Abstract
Microsatellite expansions cause more than 40 neurological disorders, including Huntington's disease, myotonic dystrophy, and C9ORF72 amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD). These repeat expansion mutations can produce repeat-associated non-ATG (RAN) proteins in all three reading frames, which accumulate in disease-relevant tissues. There has been considerable interest in RAN protein products and their downstream consequences, particularly for the dipeptide proteins found in C9ORF72 ALS/FTD. Understanding how RAN translation occurs, what cellular factors contribute to RAN protein accumulation, and how these proteins contribute to disease should lead to a better understanding of the basic mechanisms of gene expression and human disease.
Keywords: translation, translation initiation, trinucleotide repeat disease, Huntington's disease, myotonic dystrophy, amyotrophic lateral sclerosis (ALS) (Lou Gehrig disease), C9ORF72 ALS/FTD, mouse models, nucleocytoplasmic transport, RAN translation, spinocerebellar ataxia
Introduction and background
More than 40 different neurological diseases are caused by unstable microsatellite sequences (e.g. CAG, CCG, or G4C2) that are repeated multiple times at specific human genetic loci. For more than 25 years, research into these disorders has focused on the anticipated effects of the expansion mutations based on whether the mutations lie within or outside annotated protein-coding regions. In 2011, Zu et al. (1) discovered that repeat expansion mutations can produce a set of unexpected mutant proteins in multiple reading frames without the canonical AUG initiation codon. The discoveries of repeat-associated non-ATG (RAN)2 translation (1) and that repeat expansion mutations are often bidirectionally transcribed (2, 3) mean that a single repeat expansion mutation can produce mutant proteins in all three reading frames from both sense and antisense transcripts (4–6). The discovery that expansion mutations express proteins without a canonical AUG-initiation codon raises mechanistic questions about how RAN proteins are initiated. Given the prevalence of repeats in the human genome (7, 8), RAN translation may increase the diversity and function of the proteome. This Minireview will discuss mechanistic insights and disease implications of RAN translation.
The discovery of RAN translation added a new twist to the already complex field of repeat-expansion disorders (9–11). Unlike traditional mutations, expansion mutations are unstable and can change in length between generations (intergenerational instability) as well as within an individual (somatic instability) (12, 13). Intergenerational instability can cause anticipation or a decrease in age of onset and an increase in disease severity from one generation to the next (14, 15). Disease mechanisms in these disorders have traditionally been categorized based on the location of the expansion mutation within the corresponding gene. For example, expanded CAG repeats located within traditional protein-coding open reading frames (ORFs) have been considered to be caused by gain-of-function (GOF) effects of the corresponding mutant expansion protein (16) (e.g. Huntington's disease (HD)). In contrast, expanded mutations located in noncoding regions have been considered protein loss-of-function (LOF) (e.g. fragile X syndrome) or RNA GOF disorders (2, 9, 10) (e.g. myotonic dystrophy type 1 (DM1) or type 2 (DM2)). These “noncoding” expansion RNAs accumulate as nuclear foci, which sequester RNA-binding proteins (RBPs) and lead to a loss of their normal function (5, 6). For example, in DM1 and DM2, CUG and CCUG expansion RNAs sequester MBNL proteins from their normal splicing targets, and MBNL LOF leads to alternative splicing dysregulation (7–10). Although there is substantial evidence for both protein and RNA GOF mechanisms, the pathology and symptoms of these diseases are not fully explained by these mechanisms. For example, the restricted disease-specific populations of vulnerable neurons in the various diseases do not correlate well with the much broader CNS expression of the various expansion mutations. RAN translation and its downstream consequences offer new insights into disease mechanisms and more importantly new avenues for therapeutic interventions.
Translation and translational initiation
Although RAN proteins have been identified in a growing number of diseases (Table 1), very little is known about the underlying mechanisms of RAN translation. It is likely that RAN translation shares at least some features in common with canonical and/or internal-ribosome entry (IRES) initiation (Fig. 1). Canonical translation initiation is a complex process involving the stepwise activities of multiple protein complexes, including the following: 1) the recognition of the 5′-methyl-7-guanosine cap; 2) the recruitment of the 43S preinitiation complex, which scans through the 5′UTR of an mRNA until an AUG codon in the proper context pairs with the CAU anti-codon loop of the Met-tRNAi; 3) eukaryotic initiation factor 2 (eIF2) then hydrolyzes its bound GTP, and the 60S ribosomal subunit is recruited; and 4) the majority of eIFs are then released, and translation elongation begins (see Fig. 1A) (17–20).
Table 1.
Microsatellite repeats and their sense and antisense protein products
| Disease | Gene | Repeat (sense·antisense) | Sense protein motif(s) | Antisense protein motif(s) | Evidence in patients for |
|---|---|---|---|---|---|
| DM1 | DMPK | CTG·CAG | Leu, Cys, Ala | Gln, Ala, Ser | GlnAS (1) |
| DM2 | CNBP | CCTG·CAGG | (Leu–Pro–Ala–Cys) | (Gln–Ala–Gly–Arg) | Leu–Pro–Ala–CysS (63) |
| Gln–Ala–Gly–ArgAS (63) | |||||
| FECD | TCF4 | CTG·CAG | Leu, Cys, Ala | Gln, Ala, Ser | CysS (70) |
| FXTAS | FMR1 | CGG·CCG | Gly, Ala, Arg | Ala, Arg, Pro | GlyS (32), AlaAS (46), ProAS (46) |
| C9orf72 ALS/FTD | C9ORF72 | GGGGCC·GGCCCC | Gly–Pro, Gly–Ala, Gly–Arg | Gly–Pro, Pro–Ala, Pro–Arg | Gly–ProS/AS (33–35, 124, 191), Gly–AlaS (33–35, 191), Gly–ArgS (33–35, 123, 191) |
| Pro–AlaAS (35, 124), Pro–ArgAS (33–35, 123, 124, 191) | |||||
| SCA8 | ATXN8 | CAG·CTG | Gln, Ala, Ser | Leu, Cys, Ala | AlaAS (1), SerS (30) |
| SCA31 | BEAN | TGGAA·TTCCA | (Trp–Asn–Gly–Met–Glu) | (Ser–Ile–Pro–Phe–His) | Trp–Asn–Gly–Met–Glu) (37) |
| SCA37 | DAB1 | ATTTC·GAAAT | (Phe–Ile–Ser–Phe–His) | (Asn–Glu–Met–Lys STOP) | NDa |
a ND means not determined.
Figure 1.
Translation initiation mechanisms. A, canonical translation initiation involves the binding of the 5′ mRNA cap (eIF4F consisting of eIF4G, eIF4E, eIF4A, and eIF4B) and mRNA poly(A) tail (PABP), unwinding of the mRNA by the helicase activity of eIF4A, and recruitment of 43S complex (eIF5, eIF3, eIF2, and 40S ribosomal subunit) followed by scanning of the mRNA 5′ UTR in a 5′ to 3′ direction by the engaged 43S complex. Recognition of the initiation codon results in the 48S initiation complex formation and displacement of several initiation factors. B, internal ribosome entry site initiation occurs in a cap-independent manner from multiple viral and cellular RNA sequences that involve the recruitment of the cellular 43S ribosomal complex to internal sites with the RNA by specific initiation translation factors (ITAFs). Depending upon the viral IRES group (I–IV), all, some, or none of the typical canonical translation factors, including the initiation codon, may be required for translation initiation. C, repeat-associated non-ATG translation initiation is a repeat-length–dependent process that allows for initiation at noncanonical codons either within or adjacent to the expanded repeat tract. Evidence from the FXTAS CGG repeats and some reports for G4C2 repeats support a requirement for 5′ mRNA cap, eIF4E, and eIF4A suggesting cap-dependent and scanning mechanisms. However, other reports support cap-independent translation initiation mechanisms more similar to IRES initiation. The identity and requirement of other cellular initiation factors involved in RAN translation have yet to be determined.
Although the majority of mRNAs are initiated by canonical scanning and AUG initiation, alternative initiation pathways have also been described. For example, many viral and a growing number of cellular mRNAs have been shown to use IRESs, complex RNA structures that direct ribosomal and eIF recruitment directly, for initiation (see Fig. 1B) (21). Close-cognate start codons, which differ from the AUG start codons by one nucleotide (22) and use the Met-tRNAi and methionine as the initiating amino acid, can also be used in mammalian cells. Starck et al. (23) showed that nonrepetitive CUG-codons initiate translation of the peptide-loaded major histocompatibility complex class I molecules (24) with Leu-tRNALeu as the initiating tRNA. Although these alternative translation initiation mechanisms may share some similarities with RAN translation, RAN translation differs in that it is associated with repeat expansion mutations, can occur in the absence of close-cognate initiation codons, and can produce proteins in all three reading frames (Fig. 1C). Because RAN translation can also occur across repeats located in traditional ATG-initiated ORFs (1, 25), a single repeat expansion transcript may undergo ATG-initiated translation in one frame and RAN translation in the other two reading frames.
Discovery and initial characterization of RAN translation
The first evidence for RAN translation arose from studies attempting to separate the RNA and protein GOF effects in spinocerebellar ataxia type 8 (SCA8) (1). SCA8 is a dominantly inherited, slowly progressive neurodegenerative disorder caused by a CTG·CAG repeat expansion (26, 27). Prior to the discovery of RAN translation, SCA8 was the only disorder in which both RNA and protein gain-of-function disease mechanisms had been implicated (28). Bidirectional transcription of the SCA8 expansion mutation produces CUG expansion transcripts that form RNA foci (29) and a CAG expansion transcript expressed in the opposite direction that encodes a nearly pure ATG-initiated polyglutamine (poly(Gln)) expansion protein (28). Surprisingly, mutating the ATG-initiation codon did not prevent expression of the poly(Gln) expansion protein (1). Zu et al. (1) demonstrated that CAG expansions lacking an ATG initiation codon can produce homopolymeric expansion proteins in all three reading frames (i.e. poly(Gln), poly(Ser), and poly(Ala)). Additional experiments showed the following: 1) no evidence of RNA editing that could have introduced a start codon; 2) frameshifting was not required for protein expression in multiple frames; 3) translation appeared by MS data to begin within the repeat itself, at least for the poly(Ala) frame; 4) multiple RAN products from different frames can be produced in the same cell; 5) RAN translation is repeat length-dependent and favored by RNAs that form secondary structures; and 6) RAN proteins were shown to be toxic to cells. Subsequent analyses showed in vivo evidence that a novel SCA8 RAN poly(Ala) protein accumulates in cerebellar Purkinje cells in SCA8 mice and human autopsy tissue. More recently, Ayhan et al. (30) demonstrated that a novel RAN poly(Ser) protein expressed from ATXN8 CAG expansion transcripts accumulates in white matter regions in SCA8 mouse and human cerebella. Additionally, these authors showed that steady-state levels of SCA8 poly(Ser) and other RAN but not ATG-initiated proteins were reduced by knockdown of the initiation factor eIF3F. A novel CAG-encoded RAN poly(Gln) protein was also detected in myotonic dystrophy type 1 (DM1) mouse and human tissues, including patient myoblasts, skeletal muscle, and blood (1). Additionally, in both humans and mice, poly(Gln) aggregates were shown to co-localize with caspase-8 (1), an early indicator of poly(Gln)-induced apoptosis (31). The 2011 discovery of RAN translation in SCA8 and DM1 generated substantial interest by the scientific community into both the mechanisms of this novel type of promiscuous translation and the role of RAN proteins in neurodegenerative disease.
Since the discovery of RAN translation in SCA8 and DM1 (1), RAN proteins have been reported in fragile X tremor ataxia syndrome (FXTAS) (32), C9ORF72 ALS/frontotemporal dementia (C9-ALS/FTD) (33–35), Huntington's disease (25), and spinocerebellar ataxia type 31 (37). RAN translation has now been shown to occur across several different types of repeat motifs (CAG·CTG, CGG·CCG, G4C2·G2C4, and TG2A2·T2C2A), which share some common themes, including repeat length-dependence and the formation of unusual RNA secondary structures (38–40). Given the potential impact of RAN translation in disease, much of the research on RAN translation has focused on the expression of RAN proteins and the characterization of their downstream consequences. This Minireview will focus on recent discoveries of RAN translation in disease and what is currently known about the mechanisms of RAN translation.
Fragile X tremor ataxia syndrome
Expansion of the FMR1 CGG repeat to between 55 and 200 repeats results in FXTAS, a late-onset disease primarily affecting males that is characterized by tremor, ataxia, parkinsonism, and cognitive decline (41). FXTAS patients have increased expression of the repeat-containing RNA (42, 43) and show ubiquitin-positive inclusions throughout the cerebral cortex, brainstem, and cerebellum (43, 44). Although some studies support the contribution of a toxic RNA GOF mechanism, the discovery and characterization of RAN proteins in FXTAS suggest RAN proteins also contribute to disease.
In 2013, Todd et al. (32) demonstrated that translation of expanded CGG repeats results in the expression of RAN proteins in the polyglycine (FMR–poly(Gly)) and polyalanine (FMR–poly(Ala)) but not polyarginine (FMR–poly(Arg)) reading frames in cell culture. The FMR–poly(Gly) protein has been shown to co-localize with the ubiquitinated inclusions previously reported in FXTAS patient brain samples (32) and in the ovaries of fragile X premutation ovarian insufficiency (FXPOI) patients (45). Krans et al. (46) showed in vitro evidence that polyproline (poly(ProAS)), polyarginine (poly(ArgAS)), and polyalanine (poly(AlaAS)) expansion proteins are expressed across expanded antisense CCG transcripts. Similar to the sense FMR–poly(Gly) protein, both the poly(ProAS) and poly(AlaAS) proteins were shown to accumulate in patient brains (46). Mechanistic studies by Kearse et al. (47) showed translation in both FMR–poly(Gly) and poly(Ala) reading frames depends on a 5′ cap, eIF4E, and eIF4A, suggesting a cap-binding and scanning mode of translation initiation for those reading frames. Similar to RAN translation across a CAG repeat (1), steady-state levels of individual RAN proteins vary by reading frame. When fused with green fluorescent protein (GFP), the GFP–FMR–poly(Gly) fusion protein was observed with as few as 30 repeats, whereas GFP–FMR–poly(Ala) was not detected at lengths below 88 repeats (32). Insertion of stop codons into the upstream region of the FMR–poly(Gly) prevented protein expression, whereas a similar insertion did not prevent FMR–poly(Ala) expression. Additional luciferase experiments show that the FMR–poly(Gly) protein initiates at close-cognate AUG-like codons upstream of the CGG repeat (47), and this initiation occurs independent of the repeat tract itself (47). Taken together, these data suggest that RNA structures independent of the FMR repeat may promote initiation at multiple upstream non-AUG start codons in the poly(Gly) reading frame. Almost half of mammalian mRNAs harbor upstream ORFs (uORFs), many of which initiate from close-cognate start codons (48–52).
In contrast to FMR–poly(Gly) expression and similar to SCA8 poly(Ala) expression (1), mutagenesis experiments suggest the FMR–poly(Ala) frame may initiate from within the repeat expansion (47) as stop codons inserted before the repeat did not prevent expression. Taken together, these results suggest RAN translation can differ mechanistically in different reading frames (53). Translation initiation in the FMR–poly(Gly) frame has more in common with initiation at close cognate uORFs than with the more permissive characteristics of RAN translation observed in the FMR–poly(Ala) frame and in other repeat expansions disorders. RAN translation has been shown to occur in multiple reading frames, even when expansion mutations are located within larger ORFs (e.g. HD) (25). Similarly, RAN translation in the FMR poly(Ala) reading frame occurs along with close-cognate uORF expression in the poly(Gly) frame (1, 25). Taken together, these results highlight the complexity of translation mechanisms at repeat expansion loci.
The role of the FMR–poly(Gly) protein and the ubiquitin-positive inclusions was further characterized using inducible mouse models with 90 CGG repeats (54, 55). Hukema et al. (54) showed that turning off expression of the CGG transgene and therefore FMR–poly(Gly) expression in these mice reduced the number of ubiquitin and FMR–poly(Gly) inclusions at 8 weeks and halted deterioration of eye movement abnormalities suggesting a pathogenic contribution of the FMR–poly(Gly) protein. These mice were more recently used to examine anxiety, motor coordination deficits, and impaired gait, in which ablation of CGG transgene expression rescued behavioral but not motor phenotypes (55). Behavioral features in this model paralleled the formation of intranuclear inclusion in various brain regions (55). More recently, Sellier et al. (56) demonstrated that expression of FMR–poly(Gly) is pathogenic, whereas the sole expression of CGG RNA is not. Poly(Gly) inclusions have also been observed in the ovaries of FXPOI patients as well as in ovaries of older (40 weeks) but not younger (20 weeks) knockin CGG mice (45). These data suggest FMR–poly(Gly) may contribute to premature ovarian insufficiency.
Huntington's disease: RAN translation in an ORF
HD is a relentlessly progressive neurodegenerative disorder characterized by movement abnormalities, cognitive decline, and psychiatric problems (11). Most HD patients have expanded CAG repeats in the 38–55 repeat range and develop symptoms during middle age, but larger repeats (>60 CAG) cause a juvenile onset form of the disease (57). Most research into HD and other poly(Gln) disorders has focused on understanding the toxic effects of the poly(Gln) expansion proteins (58). Although RAN translation had been reported in a number of noncoding disorders, the location of the CAG repeats within canonical open reading frames and their smaller size made it unclear whether these active open reading frames also produce RAN proteins in alternative reading frames. Bañez-Coronel et al. (25) tested whether RAN translation can occur in a polyglutamine disease by examining the most common of these disorders, Huntington's disease. The authors showed that four novel homopolymeric RAN expansion proteins (poly(Ala) and poly(Ser) from the HTT sense transcript and poly(Leu) and poly(Cys) from the HTT antisense strand) accumulate in HD human autopsy brains. These proteins accumulate in affected brain regions, including the striatum and frontal cortex, and in regions with neuronal loss, microglial activation, and apoptosis. Some regions, including the caudate/putamen, showed both poly(Gln) and RAN protein staining, and other regions, including caudate and putamen white matter bundle regions, showed RAN but not poly(Gln) protein staining (25). These data suggest RAN proteins may play a role in previously described HD white matter abnormalities (59–62). Additionally, Bañez-Coronel et al. (25) found evidence for robust RAN protein but minimal poly(Gln) accumulation throughout the degenerating cerebellar layers of juvenile onset HD cases with severe cerebellar atrophy. The region-specific accumulation of HD–RAN proteins could indicate that the degradation pathways that handle RAN proteins and/or the process of RAN translation itself varies in efficiency between different cell types. Taken together, these data suggest RAN proteins play a role in the neurodegenerative changes and white matter abnormalities in HD (59–62).
Myotonic dystrophy type 2
In 2017, Zu et al. (63) showed that the myotonic dystrophy type 2 (DM2) intronic CCTG expansion mutation located in the cellular nucleic acid-binding protein (CNBP) gene can undergo both bidirectional transcription and RAN translation. DM2 (64) is a multisystemic disorder clinically similar to myotonic dystrophy type 1, which includes a late-onset CNS phenotype involving executive function deficits and white matter abnormalities (65, 66). Although the DM2 expansions produce the same tetrapeptide repeat motifs in all three reading frames: leucine–proline–alanine–cysteine (LPAC) in the sense direction and glutamine–alanine–glycine–arginine (QAGR) in the antisense direction (63), the C-terminal regions in each reading frame differ, resulting in the expression of six unique proteins. The LPAC and QAGR RAN proteins accumulate in DM2 autopsy brains in distinct patterns, with LPAC primarily found in gray matter and QAGR in white matter regions of the brain. Codon-replacement studies show LPAC and QAGR RAN proteins are toxic-independent of CCUG- or CAGG-induced RNA GOF effects. These authors also showed that the nuclear sequestration of CCUG expansion transcripts into RNA foci decreases the steady-state levels of the LPAC RAN proteins. These data support a nuclear sequestration failure model in which RNA GOF effects predominate until the capacity to sequester expansion RNAs in the nuclei is exceeded and expansion RNAs are exported to the cytoplasm (sequestration failure) where they undergo RAN translation. This model predicts a highly variable pattern of RNA foci and RAN protein accumulation depending on the capacity of individual cells to sequester expansion RNAs and/or undergo RAN translation.
Spinocerebellar ataxia type 31: A pentanucleotide repeat
Spinocerebellar ataxia type 31 (SCA31) is an autosomal-dominant disease caused by expansion of a complex pentanucleotide repeat. The repeat tract, which includes TGGAA, TAGAA, TAAAATAGAA, and TAAAA repeat motifs, is located within an intron shared by the NEDD4-1 (BEAN1) and thymidine kinase 2 (TK2) genes (67). The presence of UGGAA-containing RNA foci in Purkinje cell nuclei of SCA31 patients but not controls supports a toxic RNA GOF mechanism (68). This repeat expansion is also translated into pentapeptide repeat (PPR) proteins (poly(Tyr–Asp–Gly–Met–Glu)) that accumulate in both SCA31 patient brain autopsy tissue and Drosophila models (37). Because AUG-initiation codons are embedded within the repeat tract, it is unclear whether ATG-initiated or RAN translation is responsible for protein production. In either case, PPR protein production is repeat length-dependent as these proteins are only detected in SCA31 patients and fly models with repeat expansions (37). Several RBPs, including TDP-43, were shown to suppress RNA foci formation and PPR protein accumulation (37). For TDP-43, inhibition of RNA aggregates occurred in an ATP-independent manner in vitro (37), suggesting that RBPs, like TDP-43, may play a role in RNA quality control and/or regulation of translation. Another AT-rich repeat, an (ATTTC) insertion in the noncoding region of the DAB1 gene, has been associated with SCA37 (69) and shows RNA aggregates in human cells overexpressing (ATTTC)58 but not in (ATTTT)139 repeats. Although it remains to be determined whether bidirectional transcription and/or RAN translation occurs in SCA37, future experiments with these AT-rich repeats will provide insight into the mechanisms of neurodegenerative disorders.
Fuchs endothelial corneal dystrophy (FECD)
The most recent repeat expansion disorder shown to express RAN proteins is FECD (70). FECD involves the slowly-progressive degeneration of the corneal endothelium, which ultimately results in vision loss. An intronic CTG repeat located in the third intron of the transcription factor 4 (TCF4) gene was shown to be a common genetic cause of FECD (71). The CTG expansion mutation results in RNA foci and RNA splicing defects in endothelial cells from FECD patients, supporting an RNA GOF mechanism for this disease (72, 73). Soragni et al. (70) recently demonstrated that the intronic CTG·CAG expansion in TCF4 undergoes RAN translation in transfected cells and that the resulting antisense poly(Ser) and poly(Gln) RAN proteins are toxic to immortalized corneal endothelial cells. Additionally, the authors developed a C-terminal antibody against the sense poly(Cys) expansion protein expressed from the TCF4 CUG expansion RNAs and provided evidence for the accumulation of a poly(Cys) protein in patients' endothelial samples (70). These data provide another opportunity to understand the mechanisms of RAN translation and the tissue-specific pathogenic consequences of the protein products of a repeat expansion.
C9ORF72 ALS/FTD: Accelerating the pace of RAN translation discovery
The 2011 discovery that the most common known genetic forms of ALS and FTD are caused by a hexanucleotide (G4C2) expansion in the C9ORF72 (74, 75) gene has raised an enormous level of scientific interest because it linked the microsatellite expansion field to more common neurodegenerative diseases like ALS and dementia. C9-ALS/FTD mutation carriers may develop ALS, which causes upper and lower motor neuron loss and muscle atrophy, typically leading to death from respiratory failure within 3–5 years of onset (76). This mutation can also result in FTD, a disease characterized by behavioral and personality changes with language dysfunction followed by dementia later in the disease (77, 78). Disease mechanisms previously described in other repeat expansion diseases (4, 79) have been proposed for C9-ALS/FTD (80, 81), including the following: 1) protein LOF due to C9ORF72 protein haploinsufficiency (74, 75, 82); 2) RNA GOF and RNA processing abnormalities caused by sequestration of one or more RNA-binding proteins to C9-expansion RNAs (79, 84–89); and 3) RAN protein toxicity (33–35). Bidirectional transcription, another common feature of expansion mutations (2, 28, 90, 91), is also found in C9-ALS/FTD. Both sense and antisense transcripts accumulate as RNA foci and produce RAN proteins (sense: poly(Gly-Ala) (GA), poly(Gly-Arg) (GR), and poly(Gly-Pro) (GP); and antisense: poly(Gly-Pro) (GP), poly(Pro-Arg) (PR), and poly(Pro-Ala) (PA)) that accumulate in patient autopsy brains (33–35).
The mechanism of translational initiation from the C9ORF72 repeat expansion has been the subject of investigation. Using a cell-free in vitro translation system, Tabet et al. (92) showed that translation from the expanded G4C2 transcript operates via a 5′–3′ cap-dependent scanning mechanism that utilizes an upstream CUG codon, eIF4E, and is regulated by uORF. Similarly, Green et al. (93) also showed that translation across G4C2 repeat expansions is cap- and eIF4A-dependent and utilized the same near-cognate initiation codon. In contrast, data generated using cell-based studies by Cheng et al. (94) support cap-independent translation initiation for C9ORF72 expansion transcripts. Data from this cellular system support a model in which translation initiation occurs on uncapped spliced repeat-containing intronic RNA following export to the cytoplasm. Similar to IRES-driven translation, C9 cap-independent translation was shown to be up-regulated by ER stress pathways, through eIF2α phosphorylation. Cheng et al. (94) and Green et al. (93) speculate that disease is exacerbated by a feed-forward mechanism in which RAN proteins increase ER and oxidative stress, which leads to increased eIF2α phosphorylation and RAN protein expression. Additionally, increased R-loop formation and double-strand breaks as well as defective ataxia telangiectasia-mutated (ATM)-mediated repair associated with C9-expanded repeats (95) may also factor into this process by promoting intracellular stress.
In support of a possible role for protein loss-of-function mechanisms in C9-ALS/FTD, lower levels of C9ORF72 protein are observed in C9-ALS/FTD patients (75, 82, 87, 96, 97). Arguments against a role for this mechanism include that there are no known cases of ALS or FTD patients with null or missense C9ORF72 mutations. Furthermore, C9ORF72 knockout mice display an altered immune response but do not develop motor neuron degeneration or other features of ALS or FTD (98–103), suggesting loss-of-function of C9ORF72 is not a primary driver of disease. Data supporting an RNA gain-of-function mechanism includes the accumulation of both sense and antisense RNA foci in C9 patients and various model systems (35, 75, 104), splicing defects in C9 patient cells (105–107), and the in vitro identification of multiple potential C9ORF72 RNA-binding proteins (96, 104, 108–116). These findings, particularly the C9-associated RNA-binding proteins, have shown considerable variability leaving the contributions of specific RBPs in disease unclear. There has also been considerable research focused on understanding the role of RAN proteins in C9-ALS/FTD. Both the sense and antisense dipeptide repeat (DPR) or C9-RAN proteins accumulate as aggregates in the autopsy tissue of C9 patients. DPR proteins are a hallmark feature of C9-ALS/FTD and have been found in neurons and glia throughout the brain and spinal cord (34, 111, 117). Although there is general agreement that RAN proteins are toxic in model systems, the contribution of individual RAN proteins to disease is the subject of debate.
When overexpressed or delivered to cultured cells or animal models, C9-RAN proteins are generally toxic (33–35, 84, 87, 116, 118–124) with the arginine-containing proteins (poly(PR) and poly(GR)) showing the strongest toxicity in most studies (84, 119, 120, 125). PR and GR proteins interact with nuclear proteins causing splicing aberrations (118, 126), nucleolar stress (118, 127), abnormal stress granule formation (120), and translational dysregulation (120, 128). PR and GR proteins interact with other low complexity domain proteins, altering the physiology of phase separation and impairing the assembly and function of membrane-less organelles (125, 126). PR and GR RAN proteins have also been shown to associate with the U2 small nuclear ribonucleoprotein resulting in its cytoplasmic mislocalization and the blockage of spliceosome assembly and splicing (129), including genes related to mitochondrial, neuronal, and pre-mRNA splicing function. The GA RAN protein, which is moderately toxic in cell culture (116, 121, 122), was also shown to be toxic in zebrafish (130) and to induce neurodegeneration when overexpressed by AAV delivery in mice (116). GA proteins have been reported to interact with components of the ubiquitin proteosome system (UPS) and UPS-related proteins (116, 131) and to cause disruptions in the UPS system (116, 121, 122). Additionally, long poly(GA)80 proteins have been shown to recruit poly(GR)80 into cytoplasmic inclusions and thereby partially decrease GR-induced toxicity and Notch signaling defects (132). Recently, an elegant 3D cryo-electron tomography study by Guo et al. (133) showed that poly(GA) proteins form aggregates that selectively trap macromolecular complexes, including proteasomes, which may contribute to protein homeostasis problems. Interestingly, poly(Gln) aggregates have a fibril-like structure and cause vesicle and ER membrane deformation (134). It is important to note that the majority of these toxicity studies have utilized relatively short repeats (<90 repeats) compared with the hundreds or thousands of repeats found in C9-ALS/FTD patients. The length of the repeat tract can influence a number of events, including nuclear/cytoplasmic localization (118, 132, 135), inclusion formation (121), and toxicity (118, 128). Understanding the structure, behavior, and toxicity of additional types of RAN proteins found in patients, where repeats can be hundreds or thousands of units long, will be important for understanding the pathogenic mechanisms of C9-ALS/FTD and other RAN protein diseases.
Nucleocytoplasmic transport
The nuclear pore complex and nucleocytoplasmic transport deficits have been associated with several repeat expansion disorders. Chromosomal DNA with expanded CAG repeats associates with nuclear pores in yeast and NPC-associated factors (136). In DM1, the transcription factor SHARP (SMART/HDAC1-associated repressor protein) is mislocalized to the cytoplasm, due to increased CRM1-mediated export, although the nucleocytoplasmic shuttling of other CRM1-mediated targets is not affected (137). In HD, the HTT protein contains a highly conserved nuclear export signal (138), and NPC components have been detected in synthetic poly(Gln) aggregates (139). Huntingtin N-terminal poly(Gln) fragments reduce huntingtin interaction with the nuclear export protein, translocated nuclear pore (TPR) (140). Additionally, poly(Gln) expansion proteins from repeat expansion diseases have reduced rates of nuclear export (141, 142) or nuclear pore abnormalities (143, 144). These older studies were conducted prior to the discovery of RAN translation and warrant reinvestigation to examine the potential contribution of RAN proteins to alterations in the nuclear pore complex and nucleocytoplasmic transport. Grima et al. (145) recently demonstrated severe mislocalization and aggregation of nucleoporins (NUPs) and defective nucleocytoplasmic transport can be induced by a mixture of HD–RAN proteins and mediated by RanGAP. Another group, Gasset-Rosa et al. (146), found, using HD mouse models, multiple nuclear membrane or NPC defects, including aggregated nuclear pore factors that appeared to colocalize with mouse HTT aggregates. In another study, cytoplasmic, but not nuclear, aggregates of amyloid-like proteins, including mutant huntingtin, interfered with nucleocytoplasmic transport of both proteins and RNA (147). Expression of the FMR–poly(Gly) has also been linked to altered nuclear lamina architecture that may contribute to FXTAS (56).
In C9ORF72 ALS/FTD, both the direct interaction of C9ORF72–RNA fragments with nuclear pore complex proteins (86) and the disruption of NPC function by RAN proteins (85, 148–150) have been suggested as neurodegenerative pathways. Additional support for the role of nucleocytoplasmic transport deficits comes from the observation that misregulated nuclear transport factors are found in multiple forms of ALS and FTD patient autopsy material and patient-derived induced pluripotent stem cell neurons (86, 151–153). TDP-43 pathology also triggers structural defects in the NPC and nucleocytoplasmic transport across multiple types of ALS/FTD (154). Several studies have identified nucleocytoplasmic transport components, including RanGAP (86), as suppressors or enhancers of disease using Drosophila (84, 86), yeast (85), and siRNA-based human cell (155) screens. Additionally it has been shown that PR interacts directly with the nuclear pore by plugging it and thereby hindering nuclear export (156). RAN poly(PR) peptides hinder nuclear export, likely by binding directly to the central channel of the nuclear pore, through a direct interaction between poly(PR) and nuclear pore proteins enriched in phenylalanine/glycine repeats (156). Although nuclear pore pathology is a common in many neurodegenerative diseases, further studies are needed to understand whether these deficits are a cause or consequence of other cellular problems.
Mouse models
Although there has been considerable progress in understanding C9-ALS/FTD from the analysis of cell culture, simple animal models, and patient-derived tissues, the progress on the development of transgenic mouse models has been slow until recently (98–103, 157–162). Because the regulation of the sense and antisense genes is complex, several groups decided to generate BAC transgenic models of C9ORF72 ALS/FTD to allow expression of these overlapping genes to be driven by their endogenous human promoters. Whereas animals from all four different BAC transgenic models produce RAN proteins and RNA foci, disease presentation varies greatly. Two groups did not observe behavioral or pathological phenotypes (157, 158). Mice developed by a third group showed spatial learning and working memory deficits with mild hippocampal degeneration, but not the severe neurodegenerative phenotypes found in patients with C9ORF72 ALS/FTD (159). A more severe phenotype was observed in a BAC transgenic mouse model developed by a fourth group (160). These mice show classic features of both ALS and FTD, including decreased survival, paralysis, muscle denervation, motor neuron loss, anxiety-like behavior, and cortical and hippocampal neurodegeneration. Although both sense and antisense foci are found in these mice, antisense foci preferentially accumulate in ALS/FTD-vulnerable cell populations. As these animals age and the disease progresses, RAN protein accumulation increases, with end-stage animals displaying the typical TDP-43 inclusions in degenerating regions of the brain. A recent transgenic mouse model, expressing poly(GA) protein independent of RNA GOF effects, demonstrated accumulation of the poly(GA) protein results in mild motor phenotypes, including gait and balance abnormalities, but not the overt phenotypes typical of ALS/FTD, including paralysis and death (163). Similar to other models, RAN protein inclusions develop before symptoms appear in the poly(GA) mice (163), supporting the idea that the molecular effects of these mutations precede overt disease phenotypes. Mouse models with ALS/FTD phenotypes are critical for the development and testing of therapeutic strategies. Additionally, understanding and comparing the molecular differences between phenotypic and nonphenotypic models should provide insight into disease modifiers.
Therapeutic approaches
The plethora of potential pathogenic elements associated with expanded repeats (Fig. 2) makes it complicated to develop and assess therapeutic interventions. Antisense oligonucleotides (ASOs), which mediate cleavage of target RNAs via nuclear RNase H, have been or are currently in clinical trials for targeting the sense transcripts for both HD (164, 165) and DM1 (166, 167). It is important to note that in these trials, the antisense transcripts and/or RAN proteins would not be down-regulated. ASO strategies are also being developed for other expansion disorders, including SCA2, SCA3, and C9ORF72 (168, 169). A single dose of sense-transcript targeting ASOs administered in a C9-BAC mouse model decreased expanded C9ORF72 transcript levels, sense foci, and both poly(GA) and poly(GP) protein levels (159). Although ASO injections in older mice improved cognitive test performance, it is important to note that these mice do not develop TDP-43 inclusions or the motor neuron loss characteristic of C9ORF72 ALS/FTD (159). Interestingly, beneficial effects in treated mice were observed 6 months following injection, at a time when the expanded RNA transcript levels were no longer reduced and poly(GP) and poly(GA) levels remained lower (159). An alternative ASO approach has utilized the knockdown of the SUPT4H1–SUPT5H transcriptional elongation factor complex, which reduces transcription of genes with long stretches of expanded repeats without genome-wide changes in the expression of other RNAs (170, 171). Reducing SUPT4H1 levels by either ASO knockdown or genetic deletion results in reductions in mutant-expanded HTT mRNA, Htt aggregates, and phenotypic recovery (172). A similar knockdown of SUPT5H in patient-derived C9ORF72 cells reduced both sense and antisense RNA foci as well as poly(GP) protein without large-scale changes in other transcripts (170). While promising, this treatment strategy does not reduce expanded repeat transcript levels as much as direct targeting with ASOs (100, 159, 170), and SUPT4H1/SUPT5H alters the expression of a number of other genes (170) that may have deleterious consequences.
Figure 2.
Pathogenesis of a microsatellite repeat expansion disorder. An illustration of the three nonexclusive disease mechanisms proposed for most microsatellite expansion disorders, using C9ORF72 ALS/FTD as an example, is shown. A, microsatellite repeat expansion mutation (G4C2·G2C4 for C9-ALS/FTD) results in transcriptional inhibition and/or epigenetic silencing that reduces the levels of the resulting protein product (75, 82, 83). B, expansion mutations produce up to six toxic RAN proteins from both sense and antisense mutant transcripts. These proteins disrupt normal cellular functions (e.g. nucleocytoplasmic transport) and/or overwhelm cellular coping mechanisms (e.g. protein homeostasis). In C9-ALS/FTD protein GOF effects lead to nucleolar dysfunction, ER stress, altered autophagy, cell to cell transmission of RAN proteins, nucleocytoplasmic transport, and nuclear envelope deficits (33–36). C, expansion RNAs sequester RBPs into nuclear foci reducing RBPs' availability and decreasing its normal function. Expansion transcripts may also interact with and disrupt the function of other cellular components, such as proteins of nuclear pore complex (86). Although the identity and altered function of the RBP protein for C9ORF72 hexanucleotide repeats are the subject of much debate (83, 96, 108, 110, 113–115), RNA GOF effects are well established for DM1 CUG repeats that sequester MBNL proteins (79, 84–88). Different therapeutic approaches (purple boxes) target the various expansion RNA and protein products. ASOs and small molecules (SM) have been used to target either the sense or antisense expansion RNAs, although the effect on the opposite strand is unclear. Alternatively, ASOs can target transcription of the expanded repeat, e.g. SUPT5H (170). Additionally, therapeutic approaches, including small molecules, have been aimed at the downstream consequences of the expansion mutations, such as increasing or improving protein clearance mechanisms. Antibodies against RAN proteins (188) (Ab) or overexpressing proteins involved in autophagy (190) are also therapeutic approaches.
Additional approaches, including the use of small molecules, are being applied to repeat expansion disorders. These strategies include blocking transcription (173, 174) and targeting the expansion RNAs (175–181) or downstream cellular processes (122, 182–186). Targeting nucleocytoplasmic transport defects associated with repeat diseases has also been shown to be neuroprotective in both C9ORF72-ALS models (86, 187) and HD mouse models (145). Depletion of SRSF1, which inhibits nuclear export of C9ORF72 expansion transcripts by preventing its interaction with its nuclear export receptor, has been shown to prevent neurodegeneration and locomotor deficits in flies (187). In contrast, approaches that focus on inhibiting sequestration of expansion transcripts by RBPs may allow expansion transcripts to be exported to the cytoplasm, undergo RAN translation, and further exacerbate the disease (63). Additional efforts have focused on targeting RAN proteins, and anti-GA antibodies have been shown to inhibit intracellular poly(GA) aggregation in cell culture and to block seeding activity in brain extracts (188). Overexpression of the small heat-shock protein B8 (HSPB8), which modulates autophagy-mediated disposal of misfolded aggregation-prone proteins (189), was recently shown to decrease the accumulation of most C9-RAN proteins (190). Targeting RAN proteins directly has yet to be tested for therapeutic potential in more complex disease models but doing so may also provide clues to the pathogenic role of RNA GOF versus RAN translation. A better understanding of the mechanisms of RAN translation and the role of individual RAN proteins in disease is likely to provide novel therapeutic opportunities.
Summary and future directions
RAN proteins have now been reported in eight repeat expansion disorders with different repeat motifs, pathogenic thresholds, and disease presentations. Although significant progress has been made in understanding the role of RAN proteins in disease, additional insights into the mechanisms of RAN translation will facilitate the identification of new therapeutic targets and advance our understanding of cell biology and protein translation. Although the structure, function, and C-terminal regions of individual RAN proteins differ and warrant independent consideration, targeting RAN translation could provide a single therapeutic strategy likely to impact an entire category of repeat expansion diseases. Additionally, the development of new tools and strategies to study RAN translation and RAN proteins are needed, along with models that mimic the full spectrum of molecular, pathological, and behavioral features of disease seen in patients. In summary, RAN translation is a complex biological process that we are only beginning to understand. Given the prevalence of repetitive elements in the human genome, RAN translation is likely to be found in additional diseases and possibly also contributes to normal cellular biology.
Acknowledgments
We thank our colleagues at the University of Florida and the Center for NeuroGenetics for their helpful discussions and input on this work.
This work was supported by National Institutes of Health Grants P01NS058901, R37NS040389, and R01NS098819, the ALS Association, Target ALS, Muscular Dystrophy Association, the Robert Packard Center for ALS Research, and the Myotonic Dystrophy Foundation. The authors declare that they have no conflicts of interest with the contents of this article. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
- RAN
- repeat-associated non-AUG
- RBP
- RNA-binding protein
- ALS
- amyotrophic lateral sclerosis
- ALS/FTD
- ALS/frontotemporal dementia
- GOF
- gain-of-function
- LOF
- loss-of-function
- HD
- Huntington's disease
- IRES
- internal-ribosome entry site
- ER
- endoplasmic reticulum
- CNS
- central nervous system
- FXTAS
- fragile X tremor ataxia syndrome
- eIF
- eukaryotic initiation factor
- uORF
- upstream open reading frame
- FXPOI
- fragile X premutation ovarian insufficiency
- FECD
- Fuchs endothelial corneal dystrophy
- PRR
- pentapeptide repeat
- UPS
- ubiquitin proteosome system
- DPR
- dipeptide repeat
- NPC
- nuclear pore complex
- ASO
- antisense oligonucleotide
- FMR
- fragile X mental retardation.
References
- 1. Zu T., Gibbens B., Doty N. S., Gomes-Pereira M., Huguet A., Stone M. D., Margolis J., Peterson M., Markowski T. W., Ingram M. A., Nan Z., Forster C., Low W. C., Schoser B., Somia N. V., et al. (2011) Non-ATG-initiated translation directed by microsatellite expansions. Proc. Natl. Acad. Sci. U.S.A. 108, 260–265 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Batra R., Charizanis K., and Swanson M. S. (2010) Partners in crime: bidirectional transcription in unstable microsatellite disease. Hum. Mol. Genet. 19, R77–82 10.1093/hmg/ddq132 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Groh M., Silva L. M., and Gromak N. (2014) Mechanisms of transcriptional dysregulation in repeat expansion disorders. Biochem. Soc. Trans. 42, 1123–1128 10.1042/BST20140049 [DOI] [PubMed] [Google Scholar]
- 4. Cleary J. D., and Ranum L. P. (2014) Repeat associated non-ATG (RAN) translation: new starts in microsatellite expansion disorders. Curr. Opin. Genet. Dev. 26, 6–15 10.1016/j.gde.2014.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Cleary J. D., and Ranum L. P. (2013) Repeat-associated non-ATG (RAN) translation in neurological disease. Hum. Mol. Genet. 22, R45–R51 10.1093/hmg/ddt371 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Pearson C. E. (2011) Repeat associated non-ATG translation initiation: one DNA, two transcripts, seven reading frames, potentially nine toxic entities! PLoS Genet. 7, e1002018 10.1371/journal.pgen.1002018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Lander E. S., Linton L. M., Birren B., Nusbaum C., Zody M. C., Baldwin J., Devon K., Dewar K., Doyle M., FitzHugh W., Funke R., Gage D., Harris K., Heaford A., Howland J., et al. (2001) Initial sequencing and analysis of the human genome. Nature 409, 860–921 10.1038/35057062 [DOI] [PubMed] [Google Scholar]
- 8. Tóth G., Gáspári Z., and Jurka J. (2000) Microsatellites in different eukaryotic genomes: survey and analysis. Genome Res. 10, 967–981 10.1101/gr.10.7.967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Nelson D. L., Orr H. T., and Warren S. T. (2013) The unstable repeats–three evolving faces of neurological disease. Neuron 77, 825–843 10.1016/j.neuron.2013.02.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Orr H. T. (2009) Unstable nucleotide repeat minireview series: a molecular biography of unstable repeat disorders. J. Biol. Chem. 284, 7405 10.1074/jbc.R800067200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Orr H. T., and Zoghbi H. Y. (2007) Trinucleotide repeat disorders. Annu. Rev. Neurosci. 30, 575–621 10.1146/annurev.neuro.29.051605.113042 [DOI] [PubMed] [Google Scholar]
- 12. López Castel A., Cleary J. D., and Pearson C. E. (2010) Repeat instability as the basis for human diseases and as a potential target for therapy. Nat. Rev. Mol. Cell Biol. 11, 165–170 10.1038/nrm2854 [DOI] [PubMed] [Google Scholar]
- 13. Cleary J. D., La Spada A. R., and Pearson C. E. (2006) in DNA Replication and Human Disease (DePamphilis M. L., ed) pp. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York [Google Scholar]
- 14. Harley H. G., Brook J. D., Rundle S. A., Crow S., Reardon W., Buckler A. J., Harper P. S., Housman D. E., and Shaw D. J. (1992) Expansion of an unstable DNA region and phenotypic variation in myotonic dystrophy. Nature 355, 545–546 10.1038/355545a0 [DOI] [PubMed] [Google Scholar]
- 15. Trottier Y., Biancalana V., and Mandel J. L. (1994) Instability of CAG repeats in Huntington's disease: relation to parental transmission and age of onset. J. Med. Genet. 31, 377–382 10.1136/jmg.31.5.377 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Labbadia J., and Morimoto R. I. (2013) Huntington's disease: underlying molecular mechanisms and emerging concepts. Trends Biochem. Sci. 38, 378–385 10.1016/j.tibs.2013.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Sonenberg N., and Hinnebusch A. G. (2009) Regulation of translation initiation in eukaryotes: mechanisms and biological targets. Cell 136, 731–745 10.1016/j.cell.2009.01.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Hershey J. W., Sonenberg N., and Mathews M. B. (2012) Principles of translational control: an overview. Cold Spring Harb. Perspect. Biol. 4, a011528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Hinnebusch A. G. (2014) The scanning mechanism of eukaryotic translation initiation. Annu. Rev. Biochem. 83, 779–812 10.1146/annurev-biochem-060713-035802 [DOI] [PubMed] [Google Scholar]
- 20. Jackson R. J., Hellen C. U., and Pestova T. V. (2010) The mechanism of eukaryotic translation initiation and principles of its regulation. Nat. Rev. Mol. Cell Biol. 11, 113–127 10.1038/nrm2838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Komar A. A., and Hatzoglou M. (2011) Cellular IRES-mediated translation: the war of ITAFs in pathophysiological states. Cell Cycle 10, 229–240 10.4161/cc.10.2.14472 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Peabody D. S. (1989) Translation initiation at non-AUG triplets in mammalian cells. J. Biol. Chem. 264, 5031–5035 [PubMed] [Google Scholar]
- 23. Starck S. R., Jiang V., Pavon-Eternod M., Prasad S., McCarthy B., Pan T., and Shastri N. (2012) Leucine-tRNA initiates at CUG start codons for protein synthesis and presentation by MHC class I. Science 336, 1719–1723 10.1126/science.1220270 [DOI] [PubMed] [Google Scholar]
- 24. Schwab S. R., Shugart J. A., Horng T., Malarkannan S., and Shastri N. (2004) Unanticipated antigens: translation initiation at CUG with leucine. PLoS Biol. 2, e366 10.1371/journal.pbio.0020366 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Bañez-Coronel M., Ayhan F., Tarabochia A. D., Zu T., Perez B. A., Tusi S. K., Pletnikova O., Borchelt D. R., Ross C. A., Margolis R. L., Yachnis A. T., Troncoso J. C., and Ranum L. P. (2015) RAN translation in Huntington's disease. Neuron 88, 667–677 10.1016/j.neuron.2015.10.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Koob M. D., Moseley M. L., Schut L. J., Benzow K. A., Bird T. D., Day J. W., and Ranum L. P. (1999) An untranslated CTG expansion causes a novel form of spinocerebellar ataxia (SCA8). Nat. Genet. 21, 379–384 10.1038/7710 [DOI] [PubMed] [Google Scholar]
- 27. Day J. W., Schut L. J., Moseley M. L., Durand A. C., and Ranum L. P. (2000) Spinocerebellar ataxia type 8: clinical features in a large family. Neurology 55, 649–657 10.1212/WNL.55.5.649 [DOI] [PubMed] [Google Scholar]
- 28. Moseley M. L., Zu T., Ikeda Y., Gao W., Mosemiller A. K., Daughters R. S., Chen G., Weatherspoon M. R., Clark H. B., Ebner T. J., Day J. W., and Ranum L. P. (2006) Bidirectional expression of CUG and CAG expansion transcripts and intranuclear polyglutamine inclusions in spinocerebellar ataxia type 8. Nat. Genet. 38, 758–769 10.1038/ng1827 [DOI] [PubMed] [Google Scholar]
- 29. Daughters R. S., Tuttle D. L., Gao W., Ikeda Y., Moseley M. L., Ebner T. J., Swanson M. S., and Ranum L. P. (2009) RNA gain-of-function in spinocerebellar ataxia type 8. PLoS Genet. 5, e1000600 10.1371/journal.pgen.1000600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Ayhan F., Perez B. A., Shorrock H. K., Zu T., Banez-Coronel M., Reid T. S., Furuya H., Clark H. B., Troncoso J. C., Ross C., Subramony S. H., Ashizawa T., Wang E. T., Yachnis A. T., and Ranum L. P. (2018) SCA8 RAN poly(Ser) protein preferentially accumulates in white matter regions and is regulated by eIF3F. EMBO J. 2018, e99023 10.15252/embj.201899023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. U M., Miyashita T., Ohtsuka Y., Okamura-Oho Y., Shikama Y., and Yamada M. (2001) Extended polyglutamine selectively interacts with caspase-8 and -10 in nuclear aggregates. Cell Death Differ. 8, 377–386 10.1038/sj.cdd.4400819 [DOI] [PubMed] [Google Scholar]
- 32. Todd P. K., Oh S. Y., Krans A., He F., Sellier C., Frazer M., Renoux A. J., Chen K. C., Scaglione K. M., Basrur V., Elenitoba-Johnson K., Vonsattel J. P., Louis E. D., Sutton M. A., Taylor J. P., et al. (2013) CGG repeat-associated translation mediates neurodegeneration in fragile X tremor ataxia syndrome. Neuron 78, 440–455 10.1016/j.neuron.2013.03.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Mori K., Weng S. M., Arzberger T., May S., Rentzsch K., Kremmer E., Schmid B., Kretzschmar H. A., Cruts M., Van Broeckhoven C., Haass C., and Edbauer D. (2013) The C9orf72 GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in FTLD/ALS. Science 339, 1335–1338 10.1126/science.1232927 [DOI] [PubMed] [Google Scholar]
- 34. Ash P. E., Bieniek K. F., Gendron T. F., Caulfield T., Lin W. L., Dejesus-Hernandez M., van Blitterswijk M. M., Jansen-West K., Paul J. W. 3rd., Rademakers R., Boylan K. B., Dickson D. W., and Petrucelli L. (2013) Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron 77, 639–646 10.1016/j.neuron.2013.02.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Zu T., Liu Y., Bañez-Coronel M., Reid T., Pletnikova O., Lewis J., Miller T. M., Harms M. B., Falchook A. E., Subramony S. H., Ostrow L. W., Rothstein J. D., Troncoso J. C., and Ranum L. P. (2013) RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia. Proc. Natl. Acad. Sci. U.S.A. 110, E4968–E4977 10.1073/pnas.1315438110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Westergard T., Jensen B. K., Wen X., Cai J., Kropf E., Iacovitti L., Pasinelli P., and Trotti D. (2016) Cell-to-cell transmission of dipeptide repeat proteins linked to C9orf72-ALS/FTD. Cell Rep. 17, 645–652 10.1016/j.celrep.2016.09.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Ishiguro T., Sato N., Ueyama M., Fujikake N., Sellier C., Kanegami A., Tokuda E., Zamiri B., Gall-Duncan T., Mirceta M., Furukawa Y., Yokota T., Wada K., Taylor J. P., Pearson C. E., et al. (2017) Regulatory role of RNA chaperone TDP-43 for RNA misfolding and repeat-associated translation in SCA31. Neuron 94, 108–124.e7 10.1016/j.neuron.2017.02.046 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Broda M., Kierzek E., Gdaniec Z., Kulinski T., and Kierzek R. (2005) Thermodynamic stability of RNA structures formed by CNG trinucleotide repeats. Implication for prediction of RNA structure. Biochemistry 44, 10873–10882 10.1021/bi0502339 [DOI] [PubMed] [Google Scholar]
- 39. Reddy K., Zamiri B., Stanley S. Y., Macgregor R. B. Jr., and Pearson C. E. (2013) The disease-associated r(GGGGCC)n repeat from the C9orf72 gene forms tract length-dependent uni- and multimolecular RNA G-quadruplex structures. J. Biol. Chem. 288, 9860–9866 10.1074/jbc.C113.452532 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Fratta P., Mizielinska S., Nicoll A. J., Zloh M., Fisher E. M., Parkinson G., and Isaacs A. M. (2012) C9orf72 hexanucleotide repeat associated with amyotrophic lateral sclerosis and frontotemporal dementia forms RNA G-quadruplexes. Sci. Rep. 2, 1016 10.1038/srep01016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Hagerman P. J., and Hagerman R. J. (2015) Fragile X-associated tremor/ataxia syndrome. Ann. N.Y. Acad. Sci. 1338, 58–70 10.1111/nyas.12693 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Hagerman P. (2013) Fragile X-associated tremor/ataxia syndrome (FXTAS): pathology and mechanisms. Acta Neuropathol. 126, 1–19 10.1007/s00401-013-1138-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Hagerman R., and Hagerman P. (2013) Advances in clinical and molecular understanding of the FMR1 premutation and fragile X-associated tremor/ataxia syndrome. Lancet Neurol. 12, 786–798 10.1016/S1474-4422(13)70125-X [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Leehey M. A., and Hagerman P. J. (2012) Fragile X-associated tremor/ataxia syndrome. Handb. Clin. Neurol. 103, 373–386 10.1016/B978-0-444-51892-7.00023-1 [DOI] [PubMed] [Google Scholar]
- 45. Buijsen R. A., Visser J. A., Kramer P., Severijnen E. A., Gearing M., Charlet-Berguerand N., Sherman S. L., Berman R. F., Willemsen R., and Hukema R. K. (2016) Presence of inclusions positive for polyglycine containing protein, FMRpolyG, indicates that repeat-associated non-AUG translation plays a role in fragile X-associated primary ovarian insufficiency. Hum. Reprod. 31, 158–168 10.1093/humrep/dev280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Krans A., Kearse M. G., and Todd P. K. (2016) RAN translation from antisense CCG repeats in fragile X tremor/ataxia syndrome. Ann. Neurol. 80, 871–881 10.1002/ana.24800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Kearse M. G., Green K. M., Krans A., Rodriguez C. M., Linsalata A. E., Goldstrohm A. C., and Todd P. K. (2016) CGG Repeat-associated non-AUG translation utilizes a Cap-dependent scanning mechanism of initiation to produce toxic proteins. Mol. Cell 62, 314–322 10.1016/j.molcel.2016.02.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Calvo S. E., Pagliarini D. J., and Mootha V. K. (2009) Upstream open reading frames cause widespread reduction of protein expression and are polymorphic among humans. Proc. Natl. Acad. Sci. U.S.A. 106, 7507–7512 10.1073/pnas.0810916106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Ingolia N. T., Brar G. A., Stern-Ginossar N., Harris M. S., Talhouarne G. J., Jackson S. E., Wills M. R., and Weissman J. S. (2014) Ribosome profiling reveals pervasive translation outside of annotated protein-coding genes. Cell Rep. 8, 1365–1379 10.1016/j.celrep.2014.07.045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Ingolia N. T., Lareau L. F., and Weissman J. S. (2011) Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of mammalian proteomes. Cell 147, 789–802 10.1016/j.cell.2011.10.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Ingolia N. T. (2010) Genome-wide translational profiling by ribosome footprinting. Methods Enzymol. 470, 119–142 10.1016/S0076-6879(10)70006-9 [DOI] [PubMed] [Google Scholar]
- 52. Lee S., Liu B., Lee S., Huang S. X., Shen B., and Qian S. B. (2012) Global mapping of translation initiation sites in mammalian cells at single-nucleotide resolution. Proc. Natl. Acad. Sci. U.S.A. 109, E2424–E2432 10.1073/pnas.1207846109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Green K. M., Linsalata A. E., and Todd P. K. (2016) RAN translation–What makes it run? Brain Res. 1647, 30–42 10.1016/j.brainres.2016.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Hukema R. K., Buijsen R. A., Schonewille M., Raske C., Severijnen L. A., Nieuwenhuizen-Bakker I., Verhagen R. F., van Dessel L., Maas A., Charlet-Berguerand N., De Zeeuw C. I., Hagerman P. J., Berman R. F., and Willemsen R. (2015) Reversibility of neuropathology and motor deficits in an inducible mouse model for FXTAS. Hum. Mol. Genet. 24, 4948–4957 10.1093/hmg/ddv216 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Castro H., Kul E., Buijsen R. A. M., Severijnen L. W. F. M., Willemsen R., Hukema R. K., Stork O., and Santos M. (2017) Selective rescue of heightened anxiety but not gait ataxia in a premutation 90CGG mouse model of fragile X-associated tremor/ataxia syndrome. Hum. Mol. Genet. 26, 2133–2145 10.1093/hmg/ddx108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Sellier C., Buijsen R. A. M., He F., Natla S., Jung L., Tropel P., Gaucherot A., Jacobs H., Meziane H., Vincent A., Champy M. F., Sorg T., Pavlovic G., Wattenhofer-Donze M., Birling M. C., et al. (2017) Translation of expanded CGG repeats into FMRpolyG is pathogenic and may contribute to Fragile X tremor ataxia syndrome. Neuron 93, 331–347 10.1016/j.neuron.2016.12.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Ross C. A., Aylward E. H., Wild E. J., Langbehn D. R., Long J. D., Warner J. H., Scahill R. I., Leavitt B. R., Stout J. C., Paulsen J. S., Reilmann R., Unschuld P. G., Wexler A., Margolis R. L., and Tabrizi S. J. (2014) Huntington's disease: natural history, biomarkers and prospects for therapeutics. Nat. Rev. Neurol. 10, 204–216 10.1038/nrneurol.2014.24 [DOI] [PubMed] [Google Scholar]
- 58. Bates G. P., Dorsey R., Gusella J. F., Hayden M. R., Kay C., Leavitt B. R., Nance M., Ross C. A., Scahill R. I., Wetzel R., Wild E. J., and Tabrizi S. J. (2015) Huntington's disease. Nat. Rev. Dis. Primers 1, 15005 [DOI] [PubMed] [Google Scholar]
- 59. Bohanna I., Georgiou-Karistianis N., Sritharan A., Asadi H., Johnston L., Churchyard A., and Egan G. (2011) Diffusion tensor imaging in Huntington's disease reveals distinct patterns of white matter degeneration associated with motor and cognitive deficits. Brain Imaging Behav. 5, 171–180 10.1007/s11682-011-9121-8 [DOI] [PubMed] [Google Scholar]
- 60. Fennema-Notestine C., Archibald S. L., Jacobson M. W., Corey-Bloom J., Paulsen J. S., Peavy G. M., Gamst A. C., Hamilton J. M., Salmon D. P., and Jernigan T. L. (2004) In vivo evidence of cerebellar atrophy and cerebral white matter loss in Huntington's disease. Neurology 63, 989–995 10.1212/01.WNL.0000138434.68093.67 [DOI] [PubMed] [Google Scholar]
- 61. Paulsen J. S., Nopoulos P. C., Aylward E., Ross C. A., Johnson H., Magnotta V. A., Juhl A., Pierson R. K., Mills J., Langbehn D., Nance M., and PREDICT-HD Investigators and Coordinators of the Huntington's Study (HSG). (2010) Striatal and white matter predictors of estimated diagnosis for Huntington's disease. Brain Res. Bull. 82, 201–207 10.1016/j.brainresbull.2010.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Reading S. A., Yassa M. A., Bakker A., Dziorny A. C., Gourley L. M., Yallapragada V., Rosenblatt A., Margolis R. L., Aylward E. H., Brandt J., Mori S., van Zijl P., Bassett S. S., and Ross C. A. (2005) Regional white matter change in pre-symptomatic Huntington's disease: a diffusion tensor imaging study. Psychiatry Res. 140, 55–62 10.1016/j.pscychresns.2005.05.011 [DOI] [PubMed] [Google Scholar]
- 63. Zu T., Cleary J. D., Liu Y., Bañez-Coronel M., Bubenik J. L., Ayhan F., Ashizawa T., Xia G., Clark H. B., Yachnis A. T., Swanson M. S., and Ranum L. P. (2017) RAN translation regulated by muscleblind proteins in myotonic dystrophy type 2. Neuron 95, 1292–1305.e5 10.1016/j.neuron.2017.08.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Liquori C. L., Ricker K., Moseley M. L., Jacobsen J. F., Kress W., Naylor S. L., Day J. W., and Ranum L. P. (2001) Myotonic dystrophy type 2 caused by a CCTG expansion in intron 1 of ZNF9. Science 293, 864–867 10.1126/science.1062125 [DOI] [PubMed] [Google Scholar]
- 65. Franc D. T., Muetzel R. L., Robinson P. R., Rodriguez C. P., Dalton J. C., Naughton C. E., Mueller B. A., Wozniak J. R., Lim K. O., and Day J. W. (2012) Cerebral and muscle MRI abnormalities in myotonic dystrophy. Neuromuscul. Disord. 22, 483–491 10.1016/j.nmd.2012.01.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Cho D. H., and Tapscott S. J. (2007) Myotonic dystrophy: emerging mechanisms for DM1 and DM2. Biochim. Biophys. Acta 1772, 195–204 10.1016/j.bbadis.2006.05.013 [DOI] [PubMed] [Google Scholar]
- 67. Sato N., Amino T., Kobayashi K., Asakawa S., Ishiguro T., Tsunemi T., Takahashi M., Matsuura T., Flanigan K. M., Iwasaki S., Ishino F., Saito Y., Murayama S., Yoshida M., Hashizume Y., et al. (2009) Spinocerebellar ataxia type 31 is associated with “inserted” penta-nucleotide repeats containing (TGGAA)(n). Am. J. Hum. Genet. 85, 544–557 10.1016/j.ajhg.2009.09.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Niimi Y., Takahashi M., Sugawara E., Umeda S., Obayashi M., Sato N., Ishiguro T., Higashi M., Eishi Y., Mizusawa H., and Ishikawa K. (2013) Abnormal RNA structures (RNA foci) containing a penta-nucleotide repeat (UGGAA)n in the Purkinje cell nucleus is associated with spinocerebellar ataxia type 31 pathogenesis. Neuropathology 33, 600–611 10.1111/neup.12032 [DOI] [PubMed] [Google Scholar]
- 69. Seixas A. I., Loureiro J. R., Costa C., Ordóñez-Ugalde A., Marcelino H., Oliveira C. L., Loureiro J. L., Dhingra A., Brandão E., Cruz V. T., Timóteo A., Quintáns B., Rouleau G. A., Rizzu P., Carracedo Á., et al. (2017) A pentanucleotide ATTTC repeat insertion in the non-coding region of DAB1, mapping to SCA37, causes spinocerebellar ataxia. Am. J. Hum. Genet. 101, 87–103 10.1016/j.ajhg.2017.06.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Soragni E., Petrosyan L., Rinkoski T. A., Wieben E. D., Baratz K. H., Fautsch M. P., and Gottesfeld J. M. (2018) Repeat-associated non-ATG (RAN) translation in Fuchs' endothelial corneal dystrophy. Invest. Ophthalmol. Vis. Sci. 59, 1888–1896 10.1167/iovs.17-23265 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Wieben E. D., Aleff R. A., Tosakulwong N., Butz M. L., Highsmith W. E., Edwards A. O., and Baratz K. H. (2012) A common trinucleotide repeat expansion within the transcription factor 4 (TCF4, E2-2) gene predicts Fuchs corneal dystrophy. PLoS ONE 7, e49083 10.1371/journal.pone.0049083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Du J., Aleff R. A., Soragni E., Kalari K., Nie J., Tang X., Davila J., Kocher J. P., Patel S. V., Gottesfeld J. M., Baratz K. H., and Wieben E. D. (2015) RNA toxicity and missplicing in the common eye disease Fuchs endothelial corneal dystrophy. J. Biol. Chem. 290, 5979–5990 10.1074/jbc.M114.621607 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Wieben E. D., Aleff R. A., Tang X., Butz M. L., Kalari K. R., Highsmith E. W., Jen J., Vasmatzis G., Patel S. V., Maguire L. J., Baratz K. H., and Fautsch M. P. (2017) Trinucleotide repeat expansion in the transcription factor 4 (TCF4) gene leads to widespread mRNA splicing changes in Fuchs' endothelial corneal dystrophy. Invest. Ophthalmol. Vis. Sci. 58, 343–352 10.1167/iovs.16-20900 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Renton A. E., Majounie E., Waite A., Simón-Sánchez J., Rollinson S., Gibbs J. R., Schymick J. C., Laaksovirta H., van Swieten J. C., Myllykangas L., Kalimo H., Paetau A., Abramzon Y., Remes A. M., Kaganovich A., et al. (2011) A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72, 257–268 10.1016/j.neuron.2011.09.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. DeJesus-Hernandez M., Mackenzie I. R., Boeve B. F., Boxer A. L., Baker M., Rutherford N. J., Nicholson A. M., Finch N. A., Flynn H., Adamson J., Kouri N., Wojtas A., Sengdy P., Hsiung G. Y., Karydas A., et al. (2011) Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72, 245–256 10.1016/j.neuron.2011.09.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Rowland L. P., and Shneider N. A. (2001) Amyotrophic lateral sclerosis. N. Engl. J. Med. 344, 1688–1700 10.1056/NEJM200105313442207 [DOI] [PubMed] [Google Scholar]
- 77. van Blitterswijk M., DeJesus-Hernandez M., and Rademakers R. (2012) How do C9ORF72 repeat expansions cause amyotrophic lateral sclerosis and frontotemporal dementia: can we learn from other noncoding repeat expansion disorders? Curr. Opin. Neurol. 25, 689–700 10.1097/WCO.0b013e32835a3efb [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Rohrer J. D., Isaacs A. M., Mizielinska S., Mead S., Lashley T., Wray S., Sidle K., Fratta P., Orrell R. W., Hardy J., Holton J., Revesz T., Rossor M. N., and Warren J. D. (2015) C9orf72 expansions in frontotemporal dementia and amyotrophic lateral sclerosis. Lancet Neurol. 14, 291–301 10.1016/S1474-4422(14)70233-9 [DOI] [PubMed] [Google Scholar]
- 79. Goodwin M., and Swanson M. S. (2014) RNA-binding protein misregulation in microsatellite expansion disorders. Adv. Exp. Med. Biol. 825, 353–388 10.1007/978-1-4939-1221-6_10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Taylor J. P., Brown R. H. Jr., and Cleveland D. W. (2016) Decoding ALS: from genes to mechanism. Nature 539, 197–206 10.1038/nature20413 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Todd T. W., and Petrucelli L. (2016) Insights into the pathogenic mechanisms of chromosome 9 open reading frame 72 (C9orf72) repeat expansions. J. Neurochem. 138, Suppl. 1, 145–162 10.1111/jnc.13623 [DOI] [PubMed] [Google Scholar]
- 82. Waite A. J., Bäumer D., East S., Neal J., Morris H. R., Ansorge O., and Blake D. J. (2014) Reduced C9orf72 protein levels in frontal cortex of amyotrophic lateral sclerosis and frontotemporal degeneration brain with the C9ORF72 hexanucleotide repeat expansion. Neurobiol. Aging 35, 1779.e5–1779.e13 10.1016/j.neurobiolaging.2014.01.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Haeusler A. R., Donnelly C. J., and Rothstein J. D. (2016) The expanding biology of the C9orf72 nucleotide repeat expansion in neurodegenerative disease. Nat. Rev. Neurosci. 17, 383–395 10.1038/nrn.2016.38 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Freibaum B. D., Lu Y., Lopez-Gonzalez R., Kim N. C., Almeida S., Lee K. H., Badders N., Valentine M., Miller B. L., Wong P. C., Petrucelli L., Kim H. J., Gao F. B., and Taylor J. P. (2015) GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature 525, 129–133 10.1038/nature14974 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Jovičič A., Mertens J., Boeynaems S., Bogaert E., Chai N., Yamada S. B., Paul J. W. 3rd, Sun S., Herdy J. R., Bieri G., Kramer N. J., Gage F. H., Van Den Bosch L., Robberecht W., and Gitler A. D. (2015) Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. Nat. Neurosci. 18, 1226–1229 10.1038/nn.4085 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Zhang K., Donnelly C. J., Haeusler A. R., Grima J. C., Machamer J. B., Steinwald P., Daley E. L., Miller S. J., Cunningham K. M., Vidensky S., Gupta S., Thomas M. A., Hong I., Chiu S. L., Huganir R. L., et al. (2015) The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 525, 56–61 10.1038/nature14973 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Almeida S., Gascon E., Tran H., Chou H. J., Gendron T. F., Degroot S., Tapper A. R., Sellier C., Charlet-Berguerand N., Karydas A., Seeley W. W., Boxer A. L., Petrucelli L., Miller B. L., and Gao F. B. (2013) Modeling key pathological features of frontotemporal dementia with C9ORF72 repeat expansion in iPSC-derived human neurons. Acta Neuropathol. 126, 385–399 10.1007/s00401-013-1149-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Wen X., Westergard T., Pasinelli P., and Trotti D. (2017) Pathogenic determinants and mechanisms of ALS/FTD linked to hexanucleotide repeat expansions in the C9orf72 gene. Neurosci. Lett. 636, 16–26 10.1016/j.neulet.2016.09.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Jain A., and Vale R. D. (2017) RNA phase transitions in repeat expansion disorders. Nature 546, 243–247 10.1038/nature22386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Ladd P. D., Smith L. E., Rabaia N. A., Moore J. M., Georges S. A., Hansen R. S., Hagerman R. J., Tassone F., Tapscott S. J., and Filippova G. N. (2007) An antisense transcript spanning the CGG repeat region of FMR1 is upregulated in premutation carriers but silenced in full mutation individuals. Hum. Mol. Genet. 16, 3174–3187 10.1093/hmg/ddm293 [DOI] [PubMed] [Google Scholar]
- 91. Cho D. H., Thienes C. P., Mahoney S. E., Analau E., Filippova G. N., and Tapscott S. J. (2005) Antisense transcription and heterochromatin at the DM1 CTG repeats are constrained by CTCF. Mol. Cell 20, 483–489 10.1016/j.molcel.2005.09.002 [DOI] [PubMed] [Google Scholar]
- 92. Tabet R., Schaeffer L., Freyermuth F., Jambeau M., Workman M., Lee C. Z., Lin C. C., Jiang J., Jansen-West K., Abou-Hamdan H., Désaubry L., Gendron T., Petrucelli L., Martin F., and Lagier-Tourenne C. (2018) CUG initiation and frameshifting enable production of dipeptide repeat proteins from ALS/FTD C9ORF72 transcripts. Nat. Commun. 9, 152 10.1038/s41467-017-02643-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Green K. M., Glineburg M. R., Kearse M. G., Flores B. N., Linsalata A. E., Fedak S. J., Goldstrohm A. C., Barmada S. J., and Todd P. K. (2017) RAN translation at C9orf72-associated repeat expansions is selectively enhanced by the integrated stress response. Nat. Commun. 8, 2005 10.1038/s41467-017-02200-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Cheng W., Wang S., Mestre A. A., Fu C., Makarem A., Xian F., Hayes L. R., Lopez-Gonzalez R., Drenner K., Jiang J., Cleveland D. W., and Sun S. (2018) C9ORF72 GGGGCC repeat-associated non-AUG translation is upregulated by stress through eIF2α phosphorylation. Nat. Commun. 9, 51 10.1038/s41467-017-02495-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Walker C., Herranz-Martin S., Karyka E., Liao C., Lewis K., Elsayed W., Lukashchuk V., Chiang S. C., Ray S., Mulcahy P. J., Jurga M., Tsagakis I., Iannitti T., Chandran J., Coldicott I., et al. (2017) C9orf72 expansion disrupts ATM-mediated chromosomal break repair. Nat. Neurosci. 20, 1225–1235 10.1038/nn.4604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Donnelly C. J., Zhang P. W., Pham J. T., Haeusler A. R., Mistry N. A., Vidensky S., Daley E. L., Poth E. M., Hoover B., Fines D. M., Maragakis N., Tienari P. J., Petrucelli L., Traynor B. J., Wang J., et al. (2013) RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Neuron 80, 415–428 10.1016/j.neuron.2013.10.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Belzil V. V., Bauer P. O., Prudencio M., Gendron T. F., Stetler C. T., Yan I. K., Pregent L., Daughrity L., Baker M. C., Rademakers R., Boylan K., Patel T. C., Dickson D. W., and Petrucelli L. (2013) Reduced C9orf72 gene expression in c9FTD/ALS is caused by histone trimethylation, an epigenetic event detectable in blood. Acta Neuropathol. 126, 895–905 10.1007/s00401-013-1199-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Koppers M., Blokhuis A. M., Westeneng H. J., Terpstra M. L., Zundel C. A., Vieira de Sá R., Schellevis R. D., Waite A. J., Blake D. J., Veldink J. H., van den Berg L. H., and Pasterkamp R. J. (2015) C9orf72 ablation in mice does not cause motor neuron degeneration or motor deficits. Ann. Neurol. 78, 426–438 10.1002/ana.24453 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. O'Rourke J. G., Bogdanik L., Yáñez A., Lall D., Wolf A. J., Muhammad A. K., Ho R., Carmona S., Vit J. P., Zarrow J., Kim K. J., Bell S., Harms M. B., Miller T. M., Dangler C. A., et al. (2016) C9orf72 is required for proper macrophage and microglial function in mice. Science 351, 1324–1329 10.1126/science.aaf1064 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Lagier-Tourenne C., Baughn M., Rigo F., Sun S., Liu P., Li H. R., Jiang J., Watt A. T., Chun S., Katz M., Qiu J., Sun Y., Ling S. C., Zhu Q., Polymenidou M., et al. (2013) Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration. Proc. Natl. Acad. Sci. U.S.A. 110, E4530–E4539 10.1073/pnas.1318835110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Atanasio A., Decman V., White D., Ramos M., Ikiz B., Lee H. C., Siao C. J., Brydges S., LaRosa E., Bai Y., Fury W., Burfeind P., Zamfirova R., Warshaw G., Orengo J., et al. (2016) C9orf72 ablation causes immune dysregulation characterized by leukocyte expansion, autoantibody production, and glomerulonephropathy in mice. Sci. Rep. 6, 23204 10.1038/srep23204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Sullivan P. M., Zhou X., Robins A. M., Paushter D. H., Kim D., Smolka M. B., and Hu F. (2016) The ALS/FTLD associated protein C9orf72 associates with SMCR8 and WDR41 to regulate the autophagy-lysosome pathway. Acta Neuropathol. Commun. 4, 51 10.1186/s40478-016-0324-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Sudria-Lopez E., Koppers M., de Wit M., van der Meer C., Westeneng H. J., Zundel C. A., Youssef S. A., Harkema L., de Bruin A., Veldink J. H., van den Berg L. H., and Pasterkamp R. J. (2016) Full ablation of C9orf72 in mice causes immune system-related pathology and neoplastic events but no motor neuron defects. Acta Neuropathol. 132, 145–147 10.1007/s00401-016-1581-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Swinnen B., Bento-Abreu A., Gendron T. F., Boeynaems S., Bogaert E., Nuyts R., Timmers M., Scheveneels W., Hersmus N., Wang J., Mizielinska S., Isaacs A. M., Petrucelli L., Lemmens R., Van Damme P., et al. (2018) A zebrafish model for C9orf72 ALS reveals RNA toxicity as a pathogenic mechanism. Acta Neuropathol. 135, 427–443 10.1007/s00401-017-1796-5 [DOI] [PubMed] [Google Scholar]
- 105. Cooper-Knock J., Bury J. J., Heath P. R., Wyles M., Higginbottom A., Gelsthorpe C., Highley J. R., Hautbergue G., Rattray M., Kirby J., and Shaw P. J. (2015) C9ORF72 GGGGCC expanded repeats produce splicing dysregulation which correlates with disease severity in amyotrophic lateral sclerosis. PLoS ONE 10, e0127376 10.1371/journal.pone.0127376 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Barker H. V., Niblock M., Lee Y. B., Shaw C. E., and Gallo J. M. (2017) RNA misprocessing in C9orf72-linked neurodegeneration. Front. Cell. Neurosci. 11, 195 10.3389/fnhum.2017.00195,10.3389/fncel.2017.00195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Coyne A. N., Zaepfel B. L., and Zarnescu D. C. (2017) Failure to deliver and translate–new insights into RNA dysregulation in ALS. Front. Cell. Neurosci. 11, 243 10.3389/fncel.2017.00243 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Haeusler A. R., Donnelly C. J., Periz G., Simko E. A., Shaw P. G., Kim M. S., Maragakis N. J., Troncoso J. C., Pandey A., Sattler R., Rothstein J. D., and Wang J. (2014) C9orf72 nucleotide repeat structures initiate molecular cascades of disease. Nature 507, 195–200 10.1038/nature13124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Lee Y. B., Chen H. J., Peres J. N., Gomez-Deza J., Attig J., Stalekar M., Troakes C., Nishimura A. L., Scotter E. L., Vance C., Adachi Y., Sardone V., Miller J. W., Smith B. N., Gallo, et al. (2013) Hexanucleotide repeats in ALS/FTD form length-dependent RNA foci, sequester RNA-binding proteins, and are neurotoxic. Cell Rep. 5, 1178–1186 10.1016/j.celrep.2013.10.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Xu Z., Poidevin M., Li X., Li Y., Shu L., Nelson D. L., Li H., Hales C. M., Gearing M., Wingo T. S., and Jin P. (2013) Expanded GGGGCC repeat RNA associated with amyotrophic lateral sclerosis and frontotemporal dementia causes neurodegeneration. Proc. Natl. Acad. Sci. U.S.A. 110, 7778–7783 10.1073/pnas.1219643110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Mackenzie I. R., Arzberger T., Kremmer E., Troost D., Lorenzl S., Mori K., Weng S. M., Haass C., Kretzschmar H. A., Edbauer D., and Neumann M. (2013) Dipeptide repeat protein pathology in C9ORF72 mutation cases: clinico-pathological correlations. Acta Neuropathol. 126, 859–879 10.1007/s00401-013-1181-y [DOI] [PubMed] [Google Scholar]
- 112. Cooper-Knock J., Higginbottom A., Stopford M. J., Highley J. R., Ince P. G., Wharton S. B., Pickering-Brown S., Kirby J., Hautbergue G. M., and Shaw P. J. (2015) Antisense RNA foci in the motor neurons of C9ORF72-ALS patients are associated with TDP-43 proteinopathy. Acta Neuropathol. 130, 63–75 10.1007/s00401-015-1429-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Mori K., Lammich S., Mackenzie I. R., Forné I., Zilow S., Kretzschmar H., Edbauer D., Janssens J., Kleinberger G., Cruts M., Herms J., Neumann M., Van Broeckhoven C., Arzberger T., and Haass C. (2013) hnRNP A3 binds to GGGGCC repeats and is a constituent of p62-positive/TDP43-negative inclusions in the hippocampus of patients with C9orf72 mutations. Acta Neuropathol. 125, 413–423 10.1007/s00401-013-1088-7 [DOI] [PubMed] [Google Scholar]
- 114. Cooper-Knock J., Walsh M. J., Higginbottom A., Robin Highley J., Dickman M. J., Edbauer D., Ince P. G., Wharton S. B., Wilson S. A., Kirby J., Hautbergue G. M., and Shaw P. J. (2014) Sequestration of multiple RNA recognition motif-containing proteins by C9orf72 repeat expansions. Brain 137, 2040–2051 10.1093/brain/awu120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Sareen D., O'Rourke J. G., Meera P., Muhammad A. K., Grant S., Simpkinson M., Bell S., Carmona S., Ornelas L., Sahabian A., Gendron T., Petrucelli L., Baughn M., Ravits J., Harms M. B., et al. (2013) Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion. Sci. Transl. Med. 5, 208ra149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Zhang Y. J., Gendron T. F., Grima J. C., Sasaguri H., Jansen-West K., Xu Y. F., Katzman R. B., Gass J., Murray M. E., Shinohara M., Lin W. L., Garrett A., Stankowski J. N., Daughrity L., Tong J., et al. (2016) C9ORF72 poly(GA) aggregates sequester and impair HR23 and nucleocytoplasmic transport proteins. Nat. Neurosci. 19, 668–677 10.1038/nn.4272 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Mackenzie I. R., Frick P., and Neumann M. (2014) The neuropathology associated with repeat expansions in the C9ORF72 gene. Acta Neuropathol. 127, 347–357 10.1007/s00401-013-1232-4 [DOI] [PubMed] [Google Scholar]
- 118. Kwon I., Xiang S., Kato M., Wu L., Theodoropoulos P., Wang T., Kim J., Yun J., Xie Y., and McKnight S. L. (2014) Poly-dipeptides encoded by the C9orf72 repeats bind nucleoli, impede RNA biogenesis, and kill cells. Science 345, 1139–1145 10.1126/science.1254917 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Mizielinska S., Grönke S., Niccoli T., Ridler C. E., Clayton E. L., Devoy A., Moens T., Norona F. E., Woollacott I. O. C., Pietrzyk J., Cleverley K., Nicoll A. J., Pickering-Brown S., Dols J., Cabecinha M., et al. (2014) C9orf72 repeat expansions cause neurodegeneration in Drosophila through arginine-rich proteins. Science 345, 1192–1194 10.1126/science.1256800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Wen X., Tan W., Westergard T., Krishnamurthy K., Markandaiah S. S., Shi Y., Lin S., Shneider N. A., Monaghan J., Pandey U. B., Pasinelli P., Ichida J. K., and Trotti D. (2014) Antisense proline-arginine RAN dipeptides linked to C9ORF72-ALS/FTD form toxic nuclear aggregates that initiate in vitro and in vivo neuronal death. Neuron 84, 1213–1225 10.1016/j.neuron.2014.12.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Yamakawa M., Ito D., Honda T., Kubo K., Noda M., Nakajima K., and Suzuki N. (2015) Characterization of the dipeptide repeat protein in the molecular pathogenesis of c9FTD/ALS. Hum. Mol. Genet. 24, 1630–1645 10.1093/hmg/ddu576 [DOI] [PubMed] [Google Scholar]
- 122. Zhang Y. J., Jansen-West K., Xu Y. F., Gendron T. F., Bieniek K. F., Lin W. L., Sasaguri H., Caulfield T., Hubbard J., Daughrity L., Chew J., Belzil V. V., Prudencio M., Stankowski J. N., Castanedes-Casey M., et al. (2014) Aggregation-prone c9FTD/ALS poly(GA) RAN-translated proteins cause neurotoxicity by inducing ER stress. Acta Neuropathol. 128, 505–524 10.1007/s00401-014-1336-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Mori K., Arzberger T., Grässer F. A., Gijselinck I., May S., Rentzsch K., Weng S. M., Schludi M. H., van der Zee J., Cruts M., Van Broeckhoven C., Kremmer E., Kretzschmar H. A., Haass C., and Edbauer D. (2013) Bidirectional transcripts of the expanded C9orf72 hexanucleotide repeat are translated into aggregating dipeptide repeat proteins. Acta Neuropathol. 126, 881–893 10.1007/s00401-013-1189-3 [DOI] [PubMed] [Google Scholar]
- 124. Gendron T. F., Bieniek K. F., Zhang Y. J., Jansen-West K., Ash P. E., Caulfield T., Daughrity L., Dunmore J. H., Castanedes-Casey M., Chew J., Cosio D. M., van Blitterswijk M., Lee W. C., Rademakers R., Boylan K. B., et al. (2013) Antisense transcripts of the expanded C9ORF72 hexanucleotide repeat form nuclear RNA foci and undergo repeat-associated non-ATG translation in c9FTD/ALS. Acta Neuropathol. 126, 829–844 10.1007/s00401-013-1192-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Lee K. H., Zhang P., Kim H. J., Mitrea D. M., Sarkar M., Freibaum B. D., Cika J., Coughlin M., Messing J., Molliex A., Maxwell B. A., Kim N. C., Temirov J., Moore J., Kolaitis R. M., et al. (2016) C9orf72 dipeptide repeats impair the assembly, dynamics, and function of membrane-less organelles. Cell 167, 774–788.e17 10.1016/j.cell.2016.10.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Lin Y., Mori E., Kato M., Xiang S., Wu L., Kwon I., and McKnight S. L. (2016) Toxic PR poly-dipeptides encoded by the C9orf72 repeat expansion target LC domain polymers. Cell 167, 789–802.e12 10.1016/j.cell.2016.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Tao Z., Wang H., Xia Q., Li K., Li K., Jiang X., Xu G., Wang G., and Ying Z. (2015) Nucleolar stress and impaired stress granule formation contribute to C9orf72 RAN translation-induced cytotoxicity. Hum. Mol. Genet. 24, 2426–2441 10.1093/hmg/ddv005 [DOI] [PubMed] [Google Scholar]
- 128. Kanekura K., Yagi T., Cammack A. J., Mahadevan J., Kuroda M., Harms M. B., Miller T. M., and Urano F. (2016) Poly-dipeptides encoded by the C9ORF72 repeats block global protein translation. Hum. Mol. Genet. 25, 1803–1813 10.1093/hmg/ddw052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Yin S., Lopez-Gonzalez R., Kunz R. C., Gangopadhyay J., Borufka C., Gygi S. P., Gao F. B., and Reed R. (2017) Evidence that C9ORF72 dipeptide repeat proteins associate with U2 snRNP to cause mis-splicing in ALS/FTD patients. Cell Rep. 19, 2244–2256 10.1016/j.celrep.2017.05.056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Ohki Y., Wenninger-Weinzierl A., Hruscha A., Asakawa K., Kawakami K., Haass C., Edbauer D., and Schmid B. (2017) Glycine-alanine dipeptide repeat protein contributes to toxicity in a zebrafish model of C9orf72 associated neurodegeneration. Mol. Neurodegener. 12, 6 10.1186/s13024-016-0146-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. May S., Hornburg D., Schludi M. H., Arzberger T., Rentzsch K., Schwenk B. M., Grässer F. A., Mori K., Kremmer E., Banzhaf-Strathmann J., Mann M., Meissner F., and Edbauer D. (2014) C9orf72 FTLD/ALS-associated Gly-Ala dipeptide repeat proteins cause neuronal toxicity and Unc119 sequestration. Acta Neuropathol. 128, 485–503 10.1007/s00401-014-1329-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Yang D., Abdallah A., Li Z., Lu Y., Almeida S., and Gao F. B. (2015) FTD/ALS-associated poly(GR) protein impairs the Notch pathway and is recruited by poly(GA) into cytoplasmic inclusions. Acta Neuropathol. 130, 525–535 10.1007/s00401-015-1448-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Guo Q., Lehmer C., Martínez-Sánchez A., Rudack T., Beck F., Hartmann H., Pérez-Berlanga M., Frottin F., Hipp M. S., Hartl F. U., Edbauer D., Baumeister W., and Fernández-Busnadiego R. (2018) In situ structure of neuronal C9orf72 poly(GA) aggregates reveals proteasome recruitment. Cell 172, 696–705.e12 10.1016/j.cell.2017.12.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Bäuerlein F. J. B., Saha I., Mishra A., Kalemanov M., Martínez-Sánchez A., Klein R., Dudanova I., Hipp M. S., Hartl F. U., Baumeister W., and Fernández-Busnadiego R. (2017) In situ architecture and cellular interactions of polyQ inclusions. Cell 171, 179–187.e10 10.1016/j.cell.2017.08.009 [DOI] [PubMed] [Google Scholar]
- 135. Lopez-Gonzalez R., Lu Y., Gendron T. F., Karydas A., Tran H., Yang D., Petrucelli L., Miller B. L., Almeida S., and Gao F. B. (2016) Poly(GR) in C9ORF72-related ALS/FTD compromises mitochondrial function and increases oxidative stress and DNA damage in iPSC-derived motor neurons. Neuron 92, 383–391 10.1016/j.neuron.2016.09.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Su X. A., Dion V., Gasser S. M., and Freudenreich C. H. (2015) Regulation of recombination at yeast nuclear pores controls repair and triplet repeat stability. Genes Dev. 29, 1006–1017 10.1101/gad.256404.114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Dansithong W., Jog S. P., Paul S., Mohammadzadeh R., Tring S., Kwok Y., Fry R. C., Marjoram P., Comai L., and Reddy S. (2011) RNA steady-state defects in myotonic dystrophy are linked to nuclear exclusion of SHARP. EMBO Rep. 12, 735–742 10.1038/embor.2011.86 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Xia J., Lee D. H., Taylor J., Vandelft M., and Truant R. (2003) Huntingtin contains a highly conserved nuclear export signal. Hum. Mol. Genet. 12, 1393–1403 10.1093/hmg/ddg156 [DOI] [PubMed] [Google Scholar]
- 139. Suhr S. T., Senut M. C., Whitelegge J. P., Faull K. F., Cuizon D. B., and Gage F. H. (2001) Identities of sequestered proteins in aggregates from cells with induced polyglutamine expression. J. Cell Biol. 153, 283–294 10.1083/jcb.153.2.283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Cornett J., Cao F., Wang C. E., Ross C. A., Bates G. P., Li S. H., and Li X. J. (2005) Polyglutamine expansion of huntingtin impairs its nuclear export. Nat. Genet. 37, 198–204 10.1038/ng1503 [DOI] [PubMed] [Google Scholar]
- 141. Taylor J., Grote S. K., Xia J., Vandelft M., Graczyk J., Ellerby L. M., La Spada A. R., and Truant R. (2006) Ataxin-7 can export from the nucleus via a conserved exportin-dependent signal. J. Biol. Chem. 281, 2730–2739 10.1074/jbc.M506751200 [DOI] [PubMed] [Google Scholar]
- 142. Chai Y., Shao J., Miller V. M., Williams A., and Paulson H. L. (2002) Live-cell imaging reveals divergent intracellular dynamics of polyglutamine disease proteins and supports a sequestration model of pathogenesis. Proc. Natl. Acad. Sci. U.S.A. 99, 9310–9315 10.1073/pnas.152101299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Yazawa I. (2000) Aberrant phosphorylation of dentatorubral-pallidoluysian atrophy (DRPLA) protein complex in brain tissue. Biochem. J. 351, 587–593 10.1042/bj3510587 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Takahashi H., Egawa S., Piao Y. S., Hayashi S., Yamada M., Shimohata T., Oyanagi K., and Tsuji S. (2001) Neuronal nuclear alterations in dentatorubral-pallidoluysian atrophy: ultrastructural and morphometric studies of the cerebellar granule cells. Brain Res. 919, 12–19 10.1016/S0006-8993(01)02986-9 [DOI] [PubMed] [Google Scholar]
- 145. Grima J. C., Daigle J. G., Arbez N., Cunningham K. C., Zhang K., Ochaba J., Geater C., Morozko E., Stocksdale J., Glatzer J. C., Pham J. T., Ahmed I., Peng Q., Wadhwa H., Pletnikova O., et al. (2017) Mutant Huntingtin disrupts the nuclear pore complex. Neuron 94, 93–107.e6 10.1016/j.neuron.2017.03.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Gasset-Rosa F., Chillon-Marinas C., Goginashvili A., Atwal R. S., Artates J. W., Tabet R., Wheeler V. C., Bang A. G., Cleveland D. W., and Lagier-Tourenne C. (2017) Polyglutamine-expanded Huntingtin exacerbates age-related disruption of nuclear integrity and nucleocytoplasmic transport. Neuron 94, 48–57.e4 10.1016/j.neuron.2017.03.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Woerner A. C., Frottin F., Hornburg D., Feng L. R., Meissner F., Patra M., Tatzelt J., Mann M., Winklhofer K. F., Hartl F. U., and Hipp M. S. (2016) Cytoplasmic protein aggregates interfere with nucleocytoplasmic transport of protein and RNA. Science 351, 173–176 10.1126/science.aad2033 [DOI] [PubMed] [Google Scholar]
- 148. Ishiura H., and Tsuji S. (2015) Epidemiology and molecular mechanism of frontotemporal lobar degeneration/amyotrophic lateral sclerosis with repeat expansion mutation in C9orf72. J. Neurogenet. 29, 85–94 10.3109/01677063.2015.1085980 [DOI] [PubMed] [Google Scholar]
- 149. Boeynaems S., Bogaert E., Michiels E., Gijselinck I., Sieben A., Jovičić A., De Baets G., Scheveneels W., Steyaert J., Cuijt I., Verstrepen K. J., Callaerts P., Rousseau F., Schymkowitz J., Cruts M., et al. (2016) Drosophila screen connects nuclear transport genes to DPR pathology in c9ALS/FTD. Sci. Rep. 6, 20877 10.1038/srep20877 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Khosravi B., Hartmann H., May S., Möhl C., Ederle H., Michaelsen M., Schludi M. H., Dormann D., and Edbauer D. (2017) Cytoplasmic poly(GA) aggregates impair nuclear import of TDP-43 in C9orf72 ALS/FTLD. Hum. Mol. Genet. 26, 790–800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Kinoshita Y., Ito H., Hirano A., Fujita K., Wate R., Nakamura M., Kaneko S., Nakano S., and Kusaka H. (2009) Nuclear contour irregularity and abnormal transporter protein distribution in anterior horn cells in amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 68, 1184–1192 10.1097/NEN.0b013e3181bc3bec [DOI] [PubMed] [Google Scholar]
- 152. Kaneb H. M., Folkmann A. W., Belzil V. V., Jao L. E., Leblond C. S., Girard S. L., Daoud H., Noreau A., Rochefort D., Hince P., Szuto A., Levert A., Vidal S., André-Guimont C., Camu W., et al. (2015) Deleterious mutations in the essential mRNA metabolism factor, hGle1, in amyotrophic lateral sclerosis. Hum. Mol. Genet. 24, 1363–1373 10.1093/hmg/ddu545 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Nishimura A. L., Zupunski V., Troakes C., Kathe C., Fratta P., Howell M., Gallo J. M., Hortobágyi T., Shaw C. E., and Rogelj B. (2010) Nuclear import impairment causes cytoplasmic trans-activation response DNA-binding protein accumulation and is associated with frontotemporal lobar degeneration. Brain 133, 1763–1771 10.1093/brain/awq111 [DOI] [PubMed] [Google Scholar]
- 154. Chou C. C., Zhang Y., Umoh M. E., Vaughan S. W., Lorenzini I., Liu F., Sayegh M., Donlin-Asp P. G., Chen Y. H., Duong D. M., Seyfried N. T., Powers M. A., Kukar T., Hales C. M., Gearing M., et al. (2018) TDP-43 pathology disrupts nuclear pore complexes and nucleocytoplasmic transport in ALS/FTD. Nat. Neurosci. 21, 228–239 10.1038/s41593-017-0047-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Kramer N. J., Haney M. S., Morgens D. W., Jovičić A., Couthouis J., Li A., Ousey J., Ma R., Bieri G., Tsui C. K., Shi Y., Hertz N. T., Tessier-Lavigne M., Ichida J. K., Bassik M. C., and Gitler A. D. (2018) CRISPR-Cas9 screens in human cells and primary neurons identify modifiers of C9ORF72 dipeptide-repeat-protein toxicity. Nat. Genet. 50, 603–612 10.1038/s41588-018-0070-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Shi K. Y., Mori E., Nizami Z. F., Lin Y., Kato M., Xiang S., Wu L. C., Ding M., Yu Y., Gall J. G., and McKnight S. L. (2017) Toxic PRn poly dipeptides encoded by the C9orf72 repeat expansion block nuclear import and export. Proc. Natl. Acad. Sci. U.S.A. 114, E1111–E1117 10.1073/pnas.1620293114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. O'Rourke J. G., Bogdanik L., Muhammad A. K. M. G., Gendron T. F., Kim K. J., Austin A., Cady J., Liu E. Y., Zarrow J., Grant S., Ho R., Bell S., Carmona S., Simpkinson M., Lall D., et al. (2015) C9orf72 BAC transgenic mice display typical pathologic features of ALS/FTD. Neuron 88, 892–901 10.1016/j.neuron.2015.10.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Peters O. M., Cabrera G. T., Tran H., Gendron T. F., McKeon J. E., Metterville J., Weiss A., Wightman N., Salameh J., Kim J., Sun H., Boylan K. B., Dickson D., Kennedy Z., Lin Z., et al. (2015) Human C9ORF72 hexanucleotide expansion reproduces RNA foci and dipeptide repeat proteins but not neurodegeneration in BAC transgenic mice. Neuron 88, 902–909 10.1016/j.neuron.2015.11.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Jiang J., Zhu Q., Gendron T. F., Saberi S., McAlonis-Downes M., Seelman A., Stauffer J. E., Jafar-Nejad P., Drenner K., Schulte D., Chun S., Sun S., Ling S. C., Myers B., Engelhardt J., et al. (2016) Gain of toxicity from ALS/FTD-linked repeat expansions in C9ORF72 is alleviated by antisense oligonucleotides targeting GGGGCC-containing RNAs. Neuron 90, 535–550 10.1016/j.neuron.2016.04.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Liu Y., Pattamatta A., Zu T., Reid T., Bardhi O., Borchelt D. R., Yachnis A. T., and Ranum L. P. (2016) C9orf72 BAC mouse model with motor deficits and neurodegenerative features of ALS/FTD. Neuron 90, 521–534 10.1016/j.neuron.2016.04.005 [DOI] [PubMed] [Google Scholar]
- 161. Chew J., Gendron T. F., Prudencio M., Sasaguri H., Zhang Y. J., Castanedes-Casey M., Lee C. W., Jansen-West K., Kurti A., Murray M. E., Bieniek K. F., Bauer P. O., Whitelaw E. C., Rousseau L., Stankowski J. N., et al. (2015) Neurodegeneration. C9ORF72 repeat expansions in mice cause TDP-43 pathology, neuronal loss, and behavioral deficits. Science 348, 1151–1154 10.1126/science.aaa9344 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Batra R., and Lee C. W. (2017) Mouse models of C9orf72 hexanucleotide repeat expansion in amyotrophic lateral sclerosis/frontotemporal dementia. Front. Cell. Neurosci. 11, 196 10.3389/fncel.2017.00196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Schludi M. H., Becker L., Garrett L., Gendron T. F., Zhou Q., Schreiber F., Popper B., Dimou L., Strom T. M., Winkelmann J., von Thaden A., Rentzsch K., May S., Michaelsen M., Schwenk B. M., et al. (2017) Spinal poly(GA) inclusions in a C9orf72 mouse model trigger motor deficits and inflammation without neuron loss. Acta Neuropathol. 134, 241–254 10.1007/s00401-017-1711-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Kordasiewicz H. B., Stanek L. M., Wancewicz E. V., Mazur C., McAlonis M. M., Pytel K. A., Artates J. W., Weiss A., Cheng S. H., Shihabuddin L. S., Hung G., Bennett C. F., and Cleveland D. W. (2012) Sustained therapeutic reversal of Huntington's disease by transient repression of huntingtin synthesis. Neuron 74, 1031–1044 10.1016/j.neuron.2012.05.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Kumar A., Kumar Singh S., Kumar V., Kumar D., Agarwal S., and Rana M. K. (2015) Huntington's disease: an update of therapeutic strategies. Gene 556, 91–97 10.1016/j.gene.2014.11.022 [DOI] [PubMed] [Google Scholar]
- 166. Wheeler T. M., Leger A. J., Pandey S. K., MacLeod A. R., Nakamori M., Cheng S. H., Wentworth B. M., Bennett C. F., and Thornton C. A. (2012) Targeting nuclear RNA for in vivo correction of myotonic dystrophy. Nature 488, 111–115 10.1038/nature11362 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Gao Z., and Cooper T. A. (2013) Antisense oligonucleotides: rising stars in eliminating RNA toxicity in myotonic dystrophy. Hum. Gene Ther. 24, 499–507 10.1089/hum.2012.212 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Evers M. M., Toonen L. J., and van Roon-Mom W. M. (2015) Antisense oligonucleotides in therapy for neurodegenerative disorders. Adv. Drug Deliv. Rev. 87, 90–103 10.1016/j.addr.2015.03.008 [DOI] [PubMed] [Google Scholar]
- 169. Schoch K. M., and Miller T. M. (2017) Antisense oligonucleotides: translation from mouse models to human neurodegenerative diseases. Neuron 94, 1056–1070 10.1016/j.neuron.2017.04.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Kramer N. J., Carlomagno Y., Zhang Y. J., Almeida S., Cook C. N., Gendron T. F., Prudencio M., Van Blitterswijk M., Belzil V., Couthouis J., Paul J. W. 3rd., Goodman L. D., Daughrity L., Chew J., Garrett A., et al. (2016) Spt4 selectively regulates the expression of C9orf72 sense and antisense mutant transcripts. Science 353, 708–712 10.1126/science.aaf7791 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Liu C. R., Chang C. R., Chern Y., Wang T. H., Hsieh W. C., Shen W. C., Chang C. Y., Chu I. C., Deng N., Cohen S. N., and Cheng T. H. (2012) Spt4 is selectively required for transcription of extended trinucleotide repeats. Cell 148, 690–701 10.1016/j.cell.2011.12.032 [DOI] [PubMed] [Google Scholar]
- 172. Cheng H. M., Chern Y., Chen I. H., Liu C. R., Li S. H., Chun S. J., Rigo F., Bennett C. F., Deng N., Feng Y., Lin C. S., Yan Y. T., Cohen S. N., and Cheng T. H. (2015) Effects on murine behavior and lifespan of selectively decreasing expression of mutant huntingtin allele by supt4h knockdown. PLoS Genet. 11, e1005043 10.1371/journal.pgen.1005043 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Pinto B. S., Saxena T., Oliveira R., Méndez-Gómez H. R., Cleary J. D., Denes L. T., McConnell O., Arboleda J., Xia G., Swanson M. S., and Wang E. T. (2017) Impeding transcription of expanded microsatellite repeats by deactivated Cas9. Mol. Cell 68, 479–490.e5 10.1016/j.molcel.2017.09.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Batra R., Nelles D. A., Pirie E., Blue S. M., Marina R. J., Wang H., Chaim I. A., Thomas J. D., Zhang N., Nguyen V., Aigner S., Markmiller S., Xia G., Corbett K. D., Swanson M. S., and Yeo G. W. (2017) Elimination of toxic microsatellite repeat expansion RNA by RNA-targeting Cas9. Cell 170, 899–912.e10 10.1016/j.cell.2017.07.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Su Z., Zhang Y., Gendron T. F., Bauer P. O., Chew J., Yang W. Y., Fostvedt E., Jansen-West K., Belzil V. V., Desaro P., Johnston A., Overstreet K., Oh S. Y., Todd P. K., Berry J. D., et al. (2014) Discovery of a biomarker and lead small molecules to target r(GGGGCC)-associated defects in c9FTD/ALS. Neuron 83, 1043–1050 10.1016/j.neuron.2014.07.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Kong H. E., Zhao J., Xu S., Jin P., and Jin Y. (2017) Fragile X-associated tremor/ataxia syndrome: from molecular pathogenesis to development of therapeutics. Front. Cell. Neurosci. 11, 128 10.3389/fncel.2017.00128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Childs-Disney J. L., Hoskins J., Rzuczek S. G., Thornton C. A., and Disney M. D. (2012) Rationally designed small molecules targeting the RNA that causes myotonic dystrophy type 1 are potently bioactive. ACS Chem. Biol. 7, 856–862 10.1021/cb200408a [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Disney M. D., Liu B., Yang W. Y., Sellier C., Tran T., Charlet-Berguerand N., and Childs-Disney J. L. (2012) A small molecule that targets r(CGG)(exp) and improves defects in fragile X-associated tremor ataxia syndrome. ACS Chem. Biol. 7, 1711–1718 10.1021/cb300135h [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Yang W. Y., He F., Strack R. L., Oh S. Y., Frazer M., Jaffrey S. R., Todd P. K., and Disney M. D. (2016) Small molecule recognition and tools to study modulation of r(CGG)(exp) in fragile X-associated tremor ataxia syndrome. ACS Chem. Biol. 11, 2456–2465 10.1021/acschembio.6b00147 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Simone R., Balendra R., Moens T. G., Preza E., Wilson K. M., Heslegrave A., Woodling N. S., Niccoli T., Gilbert-Jaramillo J., Abdelkarim S., Clayton E. L., Clarke M., Konrad M. T., Nicoll A. J., Mitchell J. S., et al. (2018) G-quadruplex-binding small molecules ameliorate C9orf72 FTD/ALS pathology in vitro and in vivo. EMBO Mol. Med. 10, 22–31 10.15252/emmm.201707850 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Siboni R. B., Nakamori M., Wagner S. D., Struck A. J., Coonrod L. A., Harriott S. A., Cass D. M., Tanner M. K., and Berglund J. A. (2015) Actinomycin D specifically reduces expanded CUG repeat RNA in myotonic dystrophy models. Cell Rep. 13, 2386–2394 10.1016/j.celrep.2015.11.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Boyce M., Bryant K. F., Jousse C., Long K., Harding H. P., Scheuner D., Kaufman R. J., Ma D., Coen D. M., Ron D., and Yuan J. (2005) A selective inhibitor of eIF2α dephosphorylation protects cells from ER stress. Science 307, 935–939 10.1126/science.1101902 [DOI] [PubMed] [Google Scholar]
- 183. Elia A. E., Lalli S., Monsurrò M. R., Sagnelli A., Taiello A. C., Reggiori B., La Bella V., Tedeschi G., and Albanese A. (2016) Tauroursodeoxycholic acid in the treatment of patients with amyotrophic lateral sclerosis. Eur. J. Neurol. 23, 45–52 10.1111/ene.12664 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Bott L. C., Badders N. M., Chen K. L., Harmison G. G., Bautista E., Shih C. C., Katsuno M., Sobue G., Taylor J. P., Dantuma N. P., Fischbeck K. H., and Rinaldi C. (2016) A small-molecule Nrf1 and Nrf2 activator mitigates polyglutamine toxicity in spinal and bulbar muscular atrophy. Hum. Mol. Genet. 25, 1979–1989 10.1093/hmg/ddw073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Jackrel M. E., DeSantis M. E., Martinez B. A., Castellano L. M., Stewart R. M., Caldwell K. A., Caldwell G. A., and Shorter J. (2014) Potentiated Hsp104 variants antagonize diverse proteotoxic misfolding events. Cell 156, 170–182 10.1016/j.cell.2013.11.047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Shorter J. (2017) Designer protein disaggregases to counter neurodegenerative disease. Curr. Opin. Genet. Dev. 44, 1–8 10.1016/j.gde.2017.01.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Hautbergue G. M., Castelli L. M., Ferraiuolo L., Sanchez-Martinez A., Cooper-Knock J., Higginbottom A., Lin Y. H., Bauer C. S., Dodd J. E., Myszczynska M. A., Alam S. M., Garneret P., Chandran J. S., Karyka E., Stopford M. J., et al. (2017) SRSF1-dependent nuclear export inhibition of C9ORF72 repeat transcripts prevents neurodegeneration and associated motor deficits. Nat. Commun. 8, 16063 10.1038/ncomms16063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Zhou Q., Lehmer C., Michaelsen M., Mori K., Alterauge D., Baumjohann D., Schludi M. H., Greiling J., Farny D., Flatley A., Feederle R., May S., Schreiber F., Arzberger T., Kuhm C., et al. (2017) Antibodies inhibit transmission and aggregation of C9orf72 poly(GA) dipeptide repeat proteins. EMBO Mol. Med. 9, 687–702 10.15252/emmm.201607054 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Rusmini P., Cristofani R., Galbiati M., Cicardi M. E., Meroni M., Ferrari V., Vezzoli G., Tedesco B., Messi E., Piccolella M., Carra S., Crippa V., and Poletti A. (2017) The role of the heat-shock protein B8 (HSPB8) in motoneuron diseases. Front. Mol. Neurosci. 10, 176 10.3389/fnmol.2017.00176 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Cristofani R., Crippa V., Vezzoli G., Rusmini P., Galbiati M., Cicardi M. E., Meroni M., Ferrari V., Tedesco B., Piccolella M., Messi E., Carra S., and Poletti A. (2018) The small heat-shock protein B8 (HSPB8) efficiently removes aggregating species of dipeptides produced in C9ORF72-related neurodegenerative diseases. Cell Stress Chaperones 23, 1–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Mann D. M., Rollinson S., Robinson A., Bennion Callister J., Thompson J. C., Snowden J. S., Gendron T., Petrucelli L., Masuda-Suzukake M., Hasegawa M., Davidson Y., and Pickering-Brown S. (2013) Dipeptide repeat proteins are present in the p62 positive inclusions in patients with frontotemporal lobar degeneration and motor neurone disease associated with expansions in C9ORF72. Acta Neuropathol. Commun. 1, 68 10.1186/2051-5960-1-68 [DOI] [PMC free article] [PubMed] [Google Scholar]


