Tandem repeat disorders: from diagnosis to emerging therapeutic strategies

Article information

encephalitis. 2024;.encephalitis.2024.00122
Publication date (electronic) : 2024 December 10
doi : https://doi.org/10.47936/encephalitis.2024.00122
Rare Disease Center, Department of Genomic Medicine; Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
Correspondence: Jangsup Moon Departments of Genomic Medicine and Neurology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea E-mail: jangsup.moon@gmail.com
Received 2024 September 15; Revised 2024 October 28; Accepted 2024 October 28.

Abstract

Tandem repeat disorders (TRDs) are genetic conditions characterized by the abnormal expansion of repetitive DNA sequences within specific genes. The growing number of identified TRDs highlights their complexity, with varied molecular mechanisms ranging from toxic protein production and repeat-associated non-AUG translation to RNA toxicity and epigenetic modifications. TRDs also exhibit unique clinical features such as reduced penetrance, anticipation, and repeat motif changes. Advances in molecular diagnostics such as long-read sequencing have significantly improved the detection of TRDs, especially for large or complex repeat expansions. Additionally, emerging therapeutic strategies, particularly antisense oligonucleotides (ASOs) and gene editing technologies, are showing great promise. ASOs in particular have demonstrated success through mechanisms like allele-specific knockdown and splice modulation. In this review, we explore the classification of TRDs, advances in diagnostics, molecular mechanisms, clinical features, and innovative therapeutic strategies, highlighting the need for further research to refine treatments and improve outcomes.

Introduction

Tandem repeat disorders (TRDs) are a group of genetic conditions caused by the expansion of repetitive DNA sequences, known as tandem repeats, within specific genes [1]. These sequences, which typically consist of 2-6 consecutively repeated nucleotide units, can expand beyond the normal range, leading to gene dysfunction and a variety of clinical symptoms [2]. The abnormal expansion of these repeats disrupts cellular processes either by altering the structure and function of the encoded proteins or by interfering with gene regulation and RNA processing. The inherent genetic instability of these repetitive sequences poses significant challenges in understanding, diagnosing, and treating associated disorders [3].

Owing to the advances in molecular genetics and sequencing technologies, the number of identified TRDs has steadily increased [4]. TRDs are currently recognized as a diverse and complex group of disorders, each with unique molecular mechanisms and clinical presentations [5,6]. These disorders serve as critical models for developing novel diagnostic and therapeutic approaches. Despite significant progress, TRDs continue to present challenges in clinical management, underscoring the need for ongoing research and innovation [7]. In this review, we provide a comprehensive overview of classification, diagnostic approaches, pathogenesis, and emerging therapeutic strategies for TRDs, highlighting both current knowledge and future directions in the field.

Classification of tandem repeat disorders

Different types of tandem repeat disorders

TRDs can be classified based on the location of the repeat sequence within the gene and the nature of the repeat motif (Table 1) [2]. Tandem repeats can be found either within the coding regions of genes or in noncoding regions [1]. Repeat expansions within coding regions often lead to the production of abnormal proteins that can aggregate and disrupt cellular functions, while repeat expansions in noncoding regions can interfere with gene regulation, RNA processing, and other cellular processes without directly altering protein sequences [8]. For example, Huntington disease (HD) involves a CAG trinucleotide repeat expansion in the coding region of the HTT gene, resulting in a production of a toxic mutant protein that causes neurodegeneration [9]. On the other hand, fragile X syndrome (FXS) is caused by a CGG trinucleotide repeat expansion in the 5' untranslated region (UTR) of the FMR1 gene, leading to gene silencing through methylation [10].

Classification of tandem repeat disorders

TRDs can also be classified based on the sequence and length of the repeat motif. The most common types of repeat motifs are trinucleotide repeats, such as CAG and CGG, in disorders like HD and FXS [9,11]. Other motifs include tetranucleotide repeats (e.g., CCTG in myotonic dystrophy type 2) [12], pentanucleotide repeats (e.g., ATTCT in spinocerebellar ataxia [SCA] type 10) [13], and hexanucleotide repeats (e.g., GGGGCC in C9ORF72-related disorders) [14]. Understanding the various types of repeat motifs and their role in disease pathogenesis is crucial for developing targeted therapeutic approaches for these disorders.

Recent increases in tandem repeat disorder discovery

Since the first TRD FXS was discovered in the 1990s, there has been a steady increase in the identification of new TRDs, with approximately 50 different TRDs identified to date [1]. In the early years, new TRDs were primarily discovered through linkage analysis and microarray studies, which enabled researchers to associate specific genetic regions with disease phenotypes. The advent of next-generation sequencing (NGS) technologies accelerated the discovery of TRDs by allowing more comprehensive exploration of the genome [15]. In recent years, the development of long-read sequencing (LRS) technologies and advanced computational tools and algorithms has revolutionized the field, leading to a rapid acceleration in the discovery of TRDs [16]. Many of the newly discovered TRDs identified through LRS are in noncoding regions, often involving GC-rich motifs, repeats of variable length, or even mixture of different repeat motifs, which were previously difficult to detect with traditional short-read sequencing methods [17,18]. A detailed explanation of the diagnostic methods for TRD will be provided in the subsequent section.

Diagnostics in tandem repeat disorders

Conventional methods: polymerase chain reaction-based techniques and Southern blotting

The diagnosis of TRDs has traditionally relied on conventional molecular techniques, each suited to detect specific types and sizes of repeat expansions (Table 2). These methods include standard polymerase chain reaction (PCR) with fragment length analysis, repeat-primed PCR (RP-PCR), and Southern blotting.

Diagnostic techniques for tandem repeat disorders

Conventional PCR followed by fragment length analysis is often sufficient for the diagnosis of TRDs with relatively small to moderate repeat expansions [19]. In this approach, primers are designed to flank the repeat region, allowing for the amplification of the entire repeat sequence. The size of the PCR products is then determined and the presence of repeat expansions is determined. This method is particularly effective if the repeat expansions do not exceed the amplification capabilities of standard PCR, typically up to several hundred base pairs. However, when repeat expansions are very large or GC-rich, standard PCR is not applicable and other methods may be required [20].

RP-PCR is commonly used as a screening tool to detect the presence of expanded repeat alleles, particularly when standard PCR may fail due to the large size of the expansion [21,22]. In RP-PCR, one of the primers is specifically designed to bind within the repetitive sequence itself, while the other primers bind to the flanking regions. When a repeat expansion is present, the PCR amplification generates a characteristic “ladder” pattern of products due to the varying lengths of repeats being amplified. This pattern is typically observed as a long tail on length analysis [23]. While RP-PCR is highly sensitive for detecting the presence of repeat expansions, it does not provide precise sizing, especially for very large expansions. Therefore, RP-PCR is primarily used as a preliminary screening method, with positive results often requiring confirmation by more detailed methods like Southern blotting [24].

Southern blotting has long been considered the gold standard for confirming large tandem repeat expansions that cannot be accurately sized by PCR-based methods [25,26]. The process involves the digestion of DNA with restriction enzymes, followed by hybridization with a labeled probe that binds to the region containing the repeat. This technique is particularly useful for the detection of large repeat expansions and provides detailed information on the size of the expanded repeat alleles [27]. However, Southern blotting has significant limitations. It is technically challenging and time-consuming and requires a large amount of high-quality DNA which may not always be available from patient samples. Additionally, Southern blotting must be carefully optimized for each specific disease, which can make it difficult to set up and standardize across different conditions.

Next-generation sequencing: short-read sequencing

NGS has dramatically changed the landscape of genetic diagnostics, offering high-throughput and comprehensive analysis of the genome [28]. However, short-read NGS, which sequences DNA in small fragments of 100 to 300 base pairs, has limitations when it comes to diagnosing TRDs. The reads may not cover the entire repeat region, leading to fragmented and incomplete data, which can cause difficulties in correctly identifying and sizing the expansions.

Despite these challenges, recent advancements in bioinformatics have enabled short-read NGS to diagnose some TRDs. Specialized tools and algorithms, such as ExpansionHunter [29,30], STRetch [31], and GangSTR [32], have been developed to identify repeat expansions from short-read data by analyzing the patterns of sequencing reads that map to repeat regions. These tools have improved the ability to diagnose rare forms of TRDs using NGS. However, the approach remains inadequate for detecting all TRDs, especially those involving large or complex repeat expansions.

The need for long-read sequencing

To overcome the limitations of short-read NGS, there is a growing need for LRS technologies in the diagnosis of TRDs [33]. LRS platforms, offered by PacBio and Oxford Nanopore, enable the sequencing of much longer DNA fragments, often exceeding tens of thousands of base pairs [34]. This capability is particularly valuable for accurately detecting and sizing large repeat expansions that are difficult or impossible to resolve with short-read data [17,18]. The utility of LRS is further enhanced by integrating new technologies, such as clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9. Nanopore Cas9-targeted sequencing (nCATS) enables targeted and focused LRS of specific genomic regions, providing highly accurate sizing and structural characterization, and facilitating the diagnosis of TRDs [35].

LRS also offers the advantage of analyzing DNA methylation patterns within repeat regions [36], which is important for understanding the epigenetic factors involved in certain TRDs, such as FXS. As costs decrease and accessibility improves, LRS is set to become a key tool in TRD diagnostics, offering more comprehensive and accurate genetic analysis than currently possible with short-read NGS.

Molecular mechanisms and clinical characteristics of tandem repeat disorders

Molecular mechanism of pathogenesis

The pathogenesis of TRDs can be categorized into gain-of-function (GOF) and loss-of-function mechanisms that contribute to disease development and progression.

Toxic mutant protein production (gain-of-function)

A key GOF mechanism in TRDs is the formation of toxic mutant proteins due to repeat expansions in coding regions [37]. In polyglutamine (polyQ) diseases like HD and SCAs (SCA1, SCA2, SCA3), CAG repeat expansions in genes like HTT [38] and ataxin [39-41] produce proteins with elongated polyQ tracts, leading to misfolding, aggregation, and neurodegeneration. In other disorders, such as oculopharyngeal muscular dystrophy, GCN repeat expansions in the PABPN1 gene create an expanded polyalanine tract, causing protein aggregation in muscle cells and contributing to muscle weakness [42]. In these disorders, the accumulation of misfolded proteins is central to the disease process.

Repeat-associated non-AUG translation (gain-of-function)

Another GOF mechanism involves repeat-associated non-AUG (RAN) translation, where expanded repeats in noncoding regions trigger the synthesis of toxic peptides from normally UTRs [43,44]. This occurs without the need for a conventional start codon (AUG). Disorders such as neuronal intranuclear inclusion disease (NIID) exemplify this mechanism. In NIID, the expansion of GGC repeats in the NOTCH2NLC gene leads to the production of toxic dipeptide repeat proteins through RAN translation [17]. The accumulation of these peptides within neurons contributes to cellular dysfunction, ultimately leading to the characteristic intranuclear inclusions seen in the disease.

RNA-binding protein sequestration (gain-of-function)

In some TRDs, the pathogenic mechanism involves RNA toxicity. In myotonic dystrophy, the CTG repeats in the noncoding region of the DMPK gene, lead to the formation of abnormal RNA structures. These structures sequester RNA-binding proteins, which are essential for normal RNA processing [45]. The sequestration of these proteins disrupts RNA splicing and other critical cellular functions, leading to a toxic GOF effect.

Epigenetic modifications and gene silencing (loss-of-function)

In certain TRDs, repeat expansions in noncoding regions lead to epigenetic modifications, such as DNA methylation, histone modification, and heterochromatic protein 1 (HP1)-mediated silencing, which all contribute to the silencing of gene expression [6]. For example, in FXS, transcriptional silencing of the FMR1 gene is triggered by a combination of DNA methylation and histone modification [46]. Histone modifications have also been observed in HD, where they can compact chromatin and make the gene less accessible for transcription [47]. In Friedreich’s ataxia, transcriptional silencing is known to occur in the absence of DNA methylation, mainly by HP1-mediated gene silencing [48].

R-loop formation (loss-of-function)

R-loops are three-stranded nucleic acid structures that can form when expanded repeats cause RNA to hybridize with complementary DNA strands, leaving the other DNA strand unpaired [49]. This can disrupt the normal process of gene transcription and lead to genomic instability. In Friedreich’s ataxia, GAA repeat expansions in the FXN gene form R-loops that repress gene expression, reducing frataxin and causing mitochondrial dysfunction [50]. Similarly, in FXS, CGG repeat expansions in the FMR1 gene promote R-loops, leading to gene silencing and reduced fragile X mental retardation protein (FMRP) levels [50,51].

Reduced penetrance

In some cases of TRDs, individuals may carry intermediate repeat expansions, also known as premutations, where the repeat length is larger than normal but not sufficient to cause the full-blown disease [2]. These premutations can lead to reduced or incomplete penetrance, meaning that not all individuals with the expansion will develop the clinical symptoms associated with the disorder. For example, in HD, individuals with 36-39 CAG repeats in the HTT gene often show reduced penetrance, where they may not develop symptoms until late in life, or they may not develop symptoms at all [52]. Similarly, in fragile X-associated tremor/ataxia syndrome, individuals with a premutation in the FMR1 gene typically have 55-200 CGG repeats and may exhibit mild or late-onset symptoms, or they may remain asymptomatic throughout their lives [53]. These unstable premutations can expand in subsequent generations, increasing the risk of developing a full mutation and the associated disorder, making genetic screening crucial for early detection, management, and informed family planning.

Anticipation

Anticipation is a phenomenon observed in many TRDs where the disease severity increases and the age of onset decreases in successive generations [2]. This occurs because repeat expansions tend to increase in length when passed from parent to child, often more prominently through either the maternal or paternal germline. For example, in myotonic dystrophy type 1, the CTG repeat expansion in the DMPK gene frequently increases in size when transmitted through the maternal line, leading to earlier and more severe disease manifestations in the offspring [54]. In contrast, in HD, CAG repeats typically expand more through the paternal line, resulting in a more severe and earlier onset [55]. Although anticipation is a key factor in genetic counseling and management, further studies are needed to fully understand its mechanisms. Additionally, while expansion is common, repeat contractions can also occur, reducing repeat length in subsequent generations [56].

Interruptions and repeat motif changes

In some TRDs, the repeat sequence is not purely repetitive and may contain interruptions, or variations within the repeat sequence that can stabilize the repeat tract and reduce further expansion [57]. For example, in SCA1, CAA interruptions within the CAG repeat sequence are associated with milder disease due to their stabilizing effect on the repeat expansion [58]. Conversely, in HD, CAA interruptions are present in most cases, and the loss of interruptions (LOI) can lead to a more aggressive disease course [59]. LOI in HD can result in the formation of more toxic RNA hairpin structures, contributing to a more aggressive disease course even at lower repeat numbers, such as 36 to 39, or even at 35 or fewer repeats [60]. Additionally, in disorders related to expansions in the RFC1 gene, the most common pathogenic repeat expansion is the AAGGG motif [61], replacing the normal AAAAG sequence, but other motifs like ACAGG, AGAGG, and others can also occur, influencing the clinical presentation and disease severity [62]. Understanding sequence composition changes in TRDs is crucial for accurate genetic diagnosis and insights into the variability of clinical outcomes. LRS will be essential for elucidating these underlying mechanisms.

Somatic instability

Recent scientific advancements have significantly broadened our understanding of somatic mutations in TRDs [63]. Somatic instability, where repeat expansions continue to grow in certain tissues after birth, is a critical factor in the progression and severity of many TRDs [64-66]. These mutations are particularly prevalent in tissues such as the brain and muscles, where repeat expansions can accumulate to much higher levels than those found in germline cells. This ongoing expansion can exacerbate disease symptoms over time, contributing to variability in clinical outcomes among individuals with the same genetic mutation. Studies have shown that somatic instability is influenced by factors such as DNA repair pathways, with certain genetic variants either promoting or inhibiting further expansions [63]. This emerging knowledge highlights the importance of considering somatic mutations when assessing disease prognosis and developing targeted therapies. Despite advances in LRS and other genomic technologies, detecting and fully understanding these somatic changes remains challenging, underscoring the need for further technological improvements.

Cutting-edge research on therapeutics

Antisense oligonucleotides and RNA interference

Recent advances in therapeutics for TRDs offer new hope for managing these complex disorders. Among the most promising approaches are antisense oligonucleotides (ASOs) which are attracting attention as a safe and versatile tool for modulating RNA transcripts [67]. ASOs are short, synthetic nucleic acids that bind to specific RNA sequences, allowing precise control over gene expression. The success of U.S. Food and Drug Administration (FDA)-approved ASO drugs, such as nusinersen for spinal muscular atrophy [68] and tofersen for superoxide dismutase 1 (SOD1)-related amyotrophic lateral sclerosis (ALS) [69], has paved the way for ASO development in TRDs.

ASOs operate through several mechanisms [70]. One key approach is allele-specific knockdown, where ASOs selectively degrade mutant RNA transcripts while sparing the wild-type allele [71]. This strategy is particularly promising for TRDs driven by toxic GOF mechanisms, such as HD. However, developing allele-specific ASOs presents challenges, including the need to precisely differentiate between the mutant and wild-type alleles, which often differ by only a single nucleotide. Ensuring specificity without off-target effects is crucial, as unintended knockdown of the wild-type allele could result in undesirable side effects.

Another approach is allele-nonspecific knockdown, which targets both mutant and wild-type alleles to reduce overall gene expression [2,72]. This strategy is generally easier to develop than allele-specific knockdown since it does not require distinguishing between the mutant and normal alleles. However, the key challenge is achieving the right level of knockdown, as excessive reduction could lead to insufficient normal protein levels and potential side effects. Fine-tuning knockdown efficiency is crucial for therapeutic success.

Splice modulation is one of the most powerful and unique applications of ASO technology, enabling the correction of RNA splicing defects that other therapies cannot address [73]. This approach may be particularly useful for TRDs caused by aberrant splicing, such as those involving repeat expansions in UTRs or introns. Notably, most of the successful FDA-approved ASOs for rare diseases, such as nusinersen [68], milasen [74], and atipeksen [75], are based on splice modulation mechanisms. For instance, in FXS, ASOs can correct the splicing of the FMR1 gene, potentially restoring normal protein levels and alleviating symptoms [76]. The ability to precisely target and modulate splicing is a distinct advantage of ASO technology, offering therapeutic potential for a range of TRDs where splicing errors contribute to disease pathology.

Beyond these traditional methods, ASOs are being explored for novel strategies like blocking R-loop formation or inhibiting RAN translation. These innovative approaches could block R-loop formation in the FMR1 gene [77] or reduce RAN translation fragile X protein synthesis [78]. Further research is needed to determine whether these approaches can be effectively applied to a broader range of TRDs, potentially expanding their therapeutic impact.

Gene editing technologies

Recent advancements in gene editing technologies, particularly CRISPR/Cas9 [79], base editors [80], and prime editors [81], have opened new avenues for treating genetic disorders. Recently, base editors have shown promise in treating spinal muscular atrophies [82]. These approaches can be applied to tackle TRDs. CRISPR/Cas9 is being explored to reduce the number of repeat expansions in genes like C9ORF72 to mitigate the effects of ALS and frontotemporal dementia [83]. Base editors are now being investigated to introduce CAA interruptions in the CAG repeats of HD, potentially stabilizing the repeat expansion [84]. Prime editors, offering highly precise DNA modifications [85], are being researched to directly remove or correct expanded repeats in TRDs, potentially addressing the root cause of these disorders. While these technologies hold great promise, further research is essential to refine their application and ensure their safety and effectiveness in clinical settings.

Novel therapeutic approaches

Novel therapeutic strategies are being developed to address the molecular disruptions caused by repeat expansions in TRDs. Small molecule therapies [86] aim to target disrupted pathways by modulating protein function, enhancing the clearance of toxic aggregates, or stabilizing RNA structures to mitigate the pathogenic effects of expanded repeats. Meanwhile, gene therapy [87] and stem cell therapy [88] offer the potential to replace or repair damaged cells and restore normal function. Advances in viral vector technology and stem cell engineering are bringing these approaches closer to becoming viable therapeutic options, potentially transforming the treatment landscape for TRDs in the near future.

Conclusion

TRDs, where the expansion of repetitive DNA sequences leads to diverse clinical manifestations and outcomes, exemplify the complexity of genetic diseases. Advances in molecular genetics, particularly diagnostic tools like LRS, have significantly enhanced our ability to identify and understand these disorders. Despite this progress, a clear understanding of the mechanisms behind disease onset and progression remains elusive. Emerging therapies, including ASO, RNA interference, and gene editing technologies, offer promising therapeutic options, yet careful consideration of safety and efficacy is required during their development. As research continues to uncover the complexities of TRDs, it will be essential to refine these therapeutic strategies to improve patient outcomes and provide hope for those affected by these complex conditions.

Notes

Conflicts of Interest

Jangsup Moon has been an Associate Editor of encephalitis since October 2020. He was not involved in the review process of this original article. No other potential conflict of interest relevant to this article was reported.

References

1. Depienne C, Mandel JL. 30 years of repeat expansion disorders: what have we learned and what are the remaining challenges? Am J Hum Genet 2021;108:764–785.
2. Paulson H. Repeat expansion diseases. Handb Clin Neurol 2018;147:105–123.
3. López Castel A, Cleary JD, Pearson CE. Repeat instability as the basis for human diseases and as a potential target for therapy. Nat Rev Mol Cell Biol 2010;11:165–170.
4. Gall-Duncan T, Sato N, Yuen RK, Pearson CE. Advancing genomic technologies and clinical awareness accelerates discovery of disease-associated tandem repeat sequences. Genome Res 2022;32:1–27.
5. Usdin K, House NC, Freudenreich CH. Repeat instability during DNA repair: insights from model systems. Crit Rev Biochem Mol Biol 2015;50:142–167.
6. Malik I, Kelley CP, Wang ET, Todd PK. Molecular mechanisms underlying nucleotide repeat expansion disorders. Nat Rev Mol Cell Biol 2021;22:589–607.
7. Lamkin M, Gymrek M. The emerging role of tandem repeats in complex traits. Nat Rev Genet 2024;25:452–453.
8. Swinnen B, Robberecht W, Van Den Bosch L. RNA toxicity in non-coding repeat expansion disorders. EMBO J 2020;39e101112.
9. Lee JM, Ramos EM, Lee JH, et al. CAG repeat expansion in Huntington disease determines age at onset in a fully dominant fashion. Neurology 2012;78:690–695.
10. Kraan CM, Godler DE, Amor DJ. Epigenetics of fragile X syndrome and fragile X-related disorders. Dev Med Child Neurol 2019;61:121–127.
11. Mila M, Alvarez-Mora MI, Madrigal I, Rodriguez-Revenga L. Fragile X syndrome: an overview and update of the FMR1 gene. Clin Genet 2018;93:197–205.
12. Liquori CL, Ricker K, Moseley ML, et al. Myotonic dystrophy type 2 caused by a CCTG expansion in intron 1 of ZNF9. Science 2001;293:864–867.
13. Matsuura T, Yamagata T, Burgess DL, et al. Large expansion of the ATTCT pentanucleotide repeat in spinocerebellar ataxia type 10. Nat Genet 2000;26:191–194.
14. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 2011;72:245–256.
15. Dolzhenko E, van Vugt JJ, Shaw RJ, et al. Detection of long repeat expansions from PCR-free whole-genome sequence data. Genome Res 2017;27:1895–1903.
16. Logsdon GA, Vollger MR, Eichler EE. Long-read human genome sequencing and its applications. Nat Rev Genet 2020;21:597–614.
17. Sone J, Mitsuhashi S, Fujita A, et al. Long-read sequencing identifies GGC repeat expansions in NOTCH2NLC associated with neuronal intranuclear inclusion disease. Nat Genet 2019;51:1215–1221.
18. Nakamura H, Doi H, Mitsuhashi S, et al. Long-read sequencing identifies the pathogenic nucleotide repeat expansion in RFC1 in a Japanese case of CANVAS. J Hum Genet 2020;65:475–480.
19. Warner JP, Barron LH, Goudie D, et al. A general method for the detection of large CAG repeat expansions by fluorescent PCR. J Med Genet 1996;33:1022–1026.
20. Schröder C, Horsthemke B, Depienne C. GC-rich repeat expansions: associated disorders and mechanisms. Med Genet 2022;33:325–335.
21. Cagnoli C, Michielotto C, Matsuura T, et al. Detection of large pathogenic expansions in FRDA1, SCA10, and SCA12 genes using a simple fluorescent repeat-primed PCR assay. J Mol Diagn 2004;6:96–100.
22. Ciotti P, Di Maria E, Bellone E, Ajmar F, Mandich P. Triplet repeat primed PCR (TP PCR) in molecular diagnostic testing for Friedreich ataxia. J Mol Diagn 2004;6:285–289.
23. Cleary EM, Pal S, Azam T, et al. Improved PCR based methods for detecting C9orf72 hexanucleotide repeat expansions. Mol Cell Probes 2016;30:218–224.
24. Chen L, Hadd A, Sah S, et al. An information-rich CGG repeat primed PCR that detects the full range of fragile X expanded alleles and minimizes the need for southern blot analysis. J Mol Diagn 2010;12:589–600.
25. Tarleton J. Detection of FMR1 trinucleotide repeat expansion mutations using Southern blot and PCR methodologies. Methods Mol Biol 2003;217:29–39.
26. Hübers A, Marroquin N, Schmoll B, et al. Polymerase chain reaction and Southern blot-based analysis of the C9orf72 hexanucleotide repeat in different motor neuron diseases. Neurobiol Aging 2014;35:1214.
27. Buchman VL, Cooper-Knock J, Connor-Robson N, et al. Simultaneous and independent detection of C9ORF72 alleles with low and high number of GGGGCC repeats using an optimised protocol of Southern blot hybridisation. Mol Neurodegener 2013;8:12.
28. Bahlo M, Bennett MF, Degorski P, Tankard RM, Delatycki MB, Lockhart PJ. Recent advances in the detection of repeat expansions with short-read next-generation sequencing. F1000Res 2018;7(F1000 Faculty Rev):736.
29. Dolzhenko E, Deshpande V, Schlesinger F, et al. ExpansionHunter: a sequence-graph-based tool to analyze variation in short tandem repeat regions. Bioinformatics 2019;35:4754–4756.
30. Dolzhenko E, Bennett MF, Richmond PA, et al. ExpansionHunter Denovo: a computational method for locating known and novel repeat expansions in short-read sequencing data. Genome Biol 2020;21:102.
31. Dashnow H, Lek M, Phipson B, et al. STRetch: detecting and discovering pathogenic short tandem repeat expansions. Genome Biol 2018;19:121.
32. Mousavi N, Shleizer-Burko S, Yanicky R, Gymrek M. Profiling the genome-wide landscape of tandem repeat expansions. Nucleic Acids Res 2019;47e90.
33. Chintalaphani SR, Pineda SS, Deveson IW, Kumar KR. An update on the neurological short tandem repeat expansion disorders and the emergence of long-read sequencing diagnostics. Acta Neuropathol Commun 2021;9:98.
34. Liu Q, Tong Y, Wang K. Genome-wide detection of short tandem repeat expansions by long-read sequencing. BMC Bioinformatics 2020;21(Suppl 21):542.
35. Gilpatrick T, Lee I, Graham JE, et al. Targeted nanopore sequencing with Cas9-guided adapter ligation. Nat Biotechnol 2020;38:433–438.
36. Leitner K, Motheramgari K, Borth N, Marx N. Nanopore Cas9-targeted sequencing enables accurate and simultaneous identification of transgene integration sites, their structure and epigenetic status in recombinant Chinese hamster ovary cells. Biotechnol Bioeng 2023;120:2403–2418.
37. La Spada AR, Taylor JP. Repeat expansion disease: progress and puzzles in disease pathogenesis. Nat Rev Genet 2010;11:247–258.
38. Bates G. Huntingtin aggregation and toxicity in Huntington’s disease. Lancet 2003;361:1642–1644.
39. Petrakis S, Raskó T, Russ J, et al. Identification of human proteins that modify misfolding and proteotoxicity of pathogenic ataxin-1. PLoS Genet 2012;8e1002897.
40. Evers MM, Toonen LJ, van Roon-Mom WM. Ataxin-3 protein and RNA toxicity in spinocerebellar ataxia type 3: current insights and emerging therapeutic strategies. Mol Neurobiol 2014;49:1513–1531.
41. Latouche M, Fragner P, Martin E, et al. Polyglutamine and polyalanine expansions in ataxin7 result in different types of aggregation and levels of toxicity. Mol Cell Neurosci 2006;31:438–445.
42. Brais B, Bouchard JP, Xie YG, et al. Short GCG expansions in the PABP2 gene cause oculopharyngeal muscular dystrophy. Nat Genet 1998;18:164–167.
43. Zu T, Gibbens B, Doty NS, et al. Non-ATG-initiated translation directed by microsatellite expansions. Proc Natl Acad Sci U S A 2011;108:260–265.
44. Cleary JD, Ranum LP. Repeat-associated non-ATG (RAN) translation in neurological disease. Hum Mol Genet 2013;22(R1):R45–R51.
45. Sznajder ŁJ, Swanson MS. Short tandem repeat expansions and RNA-mediated pathogenesis in myotonic dystrophy. Int J Mol Sci 2019;20:3365.
46. Usdin K, Kumari D. Repeat-mediated epigenetic dysregulation of the FMR1 gene in the fragile X-related disorders. Front Genet 2015;6:192.
47. Sadri-Vakili G, Cha JH. Mechanisms of disease: Histone modifications in Huntington’s disease. Nat Clin Pract Neurol 2006;2:330–338.
48. De Biase I, Chutake YK, Rindler PM, Bidichandani SI. Epigenetic silencing in Friedreich ataxia is associated with depletion of CTCF (CCCTC-binding factor) and antisense transcription. PLoS One 2009;4e7914.
49. Ginno PA, Lott PL, Christensen HC, Korf I, Chédin F. R-loop formation is a distinctive characteristic of unmethylated human CpG island promoters. Mol Cell 2012;45:814–825.
50. Groh M, Lufino MM, Wade-Martins R, Gromak N. R-loops associated with triplet repeat expansions promote gene silencing in Friedreich ataxia and fragile X syndrome. PLoS Genet 2014;10e1004318.
51. Loomis EW, Sanz LA, Chédin F, Hagerman PJ. Transcription-associated R-loop formation across the human FMR1 CGG-repeat region. PLoS Genet 2014;10e1004294.
52. McNeil SM, Novelletto A, Srinidhi J, et al. Reduced penetrance of the Huntington's disease mutation. Hum Mol Genet 1997;6:775–779.
53. Jacquemont S, Leehey MA, Hagerman RJ, Beckett LA, Hagerman PJ. Size bias of fragile X premutation alleles in late-onset movement disorders. J Med Genet 2006;43:804–809.
54. De Temmerman N, Sermon K, Seneca S, et al. Intergenerational instability of the expanded CTG repeat in the DMPK gene: studies in human gametes and preimplantation embryos. Am J Hum Genet 2004;75:325–329.
55. Ranen NG, Stine OC, Abbott MH, et al. Anticipation and instability of IT-15 (CAG)n repeats in parent-offspring pairs with Huntington disease. Am J Hum Genet 1995;57:593–602.
56. Pâques F, Leung WY, Haber JE. Expansions and contractions in a tandem repeat induced by double-strand break repair. Mol Cell Biol 1998;18:2045–2054.
57. Rajan-Babu IS, Dolzhenko E, Eberle MA, Friedman JM. Sequence composition changes in short tandem repeats: heterogeneity, detection, mechanisms and clinical implications. Nat Rev Genet 2024;25:476–499.
58. Sobczak K, Krzyzosiak WJ. Patterns of CAG repeat interruptions in SCA1 and SCA2 genes in relation to repeat instability. Hum Mutat 2004;24:236–247.
59. Findlay Black H, Wright GE, Collins JA, et al. Frequency of the loss of CAA interruption in the HTT CAG tract and implications for Huntington disease in the reduced penetrance range. Genet Med 2020;22:2108–2113.
60. Murmann AE, Patel M, Jeong SY, Bartom ET, Jennifer Morton A, Peter ME. The length of uninterrupted CAG repeats in stem regions of repeat disease associated hairpins determines the amount of short CAG oligonucleotides that are toxic to cells through RNA interference. Cell Death Dis 2022;13:1078.
61. Wang Y, Wang J, Yan Z, et al. RFC1 AAGGG pentanucleotide repeats form parallel G-quadruplex: structural implications for aberrant molecular cascades in CANVAS. bioRxiv [Preprint] 2023 Aug 6. https://doi.org/10.1101/2023.08.06.552146.
62. Abdi MH, Zamiri B, Pazuki G, Sardari S, Pearson CE. Pathogenic CANVAS-causing but not nonpathogenic RFC1 DNA/RNA repeat motifs form quadruplex or triplex structures. J Biol Chem 2023;299:105202.
63. Schmidt MH, Pearson CE. Disease-associated repeat instability and mismatch repair. DNA Repair (Amst) 2016;38:117–126.
64. Long A, Napierala JS, Polak U, et al. Somatic instability of the expanded GAA repeats in Friedreich’s ataxia. PLoS One 2017;12e0189990.
65. Ashizawa T, Dubel JR, Harati Y. Somatic instability of CTG repeat in myotonic dystrophy. Neurology 1993;43:2674–2678.
66. Swami M, Hendricks AE, Gillis T, et al. Somatic expansion of the Huntington’s disease CAG repeat in the brain is associated with an earlier age of disease onset. Hum Mol Genet 2009;18:3039–3047.
67. Dhuri K, Bechtold C, Quijano E, et al. Antisense oligonucleotides: an emerging area in drug discovery and development. J Clin Med 2020;9:2004.
68. Corey DR. Nusinersen, an antisense oligonucleotide drug for spinal muscular atrophy. Nat Neurosci 2017;20:497–499.
69. Miller TM, Cudkowicz ME, Genge A, et al. Trial of antisense oligonucleotide tofersen for SOD1 ALS. N Engl J Med 2022;387:1099–1110.
70. Thakur S, Sinhari A, Jain P, Jadhav HR. A perspective on oligonucleotide therapy: approaches to patient customization. Front Pharmacol 2022;13:1006304.
71. Helm J, Schöls L, Hauser S. Towards personalized allele-specific antisense oligonucleotide therapies for toxic gain-of-function neurodegenerative diseases. Pharmaceutics 2022;14:1708.
72. Godinho BM, Malhotra M, O’Driscoll CM, Cryan JF. Delivering a disease-modifying treatment for Huntington’s disease. Drug Discov Today 2015;20:50–64.
73. Aartsma-Rus A, Garanto A, van Roon-Mom W, et al. Consensus guidelines for the design and in vitro preclinical efficacy testing N-of-1 exon skipping antisense oligonucleotides. Nucleic Acid Ther 2023;33:17–25.
74. Kim J, Hu C, Moufawad El Achkar C, et al. Patient-customized oligonucleotide therapy for a rare genetic disease. N Engl J Med 2019;381:1644–1652.
75. Kim J, Woo S, de Gusmao CM, et al. A framework for individualized splice-switching oligonucleotide therapy. Nature 2023;619:828–836.
76. Shah S, Sharp KJ, Raju Ponny S, et al. Antisense oligonucleotide rescue of CGG expansion-dependent FMR1 mis-splicing in fragile X syndrome restores FMRP. Proc Natl Acad Sci U S A 2023;120e2302534120.
77. Derbis M, Kul E, Niewiadomska D, et al. Short antisense oligonucleotides alleviate the pleiotropic toxicity of RNA harboring expanded CGG repeats. Nat Commun 2021;12:1265.
78. Rodriguez CM, Wright SE, Kearse MG, et al. A native function for RAN translation and CGG repeats in regulating fragile X protein synthesis. Nat Neurosci 2020;23:386–397.
79. Savić N, Schwank G. Advances in therapeutic CRISPR/Cas9 genome editing. Transl Res 2016;168:15–21.
80. Porto EM, Komor AC, Slaymaker IM, Yeo GW. Base editing: advances and therapeutic opportunities. Nat Rev Drug Discov 2020;19:839–859.
81. Anzalone AV, Randolph PB, Davis JR, et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019;576:149–157.
82. Arbab M, Matuszek Z, Kray KM, et al. Base editing rescue of spinal muscular atrophy in cells and in mice. Science 2023;380eadg6518.
83. Meijboom KE, Abdallah A, Fordham NP, et al. CRISPR/Cas9-mediated excision of ALS/FTD-causing hexanucleotide repeat expansion in C9ORF72 rescues major disease mechanisms in vivo and in vitro. Nat Commun 2022;13:6286.
84. Choi DE, Shin JW, Zeng S, et al. Base editing strategies to convert CAG to CAA diminish the disease-causing mutation in Huntington’s disease. Elife 2024;12:RP89782.
85. Chen PJ, Liu DR. Prime editing for precise and highly versatile genome manipulation. Nat Rev Genet 2023;24:161–177.
86. Nakamori M, Mochizuki H. Targeting expanded repeats by small molecules in repeat expansion disorders. Mov Disord 2021;36:298–305.
87. Martier R, Liefhebber JM, García-Osta A, et al. Targeting RNA-mediated toxicity in C9orf72 ALS and/or FTD by RNAi-based gene therapy. Mol Ther Nucleic Acids 2019;16:26–37.
88. Wong GK, Chiu AT. Gene therapy, gene targeting and induced pluripotent stem cells: applications in monogenic disease treatment. Biotechnol Adv 2011;29:1–10.

Article information Continued

Table 1

Classification of tandem repeat disorders

Classification criteria Category Example Key feature
Location of repeat sequence Coding region HD, SCA1, 2, 3, 6, 7, 17 (CAG) Produces toxic proteins; typically affects the nervous system
Introns Friedreich’s ataxia (GAA), SCA10 (ATTCT), SCA31 (TGGAA), SCA36 (GGCCTG), C9ORF72-related ALS/FTD (GGGGCC) Disrupts RNA processing or leads to toxic RNA/protein products
5’ UTR FXS (CGG), NIID (CGG) Leads to gene silencing (FXS) or toxic protein production via RAN translation (NIID)
3’ UTR Myotonic dystrophy type 1 (CTG) Causes RNA toxicity, sequestering RNA-binding proteins
Type of repeat motif Trinucleotide repeats HD (CAG), FXS, NIID (CGG), SCA1, 2, 3, 6, 7, 17 (CAG) Most common, often leads to severe neurodegenerative conditions
Tetranucleotide repeats Myotonic dystrophy type 2 (CCTG) Associated with muscular dystrophies; typically located in introns
Pentanucleotide repeats SCA10 (ATTCT), SCA31 (TGGAA) Often results in ataxias; typically located in introns
Hexanucleotide repeats C9ORF72-related ALS/FTD (GGGGCC), SCA36 (GGCCTG) Linked to neurodegeneration and dementia; typically located in introns

HD, Huntington's disease; SCA, spinocerebellar ataxias; ALS, amyotrophic lateral sclerosis; FTD, frontotemporal dementia; UTR, untranslated region; FXS, fragile X syndrome; NIID, neuronal intranuclear inclusion disease.

Table 2

Diagnostic techniques for tandem repeat disorders

Diagnostic technique Method Advantages Limitations
Conventional PCR Fragment length analysis Quick, widely available Limited to small/moderate expansions; difficult to conduct with GC-rich repeats
Southern blotting DNA fragment hybridization Gold standard for large expansions Time-consuming, requires large DNA samples
Repeat-primed PCR (RP-PCR) PCR with repeat-binding primers Sensitive for detecting expansions Does not provide precise sizing
Short-read NGS High-throughput sequencing Comprehensive genomic analysis Fragmentation of large repeats, incomplete data
Long-read sequencing (LRS) Sequencing long DNA fragments Accurate sizing of large, complex repeats; potential for methylation analysis High cost, limited accessibility
Nanopore Cas9-targeted sequencing (nCATS) Cas9-assisted targeted sequencing Precise, targeted analysis; cost-effective for targeted regions Complex setup, specialized equipment needed

PCR, polymerase chain reaction; RP, repeat-primed.