired and stabilise the looped-out structures,
but the intervening bases are not paired; unpaired bases also occur at the ends of loop structures and at bulges in the DNA, as shown in parts AC. These unpaired
bases are susceptible to damage, which can lead to base-excision repair (D). Transcription-coupled repair can occur as the DNA strands separate for transcription and
the DNA on the non-transcribed strand is unwound and exposed. Elements of the transcriptional machinery can cleave the DNA, and stalled transcription promotes
the formation of R-loops, which predispose to repeat instability (B). In the Fanconi anaemia pathway, FAN1, a structure-specic nuclease, and possibly other
elements of this pathway, might recognise and bind to bulky structures formed by the repeat sequences, such as G-quadruplexes, leading to DNA cleavage that
renders repair necessary and predisposes to repeat instability (C). However, no mechanistic work exists to support this hypothesis. All the DNA structures with
unpaired bases are likely to have an increased propensity for DNA damage, and extrinsic and intrinsic factors, such as oxidative stress, are likely to cause further
harm. Such damage and subsequent base-excision repair leads to DNA repair by gap-lling synthesis, and predisposes to instability of repeats.
TOPOII=topoisomerase II. RIM=repeat-induced mutagenesis.

92 www.thelancet.com/neurology Vol 16 January 2017


Rapid Review

with repeat expansion.82 In cells carrying 800 CAGCTG mechanisms parallel to the topoisomerase-II cleavage
repeats, knock down of MSH2 and MSH3 prevented suggested by Madabhushi and colleagues,50 or by
repeat expansion.74 Similar phenomena are also apparent promoting or lessening DNA expansion at the CAG
in models of Friedreichs ataxia and fragile X syndrome repeat. FAN1 cleaves DNA at inter-strand cross-links, and
in mice, carrying, respectively, GAATTC and CGGCCG is involved in repair in combination with other members
expansions,83,84 and DNA damage-response genes are of the Fanconi anaemia pathway, including FANCD2 and
downregulated in the blood of patients with fragile X the mismatch repair proteins.34 Mutations in genes
syndrome.85 A further nding of note is that histone associated with inter-strand cross-link repair cause
deacetylase enzymes promote repeat expansion via the Fanconi anaemia, but loss-of-function mutations in FAN1
MutS pathway.86 CAG repeat sequences show enhanced lead to the recessive renal syndrome and karyomegalic
convergent transcription (ie, transcription taking place interstitial nephritis,92 and heterozygous truncating
on both strands with RNA polymerases moving mutations cause some familial colorectal cancers,93
towards each other),87 which involves mismatch repair similar to other mismatch repair pathway mutations.
components but induces cell death via ATR focus Rather than recognising specic DNA sequences, FAN1
formation.88 The nal pathways of neurodegeneration recognises branched structures that mimic DNA repair.34,94
and cell death in the trinucleotide repeat disorders might, Repeat sequences, including CAG repeats, adopt non-
therefore, parallel those of other neurological diseases, helical structures in DNA, such as G-quadruplexes,68 and
such as the ataxia-telangiectasia-like diseases that can be FAN1 might target these structures rather than the DNA
caused directly by mutations in ATR. sequence itself (gure). This DNA repair activity is
probably dependent on other mismatch repair proteins,
Base-excision repair which would be consistent with the manipulation of
The response to oxidative damage of DNA by base- mismatch repair genes ameliorating Huntingtons
excision repair aects repeat expansion (gure). disease phenotypes in mice.64 Given that MLH1 is in a
8-Oxoguanine glycosylase (OGG1) removes 8-oxoguanine locus on chromosome 3 that has a signal just below
bases from DNA that has been damaged through the genome-wide signicance in the Huntingtons disease
action of reactive oxygen species. Crossing Huntingtons genome-wide association study,31 and MLH1 interacts
disease knockin mice with Ogg1-decient mice reduces with FAN1, this gene and these proteins might have
somatic expansion of the CAG repeat in Htt and delays central roles in a novel FAN1-driven activity that binds
onset of symptoms. Treatment of mice with a reactive repeats and modulates their instability.
oxygen species scavenger to prevent DNA oxidation
reduces somatic expansion and leads to improvement of Conclusions and future directions
the motor phenotype.89 Flap endonuclease 1 (FEN1) also Showing that genetic modiers exist in Mendelian
has a role in base-excision repair and repeat expansion.90 trinucleotide repeat disorders demonstrates that nding
During base-excision repair of 8-oxoguanine in the DNA genetic modiers in rare genetic diseases is possible, and
of CAG repeats, OGG1 and mutY DNA glycosylase highlights areas of biology that modulate disease in
(MUTYH), which remove adenine incorporated opposite people, such as specic aspects of the DNA damage
unrepaired 8-oxoguanine bases, generate incisions on response: mismatch repair, base-excision repair, and the
opposite DNA strands that might permit repeat Fanconi anaemia pathway. The ndings raise two
expansion,91 although events downstream of DNA important questions. The rst is whether genetic
cleavage are also involved in expansions. For instance, the association with DNA repair processes occurs across all
protein product of the ribonuclease reductase regulator the trinucleotide repeat disorders, and the second is
TP53 inducible subunit M2B gene (RRM2B) is induced whether there is a common mechanism underlying
in brain regions that show somatic expansion of repeats somatic expansion of repeats. To establish the relevance
in the R6/2 mouse model of Huntingtons disease, but of DNA repair and other modiers to the trinucleotide
not in those that do not.91 Of note, RRM2B is in the repeat disorders, more genetic studies, including
genome-wide signicant peak on chromosome 8 in the genome-wide association and sequencing studies, are
Genetic Modiers of Huntingtons Disease (GeM-HD) needed to provide adequate power to identify risk-related
study,31 and has nominal associations in other single-nucleotide polymorphisms and loci. Many of the
trinucleotide repeat disorders.32 repeat diseases have very long repeats that are dicult to
measure accurately, but new sequencing technologies,
The Fanconi anaemia repair pathway including long-read and single-cell sequencing, should
The role of the Fanconi anaemia pathway34 in trinucleotide overcome this diculty and improve accuracy in
repeat disorders is unexplored. However, the determining the exact sequence in the repeat, including
chromosome 15 locus associated with age at onset of any interruptions.95 Work being done in Huntingtons
Huntingtons disease31 contains FAN1. The product of disease and other polyglutamine diseases suggests that
this gene, the DNA nuclease FAN1, is a candidate for combining multiple diseases in one analysis might be a
modifying the onset of Huntingtons disease through the possible route to increasing power.32

www.thelancet.com/neurology Vol 16 January 2017 93


Rapid Review

presented in this Rapid Review open a new window on


Search strategy and selection criteria repeat expansion disease, with obvious avenues for
We searched PubMed for papers published from Jan 1, 2012, to therapeutic exploitation.
Sept 30, 2016, using combinations of the terms huntingt*, Contributions
spinocerebellar ataxia, trinucleotide repeat, triplet repeat, LJ did the literature search, wrote the rst draft of the review, and
prepared the gures and tables. HH and SJT edited the drafts and
repeat, or repeat disease, and DNA integrity, DNA suggested additional ndings from their knowledge of the literature.
repair, genome integrity, or genome repair. Our search was
Declaration of interests
limited to publications that included search terms in the title, We declare no competing interests.
abstract, or both. We identied further relevant papers by
References
searching the reference lists of retrieved papers and through 1 Orr HT, Zoghbi HY. Trinucleotide repeat disorders.
searches of our les. The nal reference list was based on Annu Rev Neurosci 2007; 30: 575621.
relevance to the topic of this Rapid Review. 2 Gusella JF, MacDonald ME, Lee J-M. Genetic modiers of
Huntingtons disease. Mov Disord 2014; 29: 135965.
3 Metsu S, Rooms L, Rainger J, et al. FRA2A is a CGG repeat
expansion associated with silencing of AFF3. PLoS Genet 2014;
Elucidating the mechanistic consequences of genetic 10: e1004242.
variation will be challenging. Cell and animal models of 4 Lokanga RA, Entezam A, Kumari D, et al. Somatic expansion in
mouse and human carriers of fragile X premutation alleles.
many of the trinucleotide repeat disorders are already Hum Mutat 2013; 34: 15766.
available, and could be rened with knowledge of genetic 5 Dong Y, Wu J-J, Wu Z-Y. Identication of 46 CAG repeats within
modiers and common biology. Such knowledge will PPP2R2B as probably theshortest pathogenic allele for SCA12.
allow development of common downstream assays that Parkinsonism Relat Disord 2015; 21: 398401.
6 van Blitterswijk M, DeJesus-Hernandez M, Niemantsverdriet E,
reect the disease biology that is important in human et al. Association between repeat sizes and clinical and pathological
beings. So far, however, there is no information about characteristics in carriers of C9ORF72 repeat expansions
the direct molecular eects of the genetic variation (Xpansize-72): a cross-sectional cohort study. Lancet Neurol 2013;
12: 97888.
detected. Thus, whether the function of the DNA 7 Tsuji S. Dentatorubral-pallidoluysian atrophy. Handb Clin Neurol
damage response is improved or inhibited by these 2012; 103: 58794.
changes is unknown. As in complex diseases, clues can 8 Schmidt MHM, Pearson CE. Disease-associated repeat instability
and mismatch repair. DNA Repair (Amst) 2016; 38: 11726.
be gathered by aggregating data about likely changes in
9 Todd PK, Paulson HL. RNA-mediated neurodegeneration in repeat
expression and the functional eects of coding changes expansion disorders. Ann Neurol 2010; 67: 291300.
in genes and pathways, and using genome-wide data, 10 Tanaka F, Reeves MF, Ito Y, et al. Tissue-specic somatic mosaicism
such as co-expression networks96 and protein-interaction in spinal and bulbar muscular atrophy is dependent on CAG-repeat
length and androgen receptorgene expression level.
networks.97 Am J Hum Genet 1999; 65: 96673.
Downstream from genetic discovery, to establish 11 Tezenas du Montcel S, Durr A, Bauer P, et al. Modulation of the age
whether somatic expansion or other DNA repair or at onset in spinocerebellar ataxia by CAG tracts in various genes.
Brain 2014; 137: 244455.
integrity mechanisms are responsible for the genetic
12 Evans-Galea M V, Lockhart PJ, Galea CA, Hannan AJ, Delatycki MB.
signal, cell biology and animal studies will be necessary. Beyond loss of frataxin: the complex molecular pathology of
Human tissue will be needed to investigate gene Friedreich ataxia. Discov Med 2014; 17: 2535.
expression and to show the eects of the variants in vivo. 13 Udd B, Krahe R. The myotonic dystrophies: molecular, clinical, and
therapeutic challenges. Lancet Neurol 2012; 11: 891905.
One immediate priority is establishing the eects of 14 Martins S, Seixas AI, Magalhes P, Coutinho P, Sequeiros J,
manipulating the Fanconi anaemia pathway, and work in Silveira I. Haplotype diversity and somatic instability in normal and
this area is just beginning. Potential emerging pathogenic expanded SCA8 alleles. Am J Med Genet Part B Neuropsychiatr Genet
2005; 139B: 10914.
mechanisms that require further investigation include 15 Shoubridge C, Gecz J. Polyalanine tract disorders and
the operation of repeat-induced mutagenesis, where DNA neurocognitive phenotypes. In: Hannan AJ, ed. Tandem repeat
repair activities lead to further mutagenesis around polymorphisms: genetic plasticity, neural diversity and disease.
Austin, TX: Landes Bioscience and Springer Science+Business
repeat loci,76 and the relevance of chromatin structure to Media, 2013.
repeat exposure and dynamics.98 Detailing the exact 16 Trang H, Stanley SY, Thorner P, et al. Massive CAG repeat
nature of the mechanisms of the DNA damage response expansion and somatic instability in maternally transmitted
infantile spinocerebellar ataxia type 7. JAMA Neurol 2015;
essential in neurodegeneration will allow the development 72: 21923.
of new drugs and possibly the repurposing of existing 17 Nageshwaran S, Festenstein R. Epigenetics and triplet repeat
treatments that target the DNA damage response in neurological diseases. Front Neurol 2015; 6: 262.
cancers.42 18 Cooper TA, Wan L, Dreyfuss G. RNA and disease. Cell 2009;
136: 77793.
Finally, mechanisms common to multiple diseases 19 Echeverria G V, Cooper TA. RNA-binding proteins in microsatellite
oer hope that treatments can be developed that will be expansion disorders: mediators of RNA toxicity. Brain Res 2012;
applicable across disorders. This approach can be seen in 1462: 10011.
20 Zu T, Gibbens B, Doty NS, et al. Non-ATG-initiated translation
the case of cancers, where common biological pathways directed by microsatellite expansions. Proc Natl Acad Sci USA 2011;
are dysregulated in multiple forms of the disease, and 108: 26065.
the same chemotherapeutic agents can be used in 21 Baez-Coronel M, Ayhan F, Tarabochia AD, et al. RAN translation
in Huntington disease. Neuron 2015; 88: 66777.
dierent regimens to treat various cancers.99 The ndings

94 www.thelancet.com/neurology Vol 16 January 2017


Rapid Review

22 Cleary JD, Ranum LPW. Repeat associated non-ATG (RAN) 48 Yce , West SC. Senataxin, defective in the neurodegenerative
translation: new starts in microsatellite expansion disorders. disorder ataxia with oculomotor apraxia 2, lies at the interface of
Curr Opin Genet Dev 2014; 26: 615. transcription and the DNA damage response. Mol Cell Biol 2013;
23 Tran H, Almeida S, Moore J, et al. Dierential toxicity of nuclear 33: 40617.
RNA foci versus dipeptide repeat proteins in a Drosophila model of 49 Barzilai A. The contribution of the DNA damage response to
C9ORF72 FTD/ALS. Neuron 2015; 87: 120714. neuronal viability. Antioxid Redox Signal 2006; 9: 21118.
24 Knowles TPJ, Vendruscolo M, Dobson CM. The amyloid state and 50 Madabhushi R, Gao F, Pfenning AR, et al. Activity-induced DNA
its association with protein misfolding diseases. breaks govern the expression of neuronal early-response genes.
Nat Rev Mol Cell Biol 2014; 15: 38496. Cell 2015; 161: 1592605.
25 Harrison JR, Owen MJ. Alzheimers disease: the amyloid 51 Gomez-Herreros F, Schuurs-Hoeijmakers JHM, McCormack M,
hypothesis on trial. Br J Psychiatry 2016; 208: 13. et al. TDP2 protects transcription from abortive topoisomerase
26 Sevigny J, Chiao P, Bussiere T, et al. The antibody aducanumab activity and is required for normal neural function. Nat Genet 2014;
reduces Abeta plaques in Alzheimers disease. Nature 2016; 46: 51621.
537: 5056. 52 Suberbielle E, Sanchez PE, Kravitz AV, et al. Physiologic brain
27 Lasagna-Reeves CA, Rousseaux MWC, Guerrero-Muoz MJ, et al. activity causes DNA double-strand breaks in neurons, with
A native interactor scaolds and stabilizes toxic ATAXIN-1 exacerbation by amyloid-beta. Nat Neurosci 2013; 16: 61321.
oligomers in SCA1. Elife 2015; 4: e07558. 53 Suberbielle E, Djukic B, Evans M, et al. DNA repair factor BRCA1
28 Lasagna-Reeves CA, Rousseaux MWC, Guerrero-Munoz MJ, et al. depletion occurs in Alzheimer brains and impairs cognitive
Ataxin-1 oligomers induce local spread of pathology and decreasing function in mice. Nat Commun 2015; 6: 8897.
them by passive immunization slows spinocerebellar ataxia type 1 54 Illuzzi J, Yerkes S, Parekh-Olmedo H, Kmiec EB. DNA breakage
phenotypes. Elife 2015; 4: e10891. and induction of DNA damage response proteins precede the
29 Bates GP, Dorsey R, Gusella JF, et al. Huntington disease. appearance of visible mutant huntingtin aggregates.
Nat Rev Dis Prim 2015; 1: 15005. J Neurosci Res 2009; 87: 73347.
30 Guo JL, Lee VMY. Cell-to-cell transmission of pathogenic proteins 55 Enokido Y, Tamura T, Ito H, et al. Mutant huntingtin impairs
in neurodegenerative diseases. Nat Med 2014; 20: 13038. Ku70-mediated DNA repair. J Cell Biol 2010; 189: 42543.
31 Genetic Modiers of Huntingtons Disease (GeM-HD) Consortium. 56 Jeon GS, Kim KY, Hwang YJ, et al. Deregulation of BRCA1 leads to
Identication of genetic factors that modify clinical onset of impaired spatiotemporal dynamics of gamma-H2AX and DNA
Huntingtons disease. Cell 2015; 162: 51626. damage responses in Huntingtons disease. Mol Neurobiol 2012;
32 Bettencourt C, Moss DH, Flower M, et al. DNA repair pathways 45: 55063.
underlie a common genetic mechanism modulating onset in 57 Qi M-L, Tagawa K, Enokido Y, et al. Proteome analysis of soluble
polyglutamine diseases. Ann Neurol 2016; 79: 98390 . nuclear proteins reveals that HMGB1/2 suppress genotoxic stress
33 Martins S, Pearson CE, Coutinho P, et al. Modiers of (CAG)n in polyglutamine diseases. Nat Cell Biol 2007; 9: 40214.
instability in MachadoJoseph disease (MJD/SCA3) transmissions: 58 Ito H, Fujita K, Tagawa K, et al. HMGB1 facilitates repair of
an association study with DNA replication, repair and mitochondrial DNA damage and extends the lifespan of mutant
recombination genes. Hum Genet 2014; 133: 131118. ataxin-1 knock-in mice. EMBO Mol Med 2015; 7: 78101.
34 Ceccaldi R, Sarangi P, DAndrea AD. The Fanconi anaemia 59 Jacobi H, du Montcel ST, Bauer P, et al. Long-term disease
pathway: new players and new functions. Nat Rev Mol Cell Biol 2016; progression in spinocerebellar ataxia types 1, 2, 3, and 6:
17: 33749. a longitudinal cohort study. Lancet Neurol 2015; 14: 110108.
35 San Filippo J, Sung P, Klein H. Mechanism of eukaryotic 60 Spencer CCA, Su Z, Donnelly P, Marchini J. Designing genome-
homologous recombination. Annu Rev Biochem 2008; 77: 22957. wide association studies: sample size, power, imputation, and the
36 Lieber MR. The mechanism of double-strand DNA break repair by choice of genotyping chip. PLoS Genet 2009; 5: e1000477.
the nonhomologous DNA end-joining pathway. Annu Rev Biochem 61 Wexler NS, Collett L, Wexler AR, et al. Incidence of adult
2010; 79: 181211. Huntingtons disease in the UK: a UK-based primary care study and
37 Robertson AB, Klungland A, Rognes T, Leiros I. DNA repair in a systematic review. BMJ Open 2016; 6: 009070.
mammalian cells: base excision repair: the long and short of it. 62 Evans SJW, Douglas I, Rawlins MD, Wexler NS, Tabrizi SJ,
Cell Mol Life Sci 2009; 66: 98193. Smeeth L. Prevalence of adult Huntingtons disease in the UK
38 Eker APM, Quayle C, Chaves I, van der Horst GTJ. DNA repair in based on diagnoses recorded in general practice records.
mammalian cells: direct DNA damage reversal: elegant solutions J Neurol Neurosurg Psychiatry 2013; 84: 115660.
for nasty problems. Cell Mol Life Sci 2009; 66: 96880. 63 Orth M, for the European Huntingtons Disease Network.
39 Kunkel TA, Erie DA. DNA mismatch repair. Annu Rev Biochem Observing Huntingtons disease: the European Huntingtons
2005; 74: 681710. Disease Networks REGISTRY. J Neurol Neurosurg Psychiatry 2011;
82: 140912.
40 de Laat WL, Jaspers NGJ, Hoeijmakers JHJ. Molecular mechanism
of nucleotide excision repair. Genes Dev 1999; 13: 76885. 64 Iyer RR, Pluciennik A, Napierala M, Wells RD. DNA triplet repeat
expansion and mismatch repair. Annu Rev Biochem 2015; 84: 199226.
41 Waters LS, Minesinger BK, Wiltrout ME, DSouza S, Woodru RV,
Walker GC. Eukaryotic translesion polymerases and their roles and 65 Lokanga RA, Zhao X-N, Usdin K. The mismatch repair protein
regulation in DNA damage tolerance. Microbiol Mol Biol Rev 2009; MSH2 is rate limiting for repeat expansion in a fragile X
73: 13454. premutation mouse model. Hum Mutat 2014; 35: 12936.
42 Pearl LH, Schierz AC, Ward SE, Al-Lazikani B, Pearl FMG. 66 Ezzatizadeh V, Pinto RM, Sandi C, et al. The mismatch repair
Therapeutic opportunities within the DNA damage response. system protects against intergenerational GAA repeat instability in
Nat Rev Cancer 2015; 15: 16680. a Friedreich ataxia mouse model. Neurobiol Dis 2012; 46: 16571.
43 Paull TT. Mechanisms of ATM activation. Annu Rev Biochem 2015; 67 Morales F, Vsquez M, Santamara C, Cuenca P, Corrales E,
84: 71138. Monckton DG. A polymorphism in the MSH3 mismatch repair
gene is associated with the levels of somatic instability of the
44 McKinnon PJ. DNA repair deciency and neurological disease.
expanded CTG repeat in the blood DNA of myotonic dystrophy
Nat Rev Neurosci 2009; 10: 10012.
type 1 patients. DNA Repair (Amst) 2016; 40: 5766.
45 Bras J, Alonso I, Barbot C, et al. Mutations in PNKP cause recessive
68 McMurray CT. Mechanisms of trinucleotide repeat instability
ataxia with oculomotor apraxia type 4. Am J Hum Genet 2015;
during human development. Nat Rev Genet 2010; 11: 78699.
96: 47479.
69 Veitch NJ, Ennis M, McAbney JP, Shelbourne PF, Monckton DG.
46 Weinfeld M, Mani RS, Abdou I, Aceytuno RD, Glover JNM.
Inherited CAGCTG allele length is a major modier of somatic
Tidying up loose ends: the role of polynucleotide kinase/phosphatase
mutation length variability in Huntington disease.
in DNA strand break repair. Trends Biochem Sci 2011; 36: 26271.
DNA Repair (Amst) 2007; 6: 78996.
47 Hatchi E, Skourti-Stathaki K, Ventz S, et al. BRCA1 recruitment to
70 Shelbourne PF, Keller-McGandy C, Bi WL, et al. Triplet repeat
transcriptional pause sites is required for R-loop-driven DNA
mutation length gains correlate with cell-type specic vulnerability
damage repair. Mol Cell 2015; 57: 63647.
in Huntington disease brain. Hum Mol Genet 2007; 16: 113342.

www.thelancet.com/neurology Vol 16 January 2017 95


Rapid Review

71 Fujita K, Nakamura Y, Oka T, et al. A functional deciency of TERA/ 86 Gannon A-MM, Frizzell A, Healy E, Lahue RS. MutSbeta and
VCP/p97 contributes to impaired DNA repair in multiple histone deacetylase complexes promote expansions of trinucleotide
polyglutamine diseases. Nat Commun 2013; 4: 1816. repeats in human cells. Nucleic Acids Res 2012; 40: 1032433.
72 Mirkin SM. Expandable DNA repeats and human disease. 87 Lin Y, Wilson JH. Nucleotide excision repair, mismatch repair, and
Nature 2007; 447: 93240. R-loops modulate convergent transcription-induced cell death and
73 Volker J, Plum GE, Gindikin V, Klump HH, Breslauer KJ. Impact of repeat instability. PLoS One 2012; 7: e46807.
bulge loop size on DNA triplet repeat domains: Implications for 88 Chatterjee N, Lin Y, Wilson JH. Mismatch repair enhances
DNA repair and expansion. Biopolymers 2014; 101: 112. convergent transcription-induced cell death at trinucleotide repeats
74 Axford MM, Wang Y-H, Nakamori M, Zannis-Hadjopoulos M, by activating ATR. DNA Repair (Amst) 2016; 42: 2632.
Thornton CA, Pearson CE. Detection of slipped-DNAs at the 89 Budworth H, Harris FR, Williams P, et al. Suppression of somatic
trinucleotide repeats of the myotonic dystrophy type I disease locus expansion delays the onset of pathophysiology in a mouse model of
in patient tissues. PLoS Genet 2013; 9: e1003866. Huntingtons disease. PLoS Genet 2015; 11: e1005267.
75 Nakatani R, Nakamori M, Fujimura H, Mochizuki H, 90 Liu Y, Wilson SH. DNA base excision repair: a mechanism of
Takahashi MP. Large expansion of CTGCAG repeats is exacerbated trinucleotide repeat expansion. Trends Biochem Sci 2012; 37: 16272.
by MutSbeta in human cells. Sci Rep 2015; 5: 11020. 91 Cilli P, Ventura I, Minoprio A, et al. Oxidized dNTPs and the OGG1
76 Shah KA, Mirkin SM. The hidden side of unstable DNA repeats: and MUTYH DNA glycosylases combine to induce CAG/CTG
Mutagenesis at a distance. DNA Repair (Amst) 2015; 32: 10612. repeat instability. Nucleic Acids Res 2016; 44: 5190203.
77 Iyama T, Wilson DM 3rd. DNA repair mechanisms in dividing and 92 Zhou W, Otto EA, Cluckey A, et al. FAN1 mutations cause
non-dividing cells. DNA Repair (Amst) 2013; 12: 62036. karyomegalic interstitial nephritis, linking chronic kidney failure to
78 Chatterjee N, Lin Y, Santillan BA, Yotnda P, Wilson JH. defective DNA damage repair. Nat Genet 2012; 44: 9105.
Environmental stress induces trinucleotide repeat mutagenesis in 93 Segui N, Mina LB, Lazaro C, et al. Germline mutations in FAN1
human cells. Proc Natl Acad Sci USA 2015; 112: 376469. cause hereditary colorectal cancer by impairing DNA repair.
79 Wheeler VC, Lebel L-A, Vrbanac V, Teed A, te Riele H, Gastroenterology 2015; 149: 56366.
MacDonald ME. Mismatch repair gene Msh2 modies the timing 94 Pennell S, Declais A-C, Li J, et al. FAN1 activity on asymmetric
of early disease in HdhQ111 striatum. Hum Mol Genet 2003; repair intermediates is mediated by an atypical monomeric
12: 27381. virus-type replication-repair nuclease domain. Cell Rep 2014;
80 Dragileva E, Hendricks A, Teed A, et al. Intergenerational and 8: 8493.
striatal CAG repeat instability in Huntingtons disease knock-in 95 McFarland KN, Liu J, Landrian I, et al. SMRT Sequencing of long
mice involve dierent DNA repair genes. Neurobiol Dis 2009; tandem nucleotide repeats in SCA10 reveals unique insight of
33: 3747. repeat expansion structure. PLoS One 2015; 10: e0135906.
81 Pinto RM, Dragileva E, Kirby A, et al. Mismatch repair genes Mlh1 96 Parikshak NN, Gandal MJ, Geschwind DH. Systems biology and
and Mlh3 Modify CAG instability in Huntingtons disease mice: gene networks in neurodevelopmental and neurodegenerative
genome-wide and candidate approaches. PLoS Genet 2013; disorders. Nat Rev Genet 2015; 16: 44158.
9: e1003930. 97 Szklarczyk D, Franceschini A, Wyder S, et al. STRING v10:
82 Tom S, Manley K, Simard JP, et al. MSH3 polymorphisms and proteinprotein interaction networks, integrated over the tree of
protein levels aect CAG repeat instability in Huntingtons disease life. Nucleic Acids Res 2015; 43: D44752.
mice. PLoS Genet 2013; 9: 116. 98 Viterbo D, Michoud G, Mosbach V, Dujon B, Richard G-F.
83 Bourn RL, De Biase I, Pinto RM, et al. Pms2 suppresses large Replication stalling and heteroduplex formation within CAG/CTG
expansions of the (GAA.TTC)n sequence in neuronal tissues. trinucleotide repeats by mismatch repair. DNA Repair (Amst) 2016;
PLoS One 2012; 7: e47085. 42: 94106.
84 Zhao X-N, Kumari D, Gupta S, et al. Mutsbeta generates both 99 Longley DB, Harkin DP, Johnston PG. 5-uorouracil: mechanisms
expansions and contractions in a mouse model of the fragile of action and clinical strategies. Nat Rev Cancer 2003; 3: 33038.
X-associated disorders. Hum Mol Genet 2015; 24: 708796.
85 Xu H, Rosales-Reynoso MA, Barros-Nunez P, Peprah E.
DNA repair/replication transcripts are down regulated in patients
with fragile X syndrome. BMC Res Notes 2013; 6: 90.

96 www.thelancet.com/neurology Vol 16 January 2017

Potrebbero piacerti anche