Abstract
Lafora disease is a fatal progressive myoclonus epilepsy. At root, it is due to constant acquisition of branches that are too long in a subgroup of glycogen molecules, leading them to precipitate and accumulate into Lafora bodies, which drive a neuroinflammatory response and neurodegeneration. As a potential therapy, we aimed to downregulate glycogen synthase, the enzyme responsible for glycogen branch elongation, in mouse models of the disease. We synthesized an antisense oligonucleotide (Gys1-ASO) that targets the mRNA of the brain-expressed glycogen synthase 1 gene (Gys1). We administered Gys1-ASO by intracerebroventricular injection and analysed the pathological hallmarks of Lafora disease, namely glycogen accumulation, Lafora body formation, and neuroinflammation. Gys1-ASO prevented Lafora body formation in young mice that had not yet formed them. In older mice that already exhibited Lafora bodies, Gys1-ASO inhibited further accumulation, markedly preventing large Lafora bodies characteristic of advanced disease. Inhibition of Lafora body formation was associated with prevention of astrogliosis and strong trends towards correction of dysregulated expression of disease immune and neuroinflammatory markers. Lafora disease manifests gradually in previously healthy teenagers. Our work provides proof of principle that an antisense oligonucleotide targeting the GYS1 mRNA could prevent, and halt progression of, this catastrophic epilepsy.
Keywords: Lafora disease, antisense oligonucleotides, neuroinflammation, glycogen synthase, therapy
Ahonen et al. show that an antisense oligonucleotide targeting the brain-expressed glycogen synthase 1 gene corrects the neuropathological bases of Lafora disease in mouse models. The work provides proof of principle that an antisense oligonucleotide could halt progression of this catastrophic neurodegenerative epilepsy.
Introduction
Lafora disease is a teenage-onset progressive myoclonus epilepsy due to a disturbance in glycogen metabolism. In healthly subjects, two enzymes, glycogen synthase (which elongates glycogen chains) and glycogen branching enzyme, act in concert to form glycogen molecules that have chains of a certain length and are soluble. In Lafora disease, there is a slow but continuous generation of glycogen molecules with abnormally long chains.1,2 Lafora disease is caused by loss-of-function mutations in the genes encoding the glycogen phosphatase laforin (EPM2A) or the laforin-interacting ubiquitin E3 ligase malin (EPM2B/NHLRC1).2 The mechanisms by which laforin and malin regulate glycogen chain lengths remain unclear, but it is apparent that the overlong glycogen chains cause affected molecules to precipitate and over time accumulate into disease-pathognomonic Lafora bodies. In the brain, by teenage years the ever increasing and enlarging Lafora bodies drive a progressive neuroinflammatory process, which at least in part underlies the disease’s intractable epilepsy, neurodegeneration and dementia.2-4
The Epm2a−/− and Epm2b−/− Lafora disease mouse models recapitulate the primary pathologies of the disease, namely presence of glycogen molecules with overlong branches, progressive Lafora body formation, and neuroinflammation. These abnormalities were prevented by breeding Lafora disease mice with mice deficient of glycogen synthase or its activator proteins, and thus limiting the ability of the enzyme to elongate glycogen chains. This effect was achieved both by complete or partial (30–50%) reduction of glycogen synthase activity.5–11 Additionally, conditional knockdown of glycogen synthase after the disease had progressed for a period of time, i.e. after Lafora bodies had already accumulated, halted further Lafora body formation and attenuated neuroinflammation.12,13
In the present work we designed an antisense oligonucleotide (ASO) that targets the mRNA of the brain-expressed isoform of glycogen synthase (Gys1). Delivered into the CSF of Lafora disease mouse models, the ASO halted Lafora body formation and reduced neuroinflammation. The results open a path to an ASO-based therapy for Lafora disease.
Materials and methods
Mice and delivery of antisense oligonucleotides
Epm2a −/−and Epm2b−/−mouse models were described previously.5,6 Animal procedures were approved by the Toronto Centre for Phenogenomics or Ionis Pharmaceuticals Institutional Animal Care and Use Committees. Male and female mice were anaesthetized with isofluorane, and, except where indicated otherwise, 300 µg ASO in 10 µl PBS were injected intracerebroventrically as per indicated schedule, alternating ventricles at successive time points. Stereotactic injection coordinates were 0.3 mm anterior/posterior (anterior to bregma), 1.0 mm to right or left medial/lateral and −3.0 mm dorsal/ventral. Analgesic (Metacam® 2 mg/kg) was administered before and for 2 days post-injections. The Gys1-ASO sequence was 5′-CATGCTTCATTTCTTTATTG-3′. Littermate controls were a no-target ASO, 5′-CCTATAGGACTATCCAGGAA-3′ (Ctrl-ASO), or PBS. Mice were sacrificed by cervical dislocation. One hemisphere was snap-frozen in liquid nitrogen for qRT-PCR and biochemical analyses, the other immersed in 10% neutral buffered formalin for histo- and immunohisto-pathology.
Gys1 expression analysis in mouse brain samples
For most trials, RNA was extracted with the Qiagen RNeasy® Lipid Tissue Mini Kit (Qiagen, #74804) and using a 1 ml syringe and 21 gauge needle for tissue homogenization. DNA was digested with DNase I (Thermo Scientific, #EN0521). Complementary DNA was synthesized from 1 μg RNA using the iScriptTM Advanced cDNA Synthesis Kit (Bio-Rad, #1725037). For the 3–12 month trial, tissue was homogenized in Ambion TRIzolTM (Thermo Fisher Scientific, #15596018). Homogenate was loaded onto Phase Lock Gels (VWR, #10847–802). After centrifugation the aqueous phase was loaded onto RNA columns (PureLink™ RNA Mini Kit, Thermo Fisher Scientific, #12183025). On-column DNaseI digestion was performed using the PureLink™ DNase Set (Thermo Fisher Scientific, #12185010). Complementary DNA was synthesized using the iScript™ Reverse Transcription Supermix kit (Bio-Rad, #1708841).
For qRT-PCR we used either the Stratagene Mx3005P (Agilent Technologies) or QuantStudio 7 Pro SmartStart (Applied Biosystems) real-time PCR system, and SYBR Green technology [PowerUpTM SYBRTM Green Master Mix (Applied Biosystems) or iTaqTM Universal SYBR® Green Supermix (Bio-Rad)]. Primer sequences were Gys1-F: 5′-CGCAAACAACTATGGGACAC-3′, Gys1-R: 5′-TCCTCCTTGTCCAGCATCTT-3′, Gapdh-F: 5′-AAGGGCTCATGACCACAGTC-3′, Gapdh-R: 5′-GGATGCAGGGATGATGTTCT-3′, Hprt-F: 5′-TTGCTGACCTGCTGGATTAC-3′, Hprt-R: 5′-ACTTTTATGTCCCCCGTTGA-3′, Rpl4-F: 5′-CCCTTACGCCAAGACTATGC-3′, Rpl4-R: 5′-TGGAACAACCTTCTCGGATT-3′, Lcn2-F: 5′-GCCTCAAGGACGACAACATC-3′, Lcn2-R: 5′-CACACTCACCACCCATTCAG-3′, Cxcl10-F: 5′-AAGTGCTGCCGTCATTTTCT-3′, Cxcl10-R: 5′-ATAGGCTCGCAGGGATGATT-3′, Ccl5-F: 5′-TGCCAACCCAGAGAAGAAGT-3′, Ccl5-R: 5′-AGCAAGCAATGACAGGGAAG-3′, C3-F: 5′-CTGTGTGGGTGGATGTGAAG-3′, and C3-R: 5′-TCCTGAGTGTCGTTTGTTGC-3′. Gapdh served as reference, except for the 3–12 month trial where Hprt and Rpl4 were used. ΔCt values were determined, calculating Ctgene of interest − Ctreference gene (geometric mean was used in case of two reference genes), followed by transformation into 2−ΔCt. Expression levels were further normalized to the PBS or baseline group.
Protein and glycogen analyses
Frozen tissue was homogenized with buffer including Pierce protease and phosphatase inhibitors (Thermo Scientific) and 2 mM DTT. Protein concentrations were determined using the DC Protein Assay (Bio-Rad). Equal protein amounts (30 µg) were heated in sample buffer (70°C, 10 min) and loaded to 10% SDS-PAGE for western blotting. GYS1 and GAPDH antibodies were from Cell Signaling (#3886) and Santa Cruz (sc-365062), respectively. Immunoblots were detected with HRP-conjugated secondary antibodies and Clarity Western ECL-substrate (Bio-Rad) and analysed with the ChemiDoc Imaging system (Bio-Rad). Glycogen extraction, amyloglucosidase digestion and glucose determination were as previously described.14
Histological analyses
Formalin-fixed paraffin-embedded brain tissues were sectioned and stained using periodic acid-Schiff diastase (PASD) for Lafora bodies9 or immunohistochemistry against GFAP (mouse anti-GFAP, BioGenex, #AM020-5M; dilution 1:250). Slides were scanned using Pannoramic (3DHistech) or NanoZoomer 2.0-HT (Hamamatsu Photonics) scanners. Lafora bodies and GFAP signals were quantified in the hippocampus using HistoQuant (3DHistech) by defining Lafora bodies or GFAP signals based on pixel colour. Values are expressed as per cent area.
Lafora body size distribution
Per hippocampal section, HistoQuant analysis yielded in a list of individual Lafora body sizes. Lafora bodies were assigned to 30 bins with size limits defined through third order binomial functions (Equations 1 and 2), b being the bin position.
(1) |
(2) |
The number of bodies per bin was divided by hippocampal area and expressed as area-normalized Lafora body number (count/µm2). The average area-normalized body number per bin was calculated across all hippocampi of each group (n > 7, biological replicates) and plotted against the bin centre (average of lower and upper bin size limits). To calculate fold-change from baseline, area-normalized counts of each bin were divided by the average area-normalized count of the baseline group in the same bin (also performed with individual baseline values to obtain fold-changes to baseline average for each animal). Average fold-change and standard error of the mean (SEM) were plotted against body size (bin centre). Statistical analyses were performed comparing group average area-normalized body numbers and fold-changes for each bin.
Statistical analyses
Data are presented as mean ± SEM. Statistics were performed with GraphPad Prism 8.4.3. If not stated otherwise, one-way ANOVA was performed, followed by post hoc tests with Tukey multiple comparison correction. For Figs 3A, B and 4E, two-way ANOVA was performed, also followed by post hoc tests with Tukey multiple comparison correction. For Fig. 3D and E, one-way ANOVA was performed, followed by post hoc tests with Holm-Bonferroni multiple comparison correction. For qRT-PCR data in Figs 2 and 4, Kruskal-Wallis ANOVA was used, followed by two-stage linear step-up procedure of Benjamini, Krieger and Yekutieli. Asterisks denote statistical significance as follows: *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001.
Figure 3.
Long-term ASO treatment strongly prevents glycogen and Lafora body accumulation in Epm2a−/− mice. (A) Brain total glycogen content. (B) Lafora body (LB) quantification in the hippocampus. (C) Representative images of PASD stained hippocampus. Scale bar = 50 µm. PBS, Ctrl-ASO (no-target control ASO), or Gys1-ASO were injected at 3, 6 and 9 months and brain tissue analysed at 12 months. Untreated mice, sacrificed and analysed at 3 months, served as a baseline control. Significance levels are indicated as: *P < 0.05, ***P < 0.001 and ****P < 0.0001. Asterisks in pink indicate significance levels compared with the corresponding wild-type (WT). (D) Lafora body size distribution in Epm2a−/− mice. (E) Differential Lafora body size distribution (fold change compared to baseline) in Epm2a−/− mice. (D and E) Top: P-values comparing Lafora body number between indicated experimental groups at different Lafora body size bins. All data are presented as mean ± SEM.
Figure 4.
Long-term ASO treatment rescues astrogliosis in Epm2a−/− mice. (A–E) Relative expression levels of Gys1 mRNA (A), and inflammatory and immune system response marker genes Lcn2 (B), Cxcl10 (C), Ccl5 (D), and C3 (E) analysed by qRT-PCR. (F) GFAP signal quantification in the hippocampus. (G) Representative immunohistochemistry (IHC) images of anti-GFAP in the hippocampus. Scale bar = 50 µm. PBS, Ctrl-ASO (no-target control ASO), or Gys1-ASO were injected at 3, 6 and 9 months and brain tissue analysed at 12 months. Untreated mice, sacrificed and analysed at 3 months, served as a baseline control. All data are presented as mean ± SEM. Significance levels are indicated as: *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001. Asterisks in pink indicate significance levels compared with the corresponding wild-type (WT).
Figure 2.
Later ASO administration, after Lafora disease onset, effectively slows disease progression in Epm2a−/− mice without signs of reversal. (A) Experimental design of three different trials. PBS, Ctrl-ASO (no-target control ASO), or Gys1-ASO were injected and mice sacrificed at indicated time points. Untreated mice, sacrificed at time of first injection, served as baseline control. (B–E) Results from the 3–6 month trial (trial design in A), showing Gys1 mRNA levels in Epm2a−/− (B), brain total glycogen content in Epm2a−/− (C), and wild-type (WT) (D), and Lafora body (LB) quantification in the hippocampus of Epm2a−/− mice (E). (F–H) Results from the 8–14 month trial, using 300 µg ASO for each injection (trial design in A), showing Gys1 mRNA levels (F), brain total glycogen content (G) and Lafora body quantification in the hippocampus (H) of Epm2a−/− mice. (I and J) Results the from 8–14 month trial, using a higher dose of 500 µg ASO for each injection (trial design in A), showing brain total glycogen content (I) and Lafora body quantification in the hippocampus (J) of Epm2a−/− mice. (K) Representative images of PASD stained hippocampus of Epm2a−/− mice from the three different trials, explained in A. Scale bar = 50 µm. All data are presented as mean ± SEM. Significance levels are indicated as: *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001.
Data availability
The raw data that support the findings of this study are available from the corresponding author, upon request.
Results
Identification of an active antisense oligonucleotide
We designed 300 ASOs targeting different regions of Gys1 pre-mRNA, which we screened for efficacy in mRNA downregulation. We selected Gys1-ASO for its potent (75%), yet not complete, Gys1 mRNA downregulation (Supplementary Fig. 1A). The half maximal inhibitory concentration (IC50) of Gys1-ASO in cell-based experiments was 3.4 µM (Supplementary Fig. 1A and B). We administered Gys1-ASO into ventricles of 1-month-old wild-type mice, sacrificed the mice 14 days later and measured Gys1 mRNA, which had diminished by >50% in the cortex, hippocampus, and spinal cord (Supplementary Fig. 1C–E).
Gys1-ASO prevents Lafora body formation in young mice
In the Lafora disease mouse models, Lafora bodies begin to form around 1 month of age and become increasingly visible over the following 2 months, especially in the hippocampus. We delivered Gys1-ASO by intracerebroventricular injection to Epm2a−/− mice at 1 and 2 months and sacrificed the mice at 3 months of age. Both mRNA and protein levels of Gys1 were reduced by >80% in the Gys1-ASO-treated group (Fig. 1A–C). Brain glycogen levels are elevated in Lafora disease mice due to the accumulation of malstructured glycogen in the form of Lafora bodies.14 In Gys1-ASO-treated mice, brain glycogen levels were >50% lower than in control mice (Fig. 1D) and comparable to levels normally seen in wild-type mice (∼2 µmol glucose/g tissue).14 Finally, PASD staining revealed that Lafora bodies were likewise and equivalently reduced (Fig. 1E and F). Results of these experiments in Epm2b−/− mice were similar (Supplementary Fig. 2).
Figure 1.
Gys1-ASO, administered at 1 and 2 months, leads to reduced Gys1 mRNA and GYS1 protein levels and attenuates glycogen and Lafora body accumulation in Epm2a−/− mice at 3 months. (A) Brain Gys1 mRNA relative expression levels in PBS-, Ctrl-ASO-, and Gys1-ASO-injected Epm2a−/− mice. Ctrl-ASO, a no-target control ASO. (B) Brain GYS1 western blots with GAPDH as loading control. (C) Quantification of GYS1 western blots shown in B, normalized to GAPDH. (D) Brain total glycogen content. (E) Lafora body (LB) quantification in the hippocampus. (F) Representative images of PASD stained hippocampus. Scale bar = 50 µm. All data are presented as mean ± SEM. Significance levels are indicated as: *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001.
The above experiments showed that when administered at the point in time when Lafora bodies are starting to appear, Gys1-ASO inhibits Lafora body accumulation. Results being similar using both mouse models, we limited subsequent experiments to the Epm2a−/− genotype.
Gys1-ASO prevents Lafora body formation in older mice
To determine whether the oligonucleotide is active in the context of pre-existing Lafora body accumulations, we administered Gys1-ASO to older mice (3 months and 8 months old) with different specified dosing frequencies, doses and ages at sacrifice (Figs 2 and 3A–C). These experiments were also designed in such a way as to inform us whether treatment with Gys1-ASO could reverse existing Lafora body accumulations, i.e. not only prevent new accumulations, but lead to removal of existing ones. The results can be summarized as follows. Gys1-ASO does prevent Lafora body accumulations beyond amounts present at time of treatment initiation. This effect is stronger when treatment starts earlier (3 months versus 8 months), is longer (3 months to 12 months versus 3 months to 6 months), or is dosed higher (500 µg versus 300 µg) (Figs 2 and 3A–C). Lafora bodies do not diminish below their existing levels.
Gys1-ASO prevents a shift towards larger Lafora bodies
Lafora bodies occur along a spectrum between two main morphologies, small and dust-like, and large and spherical. The first are much smaller and much more numerous and are located in countless astrocytic processes. The second are mostly juxtanuclear in neuronal perikarya, occupying varying extents of neuronal cytoplasms.15,16 We studied the size distribution of hippocampal Lafora bodies in one of our experiments (3–12 month trial) and show that across all sizes Lafora body abundance increases with Lafora disease progression (PBS/Ctrl-ASO versus baseline), with larger Lafora bodies (>1 µm2) contributing more to the overall increase of Lafora bodies (Fig. 3D and E). Gys1-ASO attenuated the growth of Lafora bodies of all sizes, in particular maintaining the abundance of larger bodies (1–60 µm2) low compared with controls where large bodies markedly increased in number (Fig. 3E).
Gys1-ASO prevents Lafora disease-related astrogliosis
A remarkable 94% of genes upregulated in Lafora disease mouse model brain transcriptomes are those involved in inflammatory and immune system pathways, strongly suggesting that immune disease at least in part underlies the disease. Of the hundreds of these genes a set of nine has been validated by qRT-PCR experiments, showing gradual increase in expression with Lafora disease progression.4 We measured expressions of four of these genes (Lcn2, Cxcl10, Ccl5 and C3) (as well as that of Gys1) in the 3–12 month study and show that Gys1-ASO imparts a strong corrective tendency on the expression levels of all (Fig. 4A–E). We next studied whether microgliosis and astrogliosis, previously reported in murine Lafora disease, are improved. Aside from Lafora bodies, astrogliosis is the earliest and most constant neuropathological abnormality in LD mouse models.7–10,17 Microgliosis was not affected (Supplementary Fig. 3), but astrogliosis was corrected and near-eliminated (Fig. 4E–F).
Discussion
Lafora disease afflicts previously healthy children with escalating and protracted devastation. While the basic mechanisms of disease are not fully elucidated, the principal cog has recently become known: abnormally long branches, generated by glycogen synthase, lead to glycogen insolubility.3,18 As such, downregulating glycogen synthase may be therapeutic to the disease, which forms the basis of the present study.
ASOs have exceptional target specificities through Watson-Crick nucleotide sequence matching. Delivered to the CSF they distribute to practically all cells of all brain regions. They are stable and active for several months between doses.19 They can act through multiple mechanisms, e.g. activating a silent homologue (SMN2) of a mutated gene (SMN1) in spinal muscular atrophy,20,21 stabilizing and augmenting the activity of the healthy allele (SCN1A) in Dravet syndrome,22 and others. However, their original and simplest mechanism is downregulation of a target mRNA through RNaseH1,23 which is what is required in Lafora disease with GYS1 as target. We here show in the mouse models of Lafora disease that an ASO targeting Gys1 mRNA prevents the pathogenic Lafora bodies from forming or proliferating, and improves and corrects immunopathological features of the disease.
Mammals possess two glycogen synthase genes, one expressed exclusively in the liver (Gys2), the other (Gys1) in all the other organs including brain.24 Patients completely lacking GYS1 (glycogen storage disease type 0 b) have no neurological disease but develop cardiac arrhythmias. Parents of these patients, with 50% GYS1 activity, are completely healthy.25–27 A proposed therapy for Lafora disease with a GYS1 targeting ASO would be administered by lumbar puncture and thus not affect cardiac GYS1, as ASOs do not cross the blood–brain or arachnoid granulation barriers.28–30 In the brain, partial downregulation of GYS1 would be aimed for and sufficient. In previous studies based on genetic crosses of Lafora disease mice with mice deficient of GYS1 or proteins that activate GYS1, we and others showed that 30–50% GYS1 downregulation suffices to prevent Lafora body formation.9,10
In the present work the ASO was effective at any disease stage, but more so in early disease and with sustained therapy. In the current era, patients with Lafora disease can be diagnosed within weeks of symptoms. Myoclonic, visual or convulsive seizures lead to an EEG, which beyond epileptiform discharges shows background dysregulation and raises suspicion of a non-benign epilepsy. Since EPM2A and EPM2B are now widely included in epilepsy gene panels, diagnosis should then be swiftly made. In early stages of disease, patients with Lafora disease are no different from other teenagers with new-onset epilepsy. As such, a therapy that halts the disease would be, in principle, tantamount to a cure.
Precisely how the laforin glycogen phosphatase and malin ubiquitin E3 ligase cooperate to fine-tune the lengths of glycogen branches to generate perfectly spherical and soluble macromolecules awaits to be uncovered. Meanwhile it is hoped that the present results will rapidly translate to a therapy for Lafora disease.
Supplementary Material
Acknowledgements
We would like to thank Jennifer P. Lee and Julia Gliwa for expert technical support.
Funding
This work was funded by the National Institutes of Health under award P01NS097197. S.A. was supported by the Sigrid Jusélius Foundation. B.A.M. holds the University of Texas Southwestern Jimmy Elizabeth Westcott Chair in Pediatric Neurology.
Competing interests
H.K. is a shareholder and employee at Ionis pharmaceuticals. T.R.G. has a patent (16/306831) pending. All other authors report no competing interests.
Supplementary material
Supplementary material is available at Brain online.
Glossary
- ASO
antisense oligonucleotide
References
- 1. Roach PJ. Glycogen and its metabolism. Curr Mol Med. 2002;2(2):101–120. [DOI] [PubMed] [Google Scholar]
- 2. Nitschke F, Ahonen SJ, Nitschke S, Mitra S, Minassian BA.. Lafora disease - from pathogenesis to treatment strategies. Nat Rev Neurol. 2018;14(10):606–617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Sullivan MA, Nitschke S, Skwara EP, et al. Skeletal muscle glycogen chain length correlates with insolubility in mouse models of polyglucosan-associated neurodegenerative diseases. Cell Rep. 2019;27(5):1334–1344.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Lahuerta M, Gonzalez D, Aguado C, et al. Reactive glia-derived neuroinflammation: A novel hallmark in Lafora progressive myoclonus epilepsy that progresses with age. Mol Neurobiol. 2020;57(3):1607–1621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Ganesh S, Delgado-Escueta AV, Sakamoto T, et al. Targeted disruption of the Epm2a gene causes formation of Lafora inclusion bodies, neurodegeneration, ataxia, myoclonus epilepsy and impaired behavioral response in mice. Hum Mol Genet. 2002;11(11):1251–1262. [DOI] [PubMed] [Google Scholar]
- 6. Turnbull J, Wang P, Girard JM, et al. Glycogen hyperphosphorylation underlies Lafora body formation. Ann Neurol. 2010;68(6):925–933. [DOI] [PubMed] [Google Scholar]
- 7. Pederson BA, Turnbull J, Epp JR, et al. Inhibiting glycogen synthesis prevents Lafora disease in a mouse model. Ann Neurol. 2013;74(2):297–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Turnbull J, Epp JR, Goldsmith D, et al. PTG protein depletion rescues Malin-deficient Lafora disease in mouse. Ann Neurol. 2014;75(3):442–446. [DOI] [PubMed] [Google Scholar]
- 9. Turnbull J, DePaoli-Roach AA, Zhao X, et al. PTG depletion removes Lafora bodies and rescues the fatal epilepsy of Lafora disease. PLoS Genet. 2011;7(4):e1002037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Duran J, Gruart A, Garcia-Rocha M, Delgado-Garcia JM, Guinovart JJ.. Glycogen accumulation underlies neurodegeneration and autophagy impairment in Lafora disease. Hum Mol Genet. 2014;23(12):3147–3156. [DOI] [PubMed] [Google Scholar]
- 11. Israelian L, Nitschke S, Wang P, et al. Ppp1r3d deficiency preferentially inhibits neuronal and cardiac Lafora body formation in a mouse model of the fatal epilepsy Lafora disease. J Neurochem. 2021;157(6):1897–1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Varea O, Duran J, Aguilera M, Prats N, Guinovart JJ.. Suppression of glycogen synthesis as a treatment for Lafora disease: Establishing the window of opportunity. Neurobiol Dis. 2021;147:105173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Nitschke S, Chown EE, Zhao X, et al. An inducible glycogen synthase-1 knockout halts but does not reverse Lafora disease progression in mice. J Biol Chem. 2021;296:100150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Nitschke F, Sullivan MA, Wang P, et al. Abnormal glycogen chain length pattern, not hyperphosphorylation, is critical in Lafora disease. EMBO Mol Med. 2017;9(7):906–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Augé E, Pelegrí C, Manich G, et al. Astrocytes and neurons produce distinct types of polyglucosan bodies in Lafora disease. GLIA. 2018;66(10):2094–2107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Rubio-Villena C, Viana R, Bonet J, et al. Astrocytes: New players in progressive myoclonus epilepsy of Lafora type. Hum Mol Genet. 2018;27(7):1290–1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. López-González I, Viana R, Sanz P, Ferrer I.. Inflammation in Lafora disease: Evolution with disease progression in Laforin and Malin knock-out mouse models. Mol Neurobiol. 2017;54(5):3119–3130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Sullivan M, Nitschke S, Steup M, Minassian B, Nitschke F.. Pathogenesis of Lafora disease: Transition of soluble glycogen to insoluble polyglucosan. Int J Mol Sci. 2017;18(8):1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Geary RS, Norris D, Yu R, Bennett CF.. Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides. Adv Drug Deliv Rev. 2015;87:46–51. [DOI] [PubMed] [Google Scholar]
- 20. Finkel RS, Mercuri E, Darras BT, et al. Nusinersen versus sham control in infantile-onset spinal muscular atrophy. N Engl J Med. 2017;377(18):1723–1732. [DOI] [PubMed] [Google Scholar]
- 21. Mercuri E, Darras BT, Chiriboga CA, et al. Nusinersen versus sham control in later-onset spinal muscular atrophy. N Engl J Med. 2018;378(7):625–635. [DOI] [PubMed] [Google Scholar]
- 22. Han Z, Chen C, Christiansen A, et al. Antisense oligonucleotides increase Scn1a expression and reduce seizures and SUDEP incidence in a mouse model of Dravet syndrome. Sci Transl Med. 2020;12(558):eaaz6100. [DOI] [PubMed] [Google Scholar]
- 23. Crooke ST. Molecular mechanisms of antisense oligonucleotides. Nucleic Acid Ther. 2017;27(2):70–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Roach PJ, Depaoli-Roach AA, Hurley TD, Tagliabracci VS.. Glycogen and its metabolism: Some new developments and old themes. Biochem J. 2012;441(3):763–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Cameron JM, Levandovskiy V, MacKay N, et al. Identification of a novel mutation in GYS1 (muscle-specific glycogen synthase) resulting in sudden cardiac death, that is diagnosable from skin fibroblasts. Mol Genet Metabol. 2009;98(4):378–382. [DOI] [PubMed] [Google Scholar]
- 26. Kollberg G, Tulinius M, Gilljam T, et al. Cardiomyopathy and exercise intolerance in muscle glycogen storage disease 0. New Engl J Med. 2007;357(15):1507–1514. [DOI] [PubMed] [Google Scholar]
- 27. Sukigara S, Liang WC, Komaki H, et al. Muscle glycogen storage disease 0 presenting recurrent syncope with weakness and myalgia. Neuromuscul Disord. 2012;22(2):162–165. [DOI] [PubMed] [Google Scholar]
- 28. Geary RS, Yu RZ, Levin AA.. Pharmacokinetics of phosphorothioate antisense oligodeoxynucleotides. Curr Opin Investig Drugs. 2001;2(4):562–573. [PubMed] [Google Scholar]
- 29. Smith RA, Miller TM, Yamanaka K, et al. Antisense oligonucleotide therapy for neurodegenerative disease. J Clin Invest. 2006;116(8):2290–2296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Rinaldi C, Wood MJA.. Antisense oligonucleotides: The next frontier for treatment of neurological disorders. Nat Rev Neurol. 2017;14(1):9–21. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The raw data that support the findings of this study are available from the corresponding author, upon request.