1,275
Views
15
CrossRef citations to date
0
Altmetric
Editorial

c9RAN translation: a potential therapeutic target for the treatment of amyotrophic lateral sclerosis and frontotemporal dementia

, &
Pages 991-995 | Published online: 12 Jul 2013

Abstract

A hexanucleotide (GGGGCC) repeat expansion within a non-coding region of the C9ORF72 gene is the most common mutation associated with both frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Elucidating how these expanded repeats (GGGGCCexp) cause ‘c9FTD/ALS' has since become an important goal of the FTD/ALS field. GGGGCCexp transcripts aggregate into discrete nuclear structures, termed RNA foci. This phenomenon, observed in various repeat expansion disorders, is associated with RNA-binding protein sequestration. Of note, recent findings show that GGGGCCexp transcripts also succumb to an alternative fate: repeat-associated non-ATG translation (RAN translation). This unconventional mode of translation, which occurs in the absence of an initiating codon, results in the production of polyGA, polyGP and polyGR peptides. Antibodies generated against these peptides detect high molecular weight, insoluble material in brain homogenates, as well as neuronal inclusions throughout the central nervous system of c9FTD/ALS cases. Given that both foci formation and RAN translation in c9FTD/ALS require the synthesis of GGGGCCexp RNA, therapeutic strategies that target these transcripts and result in their neutralization or degradation could effectively block these two potential pathogenic mechanisms and provide a much needed treatment for c9FTD/ALS.

RNA-mediated toxicity has emerged as an important pathological mechanism in several neurological disorders caused by genomic expansions of microsatellite repeats, including myotonic dystrophy (DM), spinocerebellar ataxias (SCA), and fragile X-associated tremor ataxia syndrome (FXTAS) Citation[1]. Recently added to this list are frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) caused by an expansion of the GGGGCC repeat in the C9ORF72 gene from fewer than 30 copies to hundreds or thousands of copies Citation[2,3].

The pathology of ‘c9FTD/ALS,' and its clinical presentations, conform to those of classical FTD and ALS, which are thought to represent a continuum of disease. FTD results from the degeneration of frontal and temporal lobes and encompasses a group of disorders distinguished clinically by abnormalities in behavior, language and personality. ALS is characterized by the degeneration of motor neurons, leading to muscle atrophy and paralysis. Some FTD patients develop motor neuron dysfunction resembling ALS, and ALS patients often develop abnormalities characteristic of FTD. Disease onset is variable among C9ORF72 mutation carriers, as is clinical presentation, and there is evidence of incomplete disease penetrance. Determining whether repeat length influences these features, and whether genetic or environmental modifiers are involved, will certainly offer insight into mechanisms of disease. Neuropathologically, TDP-43-positive neuronal and glial inclusions typical of both disorders are observed in c9FTD/ALS patients. In addition, TDP-43-negative, p62-positive inclusions in the cerebellum and hippocampus, as well as neuronal inclusions composed of aberrant peptides produced via unconventional translation of GGGGCCexp RNA, are a specific pathology in patients with the C9ORF72 mutation Citation[4,5].

As with many microsatellite expansion disorders, the hexanucleotide repeat in C9ORF72 is present within a non-coding region. Efforts to elucidate how such expansions cause disease have led to the discovery of various mechanisms involving the production of repeat-containing RNA. The accumulation of these transcripts, often observed as nuclear RNA foci, induces cellular toxicity by activating downstream signaling pathways and disrupting the function of RNA-binding proteins Citation[1]. For example, RNA transcripts of an expanded CTG repeat in DMPK fold into hairpins that bind and inactivate muscleblind-like 1 protein (MBNL1) in DM1 Citation[6-8]. MBNL1 sequestration results in the mis-splicing of a subset of pre-mRNAs, such as the muscle-specific chloride ion channel, which accounts for characteristic features of DM1 Citation[9,10]. Transcripts of the GGGGCCexp repeat in C9ORF72 similarly accumulate as nuclear RNA foci in c9FTD/ALS Citation[2], implicating RNA-binding protein sequestration in disease pathogenesis. A second toxic mechanism stems from the susceptibility of such transcripts to repeat-associated non-ATG translation (RAN translation). This atypical form of translation, which occurs in the absence of an initiating codon, was first described by the Ranum group, who found that RAN translation occurs in all reading frames across long CAG repeats to produce homopolymeric proteins (polyQ, polyS, or polyA) Citation[11]. While RAN translation remains poorly understood, it appears to depend on repeat-length and hairpin formation, at least for CAGexp RNA Citation[11]. It may involve an internal ribosome entry site (IRES)-like mechanism; hairpins can recruit initiation factors and ribosomal subunits to IRESs, and certain viruses use an IRES to initiate translation at non-AUG sites Citation[12].

Given that expression of RAN translation products is sufficient to cause apoptosis in cultured cells, their accumulation may contribute to the neurodegeneration in repeat diseases, such as DM1 and SCA8 Citation[11]. Researchers also recently demonstrated that the CGG repeat expansion in FXTAS triggers RAN translational initiation within the 5′ UTR of FMR1 mRNA to generate a cryptic polyG-containing protein Citation[13]. As mentioned above, expanded GGGGCC repeats undergo RAN translation in c9FTD/ALS, and this results in the synthesis of polyGA, polyGP and polyGR peptides Citation[4,5]. Antibodies generated against these peptides specifically detect high molecular weight, insoluble material in c9FTD/ALS brain homogenates, as well as neuronal inclusions in c9FTD/ALS brain sections Citation[4,5]. Moreover, evidence that the mutant C9ORF72 allele is bidirectionally transcribed Citation[5] introduces the possibility that up to five RAN translated products may be expressed in c9FTD/ALS.

While the exact mechanisms of toxicity at play in c9FTD/ALS have yet to be fully elucidated, the need for treatment is no less crucial. Taking cues from investigations of other neurodegenerative diseases, one can surmise that the accumulation of aggregation-prone peptides resulting from RAN translation could be harmful to neurons. The inclusions may themselves be neurotoxic or they may sequester proteins causing loss of function, overwhelm protein degradation systems, or displace cytoplasmic organelles. Furthermore, RNA foci in c9FTD/ALS are expected to sequester various RNA-binding proteins and cause the misregulation of numerous downstream RNA targets Citation[14-16]. For example, the RNA-binding proteins hnRNPA2/B1 and hnRNPA1, mutations in which cause ALS and related disorders Citation[17], have been found to bind GGGGCCexp transcripts Citation[16].

Given that both foci formation and RAN translation in c9FTD/ALS require the synthesis of GGGGCCexp transcripts, strategies that target these transcripts and result in their neutralization or degradation could effectively block an early culprit in these putative mechanisms of disease (). No study examining the protective effect of targeting GGGGCCexp RNA has yet been published; however, investigations have no doubt been underway since the discovery that mutant C9ORF72 causes c9FTD/ALS. Based on advances made in the development of therapeutic strategies for other repeat expansion diseases marked by RNA toxicity, initial progress is expected to be relatively rapid. Antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), and small molecules are but some of the approaches used to target pathogenic RNAs.

Figure 1. Potential mechanisms of disease in c9FTD/ALS. Expansion of the GGGGCC hexanucleotide repeat within intron 1 of the C9ORF72 gene may cause c9FTD/ALS via RNA-dependent mechanisms. RNA transcripts resulting from transcription of GGGGCCexp aggregate into nuclear RNA foci that can sequester RNA-binding proteins and cause their loss of function. GGGGCCexp RNA transcripts are also susceptible to repeat-associated non-ATG translation, producing polyGA, polyGP and polyGR peptides, which form neuronal inclusions in c9FTD/ALS. Therapeutic strategies that target GGGGCCexp RNA transcripts may block both these putative pathogenic mechanisms. Moreover, while the mechanisms regulating RAN translation are poorly understood, and the means by which GGGGCCexp transcripts make their way to the cytoplasm are not yet known, targeting these events may also provide an approach to block RAN translation.

Figure 1. Potential mechanisms of disease in c9FTD/ALS. Expansion of the GGGGCC hexanucleotide repeat within intron 1 of the C9ORF72 gene may cause c9FTD/ALS via RNA-dependent mechanisms. RNA transcripts resulting from transcription of GGGGCCexp aggregate into nuclear RNA foci that can sequester RNA-binding proteins and cause their loss of function. GGGGCCexp RNA transcripts are also susceptible to repeat-associated non-ATG translation, producing polyGA, polyGP and polyGR peptides, which form neuronal inclusions in c9FTD/ALS. Therapeutic strategies that target GGGGCCexp RNA transcripts may block both these putative pathogenic mechanisms. Moreover, while the mechanisms regulating RAN translation are poorly understood, and the means by which GGGGCCexp transcripts make their way to the cytoplasm are not yet known, targeting these events may also provide an approach to block RAN translation.

Through base pairing to complementary nucleotide sequences, ASOs modulate the function of the targeted RNA via steric block or cleavage mechanisms. Steric blocking ASOs can modify pre-mRNA splicing by masking splicing enhancer or repressor sequences, inhibiting translation, or blocking the interaction of target RNA with RNA-binding proteins. For example, a steric blocking ASO, termed CAG25, was shown to disrupt the interaction between CUGexp RNA and MBNL1 following intramuscular injection in a mouse model of DM1 Citation[18]. Following release of MBNL1 from sequestration, splicing defects and defective chloride channel conductance were reversed Citation[18]. Of note, while CAG25 was not designed to induce target degradation, levels of CUGexp RNA were nonetheless decreased. This was postulated to result from its accelerated clearance given that, despite the overall decrease in CUGexp RNA levels, the amount of this transcript was increased in the cytoplasm where it would be accessible to degradation systems. Small molecules have likewise shown promise in improving DM1-associated features, including disrupting nuclear foci formation, blocking the CUGexp RNA-MBNL1 interaction, and attenuating translational and pre-mRNA splicing defects Citation[19,20]. Given their permeability and tight binding affinities, small ligands offer an attractive therapeutic option.

In contrast to steric blocking ASOs, RNAse H-dependent ASOs and siRNAs are designed to eliminate the toxic RNA. A robust decrease in CUGexp RNA was observed in skeletal muscle of DM1 transgenic mice after subcutaneous injection or injection into the tibialis anterior muscle of RNAse H-dependent ASOs, and this was accompanied by a decrease in nuclear RNA foci Citation[21,22]. Also observed were release of MBNL1, correction of myotonia, and rescue of misregulated splicing with sustained effects for up to 1 year after treatment was discontinued Citation[21]. Similarly, delivery of siRNAs targeting CUG repeats to skeletal muscle in DM1 transgenic mice led to a robust downregulation of CUGexp transcripts, dispersal of RNA foci, and restoration of MBNL1 splicing regulation Citation[23].

Expert opinion

The above-mentioned studies provide compelling evidence that targeting pathogenic RNA is a promising therapeutic strategy for the treatment of diseases in which RNA toxicity plays a role. Nonetheless, these strategies are still being refined and barriers exist that must be overcome if such approaches are to be utilized for treating neurodegenerative diseases. Existing challenges include improving the delivery of therapeutics to the central nervous system, as well as gaining a better understanding of potential off-target effects (e.g., effects resulting from the binding of the therapeutic to alternative transcripts that contain the repeat sequence being targeted). There are also several issues specific to c9FTD/ALS drug development that must be resolved. For instance, the secondary structure of GGGGCCexp must be taken into account. Studies suggest that GGGGCC repeats can form G-quadruplexes, and secondary structure prediction modeling indicates that the repeats may also assume hairpins Citation[4,15,24]. A G-quadruplex structure may negatively influence the ability of ASOs to bind the transcript. Small molecules, however, may be used to target such structures. It should also be kept in mind that conclusive proof that RNA-mediated toxicity underlies neurodegeneration in c9FTD/ALS has yet to be shown. While it may be a correct assumption that such is the case, a better understanding of disease mechanisms is required for designing the most effective therapeutic strategies. For example, while the means by which GGGGCCexp transcripts make their way to the cytoplasm and are loaded onto ribosomes are not yet known, targeting these events could well be an efficient therapeutic approach to block RAN translation. Also to consider is the possibility that therapeutics that prevent the binding of GGGGCCexp transcripts with RNA-binding proteins but increase cytoplasmic GGGGCCexp RNA (akin to the effect of GAC25 on CUGexp) may lessen one toxic cascade but enhance another. Of course, this may be avoided altogether by choosing approaches that eliminate toxic RNA. Yet, even then, challenges may arise; while ASOs and siRNAs are expected to preferentially cause the degradation of RNA from the expanded allele, there is likely to be some level of degradation of mRNA from the wild-type allele. Given that several groups have shown that mRNA levels of at least one C9ORF72 variant are decreased in c9FTD/ALS Citation[2,3,25], a further decrease caused by a purportedly protective intervention could contribute to a loss of C9ORF72 function. The effects of RNA-targeting therapies on the wild-type allele should therefore be carefully monitored. In addition, the potential contribution of C9ORF72 loss-of-function as a neurotoxic mechanism in c9FTD/ALS should be explored, and the functions of this almost completely uncharacterized protein should be determined. It is also important to note that the co-occurrence of nuclear RNA foci, expression of RAN translated peptides, and accumulation of TDP-43 inclusions could suggest the existence of a complex pathway in which all are interconnected, or the involvement of parallel pathological processes. If the latter is true, targeting only aberrant RNA species would not alleviate TDP-43-mediated toxicity. As such, combinatorial therapeutic strategies may be needed for c9FTD/ALS. Finally, at present, a major limiting factor in c9FTD/ALS drug discovery efforts is the lack of robust experimental models that recapitulate key features of disease. Such models are necessary to gain a better understanding of disease mechanisms and will be crucial for testing efficacy of possible therapeutics. Conversely, the development of small molecules, ASOs and the like that target GGGGCCexp RNA may not only lead to the development of potential therapies, but would also serve as valuable tools to decipher the many questions still unanswered.

Declaration of interest

This work was supported by the National Institutes of Health/National Institute on Aging [R01 AG026251 (LP)]; National Institutes of Health/National Institute of Neurological Disorders and Stroke [R21 NS074121-01 (TFG), R01 NS063964 (LP); R01 NS077402 (LP); R21 NS084528-01 (LP)]; National Institute of Environmental Health Services [R01 ES20395 (LP)]; and Amyotrophic Lateral Sclerosis Association (LP).

Bibliography

  • Belzil VV, Gendron TF, Petrucelli L. RNA-mediated toxicity in neurodegenerative disease. Mol Cell Neurosci 2012; In press
  • DeJesus-Hernandez M, Mackenzie IR, Boeve BF, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 2011;72:245-56
  • Renton AE, Majounie E, Waite A, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 2011;72:257-68
  • Ash PE, Bieniek KF, Gendron TF, et al. Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron 2013;77:639-46
  • Mori K, Weng SM, Arzberger T, et al. The C9orf72 GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in FTLD/ALS. Science 2013;339:1335-8
  • Sobczak K, de Mezer M, Michlewski G, et al. RNA structure of trinucleotide repeats associated with human neurological diseases. Nucleic Acids Res 2003;31:5469-82
  • Taneja KL, McCurrach M, Schalling M, et al. Foci of trinucleotide repeat transcripts in nuclei of myotonic dystrophy cells and tissues. J Cell Biol 1995;128:995-1002
  • Mooers BH, Logue JS, Berglund JA. The structural basis of myotonic dystrophy from the crystal structure of CUG repeats. Proc Natl Acad Sci USA 2005;102:16626-31
  • Mankodi A, Takahashi MP, Jiang H, et al. Expanded CUG repeats trigger aberrant splicing of ClC-1 chloride channel pre-mRNA and hyperexcitability of skeletal muscle in myotonic dystrophy. Mol Cell 2002;10:35-44
  • Dansithong W, Paul S, Comai L, et al. MBNL1 is the primary determinant of focus formation and aberrant insulin receptor splicing in DM1. J Biol Chem 2005;280:5773-80
  • Zu T, Gibbens B, Doty NS, et al. Non-ATG-initiated translation directed by microsatellite expansions. Proc Natl Acad Sci USA 2011;108:260-5
  • Jackson RJ, Hellen CU, Pestova TV. The mechanism of eukaryotic translation initiation and principles of its regulation. Nat Rev Mol Cell Biol 2010;11:113-27
  • Todd PK, Oh SY, Krans A, et al. CGG Repeat-Associated Translation Mediates Neurodegeneration in Fragile X Tremor Ataxia Syndrome. Neuron 2013;78:440-55
  • Xu Z, Poidevin M, Li X, et al. Expanded GGGGCC repeat RNA associated with amyotrophic lateral sclerosis and frontotemporal dementia causes neurodegeneration. Proc Natl Acad Sci USA 2013;110:7778-83
  • Reddy K, Zamiri B, Stanley SY, et al. 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 2013;288:9860-6
  • Mori K, Lammich S, Mackenzie IR, et al. hnRNP A3 binds to GGGGCC repeats and is a constituent of p62-positive/TDP43-negative inclusions in the hippocampus of patients with C9orf72 mutations. Acta Neuropathol 2013;125:413-23
  • Kim HJ, Kim NC, Wang YD, et al. Mutations in prion-like domains in hnRNPA2B1 and hnRNPA1 cause multisystem proteinopathy and ALS. Nature 2013;495:467-73
  • Wheeler TM, Sobczak K, Lueck JD, et al. Reversal of RNA dominance by displacement of protein sequestered on triplet repeat RNA. Science 2009;325:336-9
  • Ofori LO, Hoskins J, Nakamori M, et al. From dynamic combinatorial 'hit' to lead: in vitro and in vivo activity of compounds targeting the pathogenic RNAs that cause myotonic dystrophy. Nucleic Acids Res 2012;40:6380-90
  • Childs-Disney JL, Hoskins J, Rzuczek SG, et al. Rationally designed small molecules targeting the RNA that causes myotonic dystrophy type 1 are potently bioactive. ACS Chem Biol 2012;7:856-62
  • Wheeler TM, Leger AJ, Pandey SK, et al. Targeting nuclear RNA for in vivo correction of myotonic dystrophy. Nature 2012;488:111-15
  • Lee JE, Bennett CF, Cooper TA. RNase H-mediated degradation of toxic RNA in myotonic dystrophy type 1. Proc Natl Acad Sci USA 2012;109:4221-6
  • Sobczak K, Wheeler TM, Wang W, et al. RNA interference targeting CUG repeats in a mouse model of myotonic dystrophy. Mol Ther 2013;21:380-7
  • Fratta P, Mizielinska S, Nicoll AJ, et al. C9orf72 hexanucleotide repeat associated with amyotrophic lateral sclerosis and frontotemporal dementia forms RNA G-quadruplexes. Sci Rep 2012;2:1016
  • Gijselinck I, Van Langenhove T, van der Zee J, et al. A C9orf72 promoter repeat expansion in a Flanders-Belgian cohort with disorders of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis spectrum: a gene identification study. Lancet Neurol 2012;11:54-65

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.