Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Apr 1.
Published in final edited form as: Rev Neurol (Paris). 2021 Jul 21;178(4):315–325. doi: 10.1016/j.neurol.2021.06.006

Lafora Disease: current biology and therapeutic approaches

Sharmistha Mitra 1, Emrah Gumusgoz 1, Berge A Minassian 1
PMCID: PMC8770683  NIHMSID: NIHMS1723608  PMID: 34301405

Abstract

The ubiquitin system impacts most cellular processes and is altered in numerous neurodegenerative diseases. However, little is known about its role in neurodegenerative diseases due to disturbances of glycogen metabolism such as Lafora disease (LD). In LD, insufficiently branched and long-chained glycogen forms and precipitates into insoluble polyglucosan bodies (Lafora bodies), which drive neuroinflammation, neurodegeneration and epilepsy. LD is caused by mutations in the gene encoding the glycogen phosphatase laforin or the gene coding for the laforin interacting partner ubiquitin E3 ligase malin. The role of the malin-laforin complex in regulating glycogen structure remains with full of gaps. In this review we bring together the disparate body of data on these two proteins and propose a mechanistic hypothesis of the disease in which malin-laforin’s role to monitor and prevent over-elongation of glycogen branch chains, which drive glycogen molecules to precipitate and accumulate into Lafora bodies. We also review proposed connections between Lafora bodies and the ensuing neuroinflammation, neurodegeneration and intractable epilepsy. Finally, we review the exciting activities in developing therapies for Lafora disease based on replacing the missing genes, slowing the enzyme - glycogen synthase - that over-elongates glycogen branches, and introducing enzymes that can digest Lafora bodies. Much more work is needed to fill the gaps in glycogen metabolism in which laforin and malin operate. However, knowledge appears already adequate to advance disease course altering therapies for this catastrophic fatal disease.

Keywords: E3 ubiquitin ligase, laforin, malin, Lafora Disease, gene therapy

Neurodegenerative diseases, ubiquitin biology and Lafora disease:

Abnormal inclusions in the central nervous system are hallmarks of various neurodegenerative disorders. These accumulations often perturb cellular physiology and homeostasis. Globally, the burden of neurological disorders continues to increase as populations are growing and ageing, demanding more research in the basic molecular mechanisms of neurodegenerative diseases associated with toxic inclusions [1].

Protein ubiquitination and related cellular pathways are frequently altered in neurodegenerative diseases [24]. Ubiquitin is a small protein (8 kD), which is covalently attached to other proteins via a process that requires three consecutive reactions. The first is activation of the ubiquitin molecule by a ubiquitin activating enzyme E1 through an ATP-dependent mechanism. This is followed by the transfer of the activated ubiquitin from E1, by a ubiquitin conjugating enzyme E2 to itself. Finally, a ubiquitin ligase enzyme, E3, covalently ligates the ubiquitin molecule to its protein substrate. Ubiquitination either targets a substrate to degradation by the ubiquitin proteasome system (UPS) or by autophagy, or serves to regulate the protein substrate in its cellular pathways, including immune signaling, endocytosis, DNA damage repair, cell cycle progression, mitophagy, autophagy and others [5, 6]. Because of this myriad functions of ubiquitin biology, it is not surprising that aberrations of ubiquitination have been implicated in multiple neurological disorders including but not limited to Alzheimer’s disease, Parkinson’s disease, Huntingtin’s disease and Angelman Syndrome [3, 7]. In a recent study, a manual database search found that among ~700 known and predicted human E3 ligases, 83 are mutated in different neurological disorders, highlighting the importance of this class of proteins in neurological disease [8].

In certain neurological disorders, the inclusions are not proteinaceous but glucan in nature, namely abnormal structured, insoluble forms of glycogen [9, 10]. In some, there is disturbance in ubiquitin biology [11, 12]. One such disease is Lafora Disease (LD), a teenage-onset intractable, fatal progressive myoclonus epilepsy [13]. In this article we review LD with a focus on ubiquitin biology. First, we briefly review clinical aspects of the disease. We next describe the unknowns about the E3 ubiquitin ligase gene mutated in the disease including its hypothetical role in glycogen metabolism and neurodegeneration. Finally, we discuss currently available and future treatment options for LD.

The clinical syndrome of Lafora disease

LD is inherited in autosomal recessive fashion. The majority of patients have a relatively uniform phenotype, with exceptional cases with missense mutations or certain splice site mutations having a later onset and protracted course. Typically, a period of decline in school performance in pre-teen years predates appearance between ages 10 and 15 years of overt symptoms, which are any or all of myoclonus, visual hallucinations or convulsive seizures [14]. Given the onset of myoclonus and seizures in teenage years, a tentative diagnosis of Juvenile Myoclonic Epilepsy (JME) is frequently made, until the EEG is obtained. The latter shows background slowing and generalized and occipital discharges of irregular conformations, indicating something more severe than JME, and leading to genetic testing [15]. All symptoms inexorably worsen in the subsequent years. Seizures become intractable to any and all combinations of anti-seizure drugs, and myoclonus becomes near constant. The myoclonus is associated with atypical absenses leading, in its constancy, the patient to become unable to complete thoughts or sentences. Dementia sets in, ambulation is lost, and eventually the patient becomes vegetative. Usually, the young patient succumbs to airway control problems and/or status epilepticus within ten years of onset [13, 14]. Thankfully, LD is rare, with a frequency not higher than 1/100,000 [16]. But as mentioned, it is a horrendous disease definitely needing better understanding and a treatment.

Malin is an E3 ubiquitin ligase deficient in approximately 50% of patients with LD:

In LD, long chained, lesser branched glycogen is formed and precipitated into insoluble polyglucosan bodies (PBs) (Lafora bodies; LBs, named after Gonzalo Lafora who first described them [17]), which likely drive the neurodegeneration and epilepsy (see below) [14, 18]. In the vast majority of patients the brain is the only clinically affected organ, even though LBs also form in other organs including skeletal muscle, heart, liver and skin [19, 20]. There have been exceedingly rare instances of cardiac arrhythmia and heart failure [21]. Typically, LD starts in otherwise healthy teenagers with myoclonus, visual and convulsive seizures, which over time that become frequent and intractable. This is associated with behavioral and cognitive decline; the patient ultimately reaches a vegetative state and dies within 10 years of onset [22]. The molecular mechanisms that underlie LD are not fully understood. However, recent research revealed unsuspected players in glycogen metabolism, namely the glycogen phosphatase laforin and its interacting protein partner, the ubiquitin E3 ligase malin.

Discovered in 2003 [23], the gene for malin, EPM2B (also known as NHLRC1), maps in chromosomal band 6p22.3 and consists of a single exon expressing an N-terminal RING (Really Interesting New Gene) domain and a C-terminal six repeats of NHL (protein domain present in Ncl-1, HT2A and Lin-41 protein) domains. The NHL domains presumably confer protein-protein interaction, and the RING domain the E3 ubiquitin ligase activity. There are over 80 LD causing mutations that have been identified in the EPM2B gene, affecting both RING and NHL domains (http://projects.tcag.ca/lafora/). The evolutionary lineage of the EPM2B gene is restricted to only vertebrate species and a branchiopod. This is in contrast to its interacting protein partner laforin, encoded by EPM2A gene, which has much broader appearance in both vertebrates, invertebrates, protozoa as well as in certain algae indicating possible malin-independent functions of laforin especially in lower class of organisms [24]. Approximately 50% of LD patients have mutations in the EPM2B gene, and the remainder in the EPM2A gene.

The malin-laforin complex may act in quality control of glycogen synthesis:

The role of malin in glycogen biology remains a knotty question, which we review starting with a focused summary of glycogen metabolism. Glycogen is the largest molecule in the cytoplasm and a critical energy store. Glucose chains 10 units or longer tend to precipitate, yet 55,000-unit glycogen is soluble, a requisite to its metabolism [25]. How this solubility is achieved was part of the foundational classical biochemical discoveries of the 20th century and until recently considered settled [26]. Glycogen branching enzyme (GBE) activity proportionate to that of glycogen synthase (GS) was thought enough to generate molecules with adequate branching (and therefore adequately short branches) to allow molecule-through hydration and thus solubility. However, in both the laforin and malin knockout mouse models, the amount of soluble GS, its activation state and activity levels remain unaltered, indicating that there are additional mechanisms in the structuring of normal glycogen beyond just balanced activities between GS and GBE at the whole-cell level [27]. One of the fascinating findings in this aspect was the discovery that laforin is a glycogen phosphatase, consistent with the observation that LD glycogen is hyperphosphorylated [2831]. However, subsequent studies showed that glycogen hyperphosphorylation, and therefore deficiency of laforin’s glycogen phosphatase function, are not at the root of abnormal glycogen (polyglucosan) formation in LD. Expression of a phosphatase-defective laforin in laforin knockout mice rescued the mice, including correcting the chain lengths of glycogen and preventing LB formation. This indicated that a laforin function other than its phosphatase is necessary to prevent LD. Expression of the same phosphatase-defective laforin in malin knockout mice did not rescue any of the above. Together these results indicated that laforin most likely acts via malin, and emphasized the critical role of malin’s ubiquitin E3 ligase activity in the regulation of glycogen structure [32, 33] [3436]. [33].

A recent study shed on the native properties of soluble glycogen added further detail. Analyses of glycogen fractions from both laforin knockout and malin knockout mouse models showed that i) soluble glycogen is heterogeneous in branch chain length distribution, ii) molecules with longer chains have a propensity to precipitate, and iii) the insoluble glycogen fractions from both mouse models are enriched with molecules with the longest chains [27]. These observations highlighted the fact that glycogen branch lengths are key determinant to solubility, and that certain soluble glycogen molecules are at the risk of precipitation due to possessing an excess of longer chains. Laforin contains a carbohydrate binding motif (CBM) [31, 37] that preferentially binds longer, versus shorter, glucan chains [38]. Considering all above, one can speculate that the malin-laforin complex tethers to these longer, ‘at-risk’, soluble glycogen molecules through laforin’s CBM, and may act to prevent ‘at-risk’ molecules from proceeding to precipitate.

If it is postulated that the malin-laforin complex acts on these ‘at risk’ soluble glycogen molecule, how does the complex regulate solubility of glycogen? The answer may resides in finding the glycogen metabolism related protein substrate(s) for the malin E3 ubiquitin ligase. Numerous malin ubiquitination substrates have been proposed at least based on in-vitro overexpression experiments (reviewed in [12]). Among them GS and Protein Targeting to Glycogen (PTG) are notable because of their direct importance in glycogen biology [3942]. Indeed, genetic downregulation of GS (described later in ‘Therapeutic approaches to LD’ of this review) activity corrected glycogen chain lengths and prevented polyglucosan formation in LD mouse models, indicating, unsurprisingly, that the latter is dependent on GS. Additionally, knocking out subunits of protein phosphatase 1α (which is a GS activator), namely the protein phosphatase 1 regulatory subunit 3c (PP1R3c, also known as PTG) or the protein phosphatase 1 regulatory subunit 3d (PP1R3d, also known as R6), reduced LB formation, emphasizing that the regulation of glycogen synthase activity and chain elongation are key to LD pathogenesis [4346].

These and other results lead to the proposal of a mechanistic model for the malin-laforin complex as a quality control system for glycogen metabolism (Figure 1, adapted from [47]): The hypothesis is that the GBE to GS activities ratio is occasionally out of balance at localized sites of particular glycogen molecules, engendering overlong chains and resultant risk of precipitation for those specific molecules. An unknown kinase phosphorylates these long chains, preventing their intertwining (in amylopectin, phosphorylation separates double helices to allow hydration and access for chain-digesting enzymes [48]). The phosphorylated long chains attract laforin, and with it malin, which ubiquitinates and inhibits GS and/or GS activating enzymes locally. The pathogenic over-elongation stalled, the chain-digesting enzyme glycogen phosphorylase re-shortens the long chains but cannot proceed beyond the phosphate (a known feature of this enzyme [49]). Laforin removes the phosphate, allowing phosphorylase to complete the shortening/remodeling (in plants the phosphatase SEX4, orthologous to and complementable by laforin, removes added phosphates to permit continued chain shortening by plant phosphorylases/exoamylases [50, 51]). Finally, malin ubiquitinates and removes laforin. In the absence of laforin or malin, chain over-elongations are not checked, and the at-risk molecules precipitate and accumulate slowly over time. Overall tissue or cell level GS and GBE activities are normal (i.e. not measurably altered), because the causative GBE/GS imbalance is local at a very small fraction of total glycogen molecules at any one time.

Figure 1. Hypothetical role of the laforin-malin complex in glycogen metabolism.

Figure 1.

An unknown kinase phosphorylates a pre-existing overly long and poorly branched glycogen chain formed due to occasional local imbalance in glycogen synthase-glycogen branching enzyme activities (1). Laforin-malin is attracted to and docks on this overlong chain (boxed) via laforin’s carbohydrate binding domain (2). Malin ubiquitinates and degrades glycogen synthase or glycogen synthase activating proteins such as protein phosphatase-1α (3). Laforin removes the phosphate (4) allowing chain re-shortening by glycogen phosphorylase (not shown in the picture) (5). Finally, malin ubiquitinates and removes laforin. The end result is a corrected chain with restoration of structural integrity and mitigation of precipitation of the glycogen molecule. Note: Steps 3–5 are shown only with the long glycogen chain.

LD at a crossroad of neuroinflammation, neurodegeneration and epilepsy:

Another major unknown in LD research is how the abnormal glycogen accumulation drives the neurodegenerative and epileptic phenotypes? Laforin and malin knockout mouse models show neurodegenerative phenotypes of neuronal loss and astrogliosis [45, 5257]. The PBs are present in both astrocytes and neurons with distinctive characteristics [58, 59]. Neuronal PBs are spherical and large in size and present in the somatic region, whereas astrocytic PBs are smaller, found in clusters mainly in the astrocytic processes, and have resemblance to Corpora Amylacea (CA) of normal aged brains [59]. How the inclusion of two different types of PBs lead to neurodegeneration is not understood. However, one can assume that accumulation of these ‘unwanted’ toxic products in neurons and astrocytes may perturb the normal physiology of brain. The accumulating LBs may also be drivers of an inflammatory or autoimmune process. In fact, malin knockout brains show reactive astrocytes, microglia and high levels of proinflammatory markers compared to wild type [60]. Indeed, both laforin and malin knockout mouse brains overexpress neuroinflammatory genes compared to WT in progressive fashion with increasing age [60, 61]. In addition, PBs may affect particular cell types, which may explain the particularly severe epilepsy. In a recent study, when glycogen synthase was ablated specifically from astrocytes in malin knockout mice, the mice no longer showed PB accumulation, astrogliosis/microgliosis and autophagy impairment. However in these mice, the epileptic phenotype was not corrected. On the other hand, over-accumulation of glycogen in astrocytes led to a neurodegnerative phenotype [62]. These results reinforce the idea that astrocytic PB accumulation could be the driving force behind neurodegeneration, but perhaps neuronal pathology underlies the epilepsy component of the disease. PBs were noted in GABAergic interneurons of laforin knockout mice, associated with a particular diminution of the numbers of these inhibitory neurons. Also, these same studies revealed decreased levels of neuronal growth factors, which in turn may contribute to the neurodegenerative process [63]. Lastly, astrocytic glutamate transporter, GLT-1 which is responsible for removing excess of glutamate from synaptic cleft, was shown to have altered distribution with less glutamate uptake activity in cultured primary astrocytes from both laforin and malin knockout mouse models [64, 65]. Recently, the Sanz group reported a possible role of malin-laforin complex in ubiquitination-mediated delay for the endocytosis of GLT-1 receptor in a cell culture-based system suggesting the importance of the complex in GLT-turnover [66]. Indeed, as shown previously, altering the activity of either GLT-1 or blocking GABA mediated neurotransmission by pharmacological interventions in malin knockout mice resulted in significantly increased glutamate levels in the brain suggesting a plausible role of glutamate homeostasis in the epileptic phenotype of LD [65].

Possible role of the malin-laforin complex in ubiquitination mediated glycophagy:

Several observations suggest a role for defective autophagy in LD, including: i) presence of ubiquitin positive proteinaceous materials in LBs [67]; ii) in-vitro interaction of malin-laforin complex with autophagy related PI3KC3 complex [68] and adaptor protein P62 [69]; iii) most often, in in-vitro conditions, malin-laforin complex ubiquitinates protein substrates with K-63 linked ubiquitin linkage – a type of ubiquitination that strictly associates with autophagy (reviewed in [12]) iv) a decrease in proteosomal activity and activation of autophagy related mTOR signaling pathway in laforin knockout mouse model [70], and lastly v) impairment of autophagosome formation in malin knockout mice [67]. Furthermore, it has been observed that autophagy related pathways such as mitophagy [71], lysosomal pathway [72], endoplasmic reticulum stress [7375] and reactive oxygen species generation are all affected in LD [76]. However, the precise connection between autophagy and LD pathology remains much debated with contrasting results from LD mouse model tissues where several of the tested autophagy markers remain unaltered [33, 77]. Nevertheless, it could be a possibility that autophagy is a secondary effect of LB formation where malin-laforin complex tethers K-63 linked ubiquitin chains onto their protein substrates attached to LBs and targets it to the autophagosome for degradation. Some interesting findings substantiate this hypothesis, such as: i) striking similarities between CA from aged brain and LBs in terms of their shape, size and low phosphate content [20] ii) CA and LBs both being decorated with ubiquitin and P62 [58, 78] and most intriguingly iii) presence of a neo-epitopes on CA that could be recognized and cleared by the body’s natural immune system [58, 79]. Overall, the precise connection between LD and autophagy is yet to be clarified.

Therapeutic approaches to LD:

The current basis of therapy for patients with LD is to control the severity and frequency of seizures and myoclonus. Anti-epileptic drugs are the only available treatments [13, 80]. Despite some advancement in understanding the disease mechanism, patients are still lacking targeted or curative therapy for the disease [13, 81].

Gene therapy offers much hope for debilitating genetic disorders and has considerably advanced the development of treatments for hereditary diseases [8284]. The limited number of genes (EPM2A or EPM2B) involved in LD makes the disease a good candidate for gene replacement therapies. The functional copy of the mutated gene can be delivered to compensate for the deficiency. In addition to the gene replacement therapies, downregulation of glycogen synthesis by targeting GS at the DNA, RNA, or protein level and degradation of LBs are also being investigated as possible therapeutic options for LD (Figure 2). Moreover, other currently available dietary modifications could be used as an addition to these more targeted approaches. In a recent paper, Israelian et al. showed that the ketogenic diet can reduce abnormal glycogen accumulation in a mouse model of LD [85]. They suggested initiation of the diet at the diagnosis of LD, preferably through an internationally organized clinical trial to clarify the particular role of the diet in patients.

Figure 2. Overview of therapeutic strategies in Lafora disease.

Figure 2.

Therapeutic approaches to LD can be classified into two main groups. The first group aims to prevent or arrest the disease progression by targeting glycogen synthase or replacing EPM2A and EPM2B genes. The second group of therapeutics aims to alleviate the existing disease by degrading Lafora bodies and improving LD related neuroinflammation.

Gene Replacement Therapy:

In LD, EPM2A or EPM2B cDNA can be delivered to make up the deficient proteins. This can be achieved by several different delivery methods including viruses, viral-like particles, gold particles, nanoparticles, exosomes, and liposomes [8691]. Currently, Adeno-Associated Viruses are the most efficient and safe gene delivery vehicles in CNS-directed gene replacement therapies [89]. This is mainly due to their non-pathogenic natures, transduction efficiencies, and long-lasting transgene expression with low-frequency integration into the host genome [92]. Although their packaging capacity is a limiting factor for many diseases, both EPM2A and EPM2B cDNAs are below the size limit. Despite many advantages, there are two main obstacles in treating CNS disorders with AAVs. The first is the blood-brain barrier (BBB) which limits the number of AAV particles that transduce brain parenchyma when injected intravenously. One way to circumvent the BBB is to inject virus directly into cerebrospinal fluid (CSF). Currently, different routes of intra-CSF injections, for example, intrathecal administration, are being investigated in clinical gene therapy trials [9395]. Intra-CSF injections have certain advantages compared to systemic injections. Delivery via the CSF might evade the loss of the virus by pre-existing neutralizing antibodies and prevent the off-target accumulation of viruses in tissues like the liver, kidney, etc. Thus, intra-CSF injections might require less amount of virus to achieve the same efficacy as with systemic injections, and fewer systemic side effects. The other area of research is to engineer new viral capsids that can better cross BBB and distribute more broadly in the brain [89, 92, 96]. While new capsids are being developed, one of the naturally occurring AAV serotypes, namely AAV9, can cross the blood-brain barrier in substantial amounts and transduce neuronal cells. This is the present-day vector of choice for CNS directed gene therapies. For example, AAV9 is the capsid used to deliver the missing gene in the recently FDA-approved CNS-directed gene replacement therapy for spinal muscular atrophy (SMA) [82, 97].

The second major hurdle in treating diseases that affect most of the brain is transduction efficiency [84, 8789]. In LD, practically every cell in the brain is diseased and abnormal PBs are formed throughout the brain. Thus, widespread CNS transduction is required. Viral capsid engineering holds hope for this problem. Despite scientific advancements in virus capsid engineering, currently, even with the earliest direct intra-CSF injections the transduction efficiency is limited [84, 89, 93, 96, 98]. Besides capsid engineering, new technologies, such as focused ultrasound, to manipulate the blood-brain barrier are being investigated and preclinical results are promising. These new methods are considered fairly safe and are routinely used in clinical practice, and could easily be used for the enhancement of viral transduction efficiency concurrently with other methods to increase the effect of gene therapies for LD [99, 100]. Once a suitable strategy is identified, early intervention will always be critical to arresting neurodegeneration and prevent extensive irreversible CNS damage.

Degradation of Lafora bodies:

Another therapeutic target for ameliorating LD is the degradation of accumulated LBs. LBs are the main drivers of the disease pathology and elimination of LBs rescues the neurological phenotype in mouse models. One way to achieve this goal is by delivering a polyglucan-degrading enzyme (amylase) to the brain. Recently, the Gentry group fused pancreatic α-amylase to a cell-penetrating antibody fragment and generated an antibody-enzyme fusion (VAL-0417) designed to penetrate cells and degrade LBs. They showed that this antibody-enzyme fusion degraded LBs in-vitro and reduced in-vivo in a mouse model of LD. Furthermore, they demonstrated that VAL-0417 reverses the physiological effects of LB accumulation. Although its effects are only tested in a mouse model, this drug has the potential for providing significant clinical benefit to LD patients as an example of precision therapy [101103].

Reducing brain glycogen synthesis:

One of the most promising therapeutic avenues for LD is the reduction of brain glycogen synthesis. Partial or full removal of the GS enzyme prevented LB formation and rescued the neurological phenotype in LD mouse models. These findings led to the hypothesis that LD could be prevented by inhibiting glycogen synthesis. Moreover, it was shown that a ~50% decrease in GS activity might be sufficient to halt disease progression [104, 105]. GS can be targeted at the DNA, RNA, or protein levels. The CRISPR–Cas9 system is the most promising tool for DNA level manipulations and has greatly simplified this approach and is currently being investigated in clinical trials [106]. A commonly used application of CRISPR–Cas9 is to knockout a target gene by introducing double-stranded DNA breaks (DSBs). In post-mitotic cells, like neurons, these DSBs are repaired by non-homologous end-joining (NHEJ), leading to permanent indel formation and a non-functional protein [107, 108]. This knockout approach can applied to LD by targeting the GS isoform expressed in the brain (GYS1) and rendering it non-functional. As for gene replacement therapy, for efficient and widespread editing, CRISPR–Cas9-mediated therapy will require AAV-mediated delivery to the CNS. In fact, in our recent study, we showed that AAV-SaCas9 decreases LB accumulation and neuroinflammation in LD mouse models [109]. Despite the practicality of the system, the CRISPR-Cas9 system has certain downsides including off-target effects and immune response to bacterial Cas9 protein [89, 110]. Improving the CRISPR-Cas9 system for human in-vivo applications is one of the hottest areas in gene therapy research currently [84, 88, 107, 108, 111, 112].

Oligonucleotide-based therapeutics or virally delivered other RNA interference (RNAi) methods can be used to target GS at the mRNA level. These are an emerging class of drugs that enable potent and efficient modulation of gene expression. These methods have made significant progress in recent years [113, 114]. Many candidates are currently in clinical trials for targeting RNAs involved in various diseases including neurodegenerative diseases, and some were approved by the FDA. Among these, Antisense oligonucleotides (ASOs) are chemically modified, short synthetic nucleic acids that bind to target mRNA and lead to its degradation [115, 116]. Recently, in one of our studies we used Gys1-ASO to prevent LB formation in young mice. In addition, the therapy stopped further LB accumulation in older LD mouse models [117]. RNAi approaches operate similarly, sequence specifically and post-transcriptionally by activating ribonucleases, but via different mechanisms in coordination with other enzymes, degrade the RNA. These types of RNAi can be achieved by the delivery of short artificial RNAs, such as microRNAs (miRNAs) and short hairpin RNAs (shRNAs), or divalent siRNAs (di-siRNA [118]. Each of these methods has certain advantages and disadvantages. ASOs do not require viral delivery, but repeated administration is needed to maintain therapeutic levels. Virally delivered miRNAs are injected once in a lifetime but their use is limited to virus-related boundaries. In this regard, recently developed divalent-siRNAs are very promising by having much longer half-lives and better transduction efficiencies compared to ASOs, while still not requiring viral or other vehicles for delivery. However, the divalent-siRNA technology is new and there are not yet many studies regarding its formulations, effectiveness in the clinic and side effects. Overall, the use of any of the methods alone or in combination with other therapies to target the mRNA encoding GS in the brain is a viable option for the treatment of LD [96, 113, 114, 118124].

Small-molecule therapies and repurposing drugs:

One of the more traditional approaches for LD is a small-molecule therapy. Although small molecules cannot replace the function of laforin or malin at the gene level but could affect the metabolic pathways that lead to abnormal glycogen accumulation. Recently, a high-throughput screening assay has been developed to identify small molecules that inhibit GS activity to treat brain glycogenosis, namely adult polyglucosan body disease, which could also be effective in LD [104]. Despite many challenges to develop a small-molecule therapy, it could provide the least invasive and the most cost-effective option for patients if administrable in the form of a digestible pill [125].

Besides searching for new small molecules, repurposing currently available drugs to target LD at a pathophysiological level is another viable and more straightforward approach in the treatment of LD. Using this approach, the Sanz group showed that treatment with metformin decreases the accumulation of LBs and polyubiquitin protein aggregates, and reduced seizure susceptibility in an LD mouse model [57, 126]. Based on this study, the European Medicines Agency granted orphan designation to metformin for the treatment of LD. Even though metformin was safe in a small cohort of LD patients, the clinical outcome remains uncertain. Recently, Molla et al., using a similar approach, investigated propranolol and epigallocatechin gallate (EGCG) as potential therapies in an LD mouse model due to their anti-inflammatory properties. Their study further highlights the potential therapeutic effectiveness of the modulators of inflammation as novel treatments in Lafora disease [75, 127129].

Conclusion:

LD is an orphan disease with much unknown basic mechanism. Therefore, the disease presents unique opportunities for basic science researchers to study diverse mechanistic pathways including glycogen biology, protein ubiquitination, neuroinflammation, neurodegeneration and epilepsy. These studies will uncover important unknown mechanisms in the brain, and also beyond. For example, LD related work recently disclosed a role for malin in small cell lung cancer [130, 131]. Finally, LD is ripe for therapy and rife with possible therapeutic approaches, including disease gene replacement, but also intervention in the already uncovered biochemical disease pathway. Advances in this or other rare diseases also lays the groundwork to therapies of more common and complex diseases of the brain.

Acknowledgments:

This work was funded by the National Institutes of Health under award P01NS097197. B.A.M. holds the University of Texas Southwestern Jimmy Elizabeth Westcott Chair in Pediatric Neurology.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References:

  • [1].Collaborators GN. Global, regional, and national burden of neurological disorders, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol 2019; 18:459–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Ardley HC, Hung CC, Robinson PA. The aggravating role of the ubiquitin-proteasome system in neurodegeneration. FEBS Lett 2005;579(3):571–6. [DOI] [PubMed] [Google Scholar]
  • [3].Ardley HC, Robinson PA. The role of ubiquitin-protein ligases in neurodegenerative disease. Neurodegener Dis 2004;1(2–3):71–87. [DOI] [PubMed] [Google Scholar]
  • [4].McKinnon C, Tabrizi SJ. The ubiquitin-proteasome system in neurodegeneration. Antioxid Redox Signal 2014;21(17):2302–21. [DOI] [PubMed] [Google Scholar]
  • [5].Popovic D, Vucic D, Dikic I. Ubiquitination in disease pathogenesis and treatment. Nat Med 2014;20(11):1242–53. [DOI] [PubMed] [Google Scholar]
  • [6].Rape M Ubiquitylation at the crossroads of development and disease. Nat Rev Mol Cell Biol 2018;19(1):59–70. [DOI] [PubMed] [Google Scholar]
  • [7].Zhu J, Tsai NP. Ubiquitination and E3 Ubiquitin Ligases in Rare Neurological Diseases with Comorbid Epilepsy. Neuroscience 2020;428:90–9. [DOI] [PubMed] [Google Scholar]
  • [8].George AJ, Hoffiz YC, Charles AJ, Zhu Y, Mabb AM. A Comprehensive Atlas of E3 Ubiquitin Ligase Mutations in Neurological Disorders. Front Genet 2018;9:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Cenacchi G, Papa V, Costa R, Pegoraro V, Marozzo R, Fanin M, et al. Update on polyglucosan storage diseases. Virchows Arch 2019;475(6):671–86. [DOI] [PubMed] [Google Scholar]
  • [10].Hedberg-Oldfors C, Oldfors A. Polyglucosan storage myopathies. Mol Aspects Med 2015;46:85–100. [DOI] [PubMed] [Google Scholar]
  • [11].Nilsson J, Schoser B, Laforet P, Kalev O, Lindberg C, Romero NB, et al. Polyglucosan body myopathy caused by defective ubiquitin ligase RBCK1. Ann Neurol 2013;74(6):914–9. [DOI] [PubMed] [Google Scholar]
  • [12].Garcia-Gimeno MA, Knecht E, Sanz P. Lafora Disease: A Ubiquitination-Related Pathology. Cells 2018;7(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Nitschke F, Ahonen SJ, Nitschke S, Mitra S, Minassian BA. Lafora disease - from pathogenesis to treatment strategies. Nat Rev Neurol 2018;14(10):606–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Turnbull J, Tiberia E, Striano P, Genton P, Carpenter S, Ackerley CA, et al. Lafora disease. Epileptic Disord 2016;18(S2):38–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].d’Orsi G, Lalla A, Palumbo O, Di Claudio MT, Valenzano A, Sabetta A, et al. The presenting symptoms of Lafora Disease: An electroclinical and genetic study in five Apulian (Southern Italy) families. Seizure 2020;83:145–53. [DOI] [PubMed] [Google Scholar]
  • [16].Brenner D, Baumgartner T, von Spiczak S, Lewerenz J, Weis R, Grimmer A, et al. Genotypes and phenotypes of patients with Lafora disease living in Germany. Neurol Res Pract 2019;1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Lafora GRG B . Beitrag zur Histopathologie der myoklonischen Epilepsie [German]. Z. Gesamte Neurol Psychiatr 1911;6:6–14. [Google Scholar]
  • [18].Andrade DM, Turnbull J, Minassian BA. Lafora disease, seizures and sugars. Acta Myol 2007;26(1):83–6. [PMC free article] [PubMed] [Google Scholar]
  • [19].Allegranza A GPSaPC. Dati istochimici in un caso di mioclonoepilepsia di Unverricht-Lundborg. Riv Pat nerv ment 1965;86:386–406. [Google Scholar]
  • [20].Cavanagh JB. Corpora-amylacea and the family of polyglucosan diseases. Brain Res Brain Res Rev 1999;29(2–3):265–95. [DOI] [PubMed] [Google Scholar]
  • [21].Oksel F, Tekgul H, Genc S, Ozyurek R, Akalin T, Tututuncuoglu S. A case of Lafora’s disease associated with cardiac arrhythmia. J Child Neurol 1999;14(11):745–6. [DOI] [PubMed] [Google Scholar]
  • [22].Girard JM, Turnbull J, Ramachandran N, Minassian BA. Progressive myoclonus epilepsy. Handb Clin Neurol 2013;113:1731–6. [DOI] [PubMed] [Google Scholar]
  • [23].Chan EM, Young EJ, Ianzano L, Munteanu I, Zhao X, Christopoulos CC, et al. Mutations in NHLRC1 cause progressive myoclonus epilepsy. Nat Genet 2003;35(2):125–7. [DOI] [PubMed] [Google Scholar]
  • [24].Roma-Mateo C, Moreno D, Vernia S, Rubio T, Bridges TM, Gentry MS, et al. Lafora disease E3-ubiquitin ligase malin is related to TRIM32 at both the phylogenetic and functional level. BMC Evol Biol 2011;11:225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Calder PC. Glycogen structure and biogenesis. Int J Biochem 1991;23(12):1335–52. [DOI] [PubMed] [Google Scholar]
  • [26].Roach PJ, Depaoli-Roach AA, Hurley TD, Tagliabracci VS. Glycogen and its metabolism: some new developments and old themes. Biochem J 2012;441(3):763–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Sullivan MA, Nitschke S, Skwara EP, Wang P, Zhao X, Pan XS, et al. Skeletal Muscle Glycogen Chain Length Correlates with Insolubility in Mouse Models of Polyglucosan-Associated Neurodegenerative Diseases. Cell Rep 2019;27(5):1334–44 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Tagliabracci VS, Turnbull J, Wang W, Girard JM, Zhao X, Skurat AV, et al. Laforin is a glycogen phosphatase, deficiency of which leads to elevated phosphorylation of glycogen in vivo. Proc Natl Acad Sci USA 2007;104(49):19262–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Tagliabracci VS, Girard JM, Segvich D, Meyer C, Turnbull J, Zhao X, et al. Abnormal metabolism of glycogen phosphate as a cause for Lafora disease. J Biol Chem 2008;283(49):33816–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Gentry MS, Dowen RH 3rd, Worby CA, Mattoo S, Ecker JR, Dixon JE. The phosphatase laforin crosses evolutionary boundaries and links carbohydrate metabolism to neuronal disease. J Cell Biol 2007;178(3):477–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Dias DM, Furtado J, Wasielewski E, Cruz R, Costello B, Cole L, et al. Biophysical characterization of laforin-carbohydrate interaction. Biochem J 2016;473(3):335–45. [DOI] [PubMed] [Google Scholar]
  • [32].Gayarre J, Duran-Trio L, Criado Garcia O, Aguado C, Juana-Lopez L, Crespo I, et al. The phosphatase activity of laforin is dispensable to rescue Epm2a−/− mice from Lafora disease. Brain 2014;137(Pt 3):806–18. [DOI] [PubMed] [Google Scholar]
  • [33].Nitschke F, Sullivan MA, Wang P, Zhao X, Chown EE, Perri AM, et al. Abnormal glycogen chain length pattern, not hyperphosphorylation, is critical in Lafora disease. EMBO Mol Med 2017;9(7):906–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Gentry MS, Worby CA, Dixon JE. Insights into Lafora disease: malin is an E3 ubiquitin ligase that ubiquitinates and promotes the degradation of laforin. Proc Natl Acad Sci USA 2005;102(24):8501–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].DePaoli-Roach AA, Tagliabracci VS, Segvich DM, Meyer CM, Irimia JM, Roach PJ. Genetic depletion of the malin E3 ubiquitin ligase in mice leads to lafora bodies and the accumulation of insoluble laforin. J Biol Chem 2010;285(33):25372–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Tiberia E, Turnbull J, Wang T, Ruggieri A, Zhao XC, Pencea N, et al. Increased laforin and laforin binding to glycogen underlie Lafora body formation in malin-deficient Lafora disease. J Biol Chem 2012;287(30):25650–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Christiansen C, Abou Hachem M, Janecek S, Vikso-Nielsen A, Blennow A, Svensson B. The carbohydrate-binding module family 20--diversity, structure, and function. FEBS J 2009;276(18):5006–29. [DOI] [PubMed] [Google Scholar]
  • [38].Chan EM, Ackerley CA, Lohi H, Ianzano L, Cortez MA, Shannon P, et al. Laforin preferentially binds the neurotoxic starch-like polyglucosans, which form in its absence in progressive myoclonus epilepsy. Hum Mol Genet 2004;13(11):1117–29. [DOI] [PubMed] [Google Scholar]
  • [39].Fernandez-Sanchez ME, Criado-Garcia O, Heath KE, Garcia-Fojeda B, Medrano-Fernandez I, Gomez-Garre P, et al. Laforin, the dual-phosphatase responsible for Lafora disease, interacts with R5 (PTG), a regulatory subunit of protein phosphatase-1 that enhances glycogen accumulation. Hum Mol Genet 2003;12(23):3161–71. [DOI] [PubMed] [Google Scholar]
  • [40].Worby CA, Gentry MS, Dixon JE. Malin decreases glycogen accumulation by promoting the degradation of protein targeting to glycogen (PTG). J Biol Chem 2008;283(7):4069–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Vernia S, Solaz-Fuster MC, Gimeno-Alcaniz JV, Rubio T, Garcia-Haro L, Foretz M, et al. AMP-activated protein kinase phosphorylates R5/PTG, the glycogen targeting subunit of the R5/PTG-protein phosphatase 1 holoenzyme, and accelerates its down-regulation by the laforin-malin complex. J Biol Chem 2009;284(13):8247–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Lohi H, Ianzano L, Zhao XC, Chan EM, Turnbull J, Scherer SW, et al. Novel glycogen synthase kinase 3 and ubiquitination pathways in progressive myoclonus epilepsy. Hum Mol Genet 2005;14(18):2727–36. [DOI] [PubMed] [Google Scholar]
  • [43].Turnbull J, DePaoli-Roach AA, Zhao X, Cortez MA, Pencea N, Tiberia E, 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]
  • [44].Pederson BA, Turnbull J, Epp JR, Weaver SA, Zhao X, Pencea N, 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]
  • [45].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):3147–56. [DOI] [PubMed] [Google Scholar]
  • [46].Israelian L, Nitschke S, Wang P, Zhao X, Perri AM, Lee JPY, et al. Ppp1r3d deficiency preferentially inhibits neuronal and cardiac Lafora body formation in a mouse model of the fatal epilepsy Lafora disease. J Neurochem 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Sullivan MA, Nitschke S, Steup M, Minassian BA, Nitschke F. Pathogenesis of Lafora Disease: Transition of Soluble Glycogen to Insoluble Polyglucosan. Int J Mol Sci 2017;18(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Hejazi M, Fettke J, Haebel S, Edner C, Paris O, Frohberg C, et al. Glucan, water dikinase phosphorylates crystalline maltodextrins and thereby initiates solubilization. Plant J 2008;55(2):323–34. [DOI] [PubMed] [Google Scholar]
  • [49].Newgard CB, Hwang PK, Fletterick RJ. The family of glycogen phosphorylases: structure and function. Crit Rev Biochem Mol Biol 1989;24(1):69–99. [DOI] [PubMed] [Google Scholar]
  • [50].Hejazi M, Fettke J, Kotting O, Zeeman SC, Steup M. The Laforin-like dual-specificity phosphatase SEX4 from Arabidopsis hydrolyzes both C6- and C3-phosphate esters introduced by starch-related dikinases and thereby affects phase transition of alpha-glucans. Plant Physiol 2010;152(2):711–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Kotting O, Santelia D, Edner C, Eicke S, Marthaler T, Gentry MS, et al. STARCH-EXCESS4 is a laforin-like Phosphoglucan phosphatase required for starch degradation in Arabidopsis thaliana. Plant Cell 2009;21(1):334–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Valles-Ortega J, Duran J, Garcia-Rocha M, Bosch C, Saez I, Pujadas L, et al. Neurodegeneration and functional impairments associated with glycogen synthase accumulation in a mouse model of Lafora disease. EMBO Mol Med 2011;3(11):667–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Ganesh S, Delgado-Escueta AV, Sakamoto T, Avila MR, Machado-Salas J, Hoshii Y, 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):1251–62. [DOI] [PubMed] [Google Scholar]
  • [54].Rubio-Villena C, Viana R, Bonet J, Garcia-Gimeno MA, Casado M, Heredia M, et al. Astrocytes: new players in progressive myoclonus epilepsy of Lafora type. Hum Mol Genet 2018;27(7):1290–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Taneja K, Ganesh S. Dendritic spine abnormalities correlate with behavioral and cognitive deficits in mouse models of Lafora disease. J Comp Neurol 2020. [DOI] [PubMed] [Google Scholar]
  • [56].Burgos DF, Cusso L, Sanchez-Elexpuru G, Calle D, Perpinya MB, Desco M, et al. Structural and Functional Brain Abnormalities in Mouse Models of Lafora Disease. Int J Mol Sci 2020;21(20). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Berthier A, Paya M, Garcia-Cabrero AM, Ballester MI, Heredia M, Serratosa JM, et al. Pharmacological Interventions to Ameliorate Neuropathological Symptoms in a Mouse Model of Lafora Disease. Mol Neurobiol 2016;53(2):1296–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Auge E, Pelegri C, Manich G, Cabezon I, Guinovart JJ, Duran J, et al. Astrocytes and neurons produce distinct types of polyglucosan bodies in Lafora disease. Glia 2018;66(10):2094–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Duran J, Gruart A, Lopez-Ramos JC, Delgado-Garcia JM, Guinovart JJ. Glycogen in Astrocytes and Neurons: Physiological and Pathological Aspects. Adv Neurobiol 2019;23:311–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Lahuerta M, Gonzalez D, Aguado C, Fathinajafabadi A, Garcia-Gimenez JL, Moreno-Estelles M, et al. Reactive Glia-Derived Neuroinflammation: a Novel Hallmark in Lafora Progressive Myoclonus Epilepsy That Progresses with Age. Mol Neurobiol 2020;57(3):1607–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Lopez-Gonzalez 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):3119–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Duran J, Hervera A, Markussen KH, Varea O, Lopez-Soldado I, Sun RC, et al. Astrocytic glycogen accumulation drives the pathophysiology of neurodegeneration in Lafora disease. Brain 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Ortolano S, Vieitez I, Agis-Balboa RC, Spuch C. Loss of GABAergic cortical neurons underlies the neuropathology of Lafora disease. Mol Brain 2014;7:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Munoz-Ballester C, Berthier A, Viana R, Sanz P. Homeostasis of the astrocytic glutamate transporter GLT-1 is altered in mouse models of Lafora disease. Biochim Biophys Acta 2016;1862(6):1074–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Munoz-Ballester C, Santana N, Perez-Jimenez E, Viana R, Artigas F, Sanz P. In vivo glutamate clearance defects in a mouse model of Lafora disease. Exp Neurol 2019;320:112959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Perez-Jimenez E, Viana R, Munoz-Ballester C, Vendrell-Tornero C, Moll-Diaz R, Garcia-Gimeno MA, et al. Endocytosis of the glutamate transporter 1 is regulated by laforin and malin: Implications in Lafora disease. Glia 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Criado O, Aguado C, Gayarre J, Duran-Trio L, Garcia-Cabrero AM, Vernia S, et al. Lafora bodies and neurological defects in malin-deficient mice correlate with impaired autophagy. Hum Mol Genet 2012;21(7):1521–33. [DOI] [PubMed] [Google Scholar]
  • [68].Sanchez-Martin P, Lahuerta M, Viana R, Knecht E, Sanz P. Regulation of the autophagic PI3KC3 complex by laforin/malin E3-ubiquitin ligase, two proteins involved in Lafora disease. Biochim Biophys Acta Mol Cell Res 2020;1867(2):118613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Sanchez-Martin P, Roma-Mateo C, Viana R, Sanz P. Ubiquitin conjugating enzyme E2-N and sequestosome-1 (p62) are components of the ubiquitination process mediated by the malin-laforin E3-ubiquitin ligase complex. Int J Biochem Cell Biol 2015;69:204–14. [DOI] [PubMed] [Google Scholar]
  • [70].Aguado C, Sarkar S, Korolchuk VI, Criado O, Vernia S, Boya P, et al. Laforin, the most common protein mutated in Lafora disease, regulates autophagy. Hum Mol Genet 2010;19(14):2867–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Lahuerta M, Aguado C, Sanchez-Martin P, Sanz P, Knecht E. Degradation of altered mitochondria by autophagy is impaired in Lafora disease. FEBS J 2018;285(11):2071–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Puri R, Suzuki T, Yamakawa K, Ganesh S. Dysfunctions in endosomal-lysosomal and autophagy pathways underlie neuropathology in a mouse model for Lafora disease. Hum Mol Genet 2012;21(1):175–84. [DOI] [PubMed] [Google Scholar]
  • [73].Vernia S, Rubio T, Heredia M, Rodriguez de Cordoba S, Sanz P. Increased endoplasmic reticulum stress and decreased proteasomal function in lafora disease models lacking the phosphatase laforin. PLoS One 2009;4(6):e5907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Sharma J, Mukherjee D, Rao SN, Iyengar S, Shankar SK, Satishchandra P, et al. Neuronatin-mediated aberrant calcium signaling and endoplasmic reticulum stress underlie neuropathology in Lafora disease. J Biol Chem 2013;288(13):9482–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Sinha P, Verma B, Ganesh S. Trehalose Ameliorates Seizure Susceptibility in Lafora Disease Mouse Models by Suppressing Neuroinflammation and Endoplasmic Reticulum Stress. Mol Neurobiol 2020. [DOI] [PubMed] [Google Scholar]
  • [76].Roma-Mateo C, Aguado C, Garcia-Gimenez JL, Ibanez-Cabellos JS, Seco-Cervera M, Pallardo FV, et al. Increased oxidative stress and impaired antioxidant response in Lafora disease. Mol Neurobiol 2015;51(3):932–46. [DOI] [PubMed] [Google Scholar]
  • [77].Wang P, Israelian L, Xue Y, Song S, Attisano L, Minassian BA. SGK1 (glucose transport), dishevelled2 (wnt signaling), LC3/p62 (autophagy) and p53 (apoptosis) proteins are unaltered in Lafora disease. All Results J Biol 2016;7(3):28–33. [PMC free article] [PubMed] [Google Scholar]
  • [78].Sinadinos C, Valles-Ortega J, Boulan L, Solsona E, Tevy MF, Marquez M, et al. Neuronal glycogen synthesis contributes to physiological aging. Aging Cell 2014;13(5):935–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Auge E, Duran J, Guinovart JJ, Pelegri C, Vilaplana J. Exploring the elusive composition of corpora amylacea of human brain. Sci Rep 2018;8(1):13525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Schorlemmer K, Bauer S, Belke M, Hermsen A, Klein KM, Reif PS, et al. Sustained seizure remission on perampanel in progressive myoclonic epilepsy (Lafora disease). Epilepsy Behav Case Rep 2013;1:118–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Verhalen B, Arnold S, Minassian BA. Lafora Disease: A Review of Molecular Mechanisms and Pathology. Neuropediatrics 2018;49(6):357–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Waldrop MA, Karingada C, Storey MA, Powers B, Iammarino MA, Miller NF, et al. Gene Therapy for Spinal Muscular Atrophy: Safety and Early Outcomes. Pediatrics 2020;146(3). [DOI] [PubMed] [Google Scholar]
  • [83].Mendell JR, Al-Zaidy SA, Rodino-Klapac LR, Goodspeed K, Gray SJ, Kay CN, et al. Current Clinical Applications of In Vivo Gene Therapy with AAVs. Mol Ther 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Martier R, Konstantinova P. Gene Therapy for Neurodegenerative Diseases: Slowing Down the Ticking Clock. Front Neurosci 2020;14:580179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Israelian L, Wang P, Gabrielian S, Zhao X, Minassian BA. Ketogenic diet reduces Lafora bodies in murine Lafora disease. Neurol Genet 2020;6(6):e533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Song X, Liu C, Wang N, Huang H, He S, Gong C, et al. Delivery of CRISPR/Cas systems for cancer gene therapy and immunotherapy. Adv Drug Deliv Rev 2021;168:158–80. [DOI] [PubMed] [Google Scholar]
  • [87].Chuang YF, Phipps AJ, Lin FL, Hecht V, Hewitt AW, Wang PY, et al. Approach for in vivo delivery of CRISPR/Cas system: a recent update and future prospect. Cell Mol Life Sci 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Wang D, Zhang F, Gao G. CRISPR-Based Therapeutic Genome Editing: Strategies and In Vivo Delivery by AAV Vectors. Cell 2020;181(1):136–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Kimura S, Harashima H. Current Status and Challenges Associated with CNS-Targeted Gene Delivery across the BBB. Pharmaceutics 2020;12(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Jiao Y, Xia ZL, Ze LJ, Jing H, Xin B, Fu S. Research Progress of nucleic acid delivery vectors for gene therapy. Biomed Microdevices 2020;22(1):16. [DOI] [PubMed] [Google Scholar]
  • [91].Vemana HP, Saraswat A, Bhutkar S, Patel K, Dukhande VV. A novel gene therapy for neurodegenerative Lafora disease via EPM2A-loaded DLinDMA lipoplexes. Nanomedicine (Lond) 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov 2019;18(5):358–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Lykken EA, Shyng C, Edwards RJ, Rozenberg A, Gray SJ. Recent progress and considerations for AAV gene therapies targeting the central nervous system. J Neurodev Disord 2018;10(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Deverman BE, Ravina BM, Bankiewicz KS, Paul SM, Sah DWY. Gene therapy for neurological disorders: progress and prospects. Nat Rev Drug Discov 2018;17(9):641–59. [DOI] [PubMed] [Google Scholar]
  • [95].Bailey RM, Armao D, Nagabhushan Kalburgi S, Gray SJ. Development of Intrathecal AAV9 Gene Therapy for Giant Axonal Neuropathy. Mol Ther Methods Clin Dev 2018;9:160–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Li C, Samulski RJ. Engineering adeno-associated virus vectors for gene therapy. Nat Rev Genet 2020;21(4):255–72. [DOI] [PubMed] [Google Scholar]
  • [97].Iftikhar M, Frey J, Shohan MJ, Malek S, Mousa SA. Current and emerging therapies for Duchenne muscular dystrophy and spinal muscular atrophy. Pharmacol Ther 2020:107719. [DOI] [PubMed] [Google Scholar]
  • [98].Pietersz KL, Martier RM, Baatje MS, Liefhebber JM, Brouwers CC, Pouw SM, et al. Transduction patterns in the CNS following various routes of AAV-5-mediated gene delivery. Gene Ther 2020. [DOI] [PubMed] [Google Scholar]
  • [99].Lin F, Wu D, Yu J, Weng H, Chen L, Meng F, et al. Comparison of efficacy of deep brain stimulation and focused ultrasound in parkinsonian tremor: a systematic review and network meta-analysis. J Neurol Neurosurg Psychiatry 2021. [DOI] [PubMed] [Google Scholar]
  • [100].Lennon JC, Hassan I. Magnetic resonance-guided focused ultrasound for Parkinson’s disease since ExAblate, 2016–2019: a systematic review. Neurol Sci 2021. [DOI] [PubMed] [Google Scholar]
  • [101].Zhou Z, Austin GL, Shaffer R, Armstrong DD, Gentry MS. Antibody-Mediated Enzyme Therapeutics and Applications in Glycogen Storage Diseases. Trends Mol Med 2019;25(12):1094–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Brewer MK, Uittenbogaard A, Austin GL, Segvich DM, DePaoli-Roach A, Roach PJ, et al. Targeting Pathogenic Lafora Bodies in Lafora Disease Using an Antibody-Enzyme Fusion. Cell Metab 2019;30(4):689–705 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Austin GL, Simmons ZR, Klier JE, Rondon A, Hodges BL, Shaffer R, et al. Central Nervous System Delivery and Biodistribution Analysis of an Antibody-Enzyme Fusion for the Treatment of Lafora Disease. Mol Pharm 2019;16(9):3791–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].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]
  • [105].Nitschke S, Chown EE, Zhao X, Gabrielian S, Petkovic S, Guisso DR, et al. An inducible glycogen synthase-1 knockout halts but does not reverse Lafora disease progression in mice. J Biol Chem 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].He S The first human trial of CRISPR-based cell therapy clears safety concerns as new treatment for late-stage lung cancer. Signal Transduct Target Ther 2020;5(1):168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Ekman FK, Ojala DS, Adil MM, Lopez PA, Schaffer DV, Gaj T. CRISPR-Cas9-Mediated Genome Editing Increases Lifespan and Improves Motor Deficits in a Huntington’s Disease Mouse Model. Mol Ther Nucleic Acids 2019;17:829–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Gyorgy B, Loov C, Zaborowski MP, Takeda S, Kleinstiver BP, Commins C, et al. CRISPR/Cas9 Mediated Disruption of the Swedish APP Allele as a Therapeutic Approach for Early-Onset Alzheimer’s Disease. Mol Ther Nucleic Acids 2018;11:429–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Gumusgoz E, Guisso DR, Kasiri S, Wu J, Dear M, Verhalen B, et al. Targeting Gys1 with AAV-SaCas9 Decreases Pathogenic Polyglucosan Bodies and Neuroinflammation in Adult Polyglucosan Body and Lafora Disease Mouse Models. Neurotherapeutics 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Sharma G, Sharma AR, Bhattacharya M, Lee SS, Chakraborty C. CRISPR-Cas9: A Preclinical and Clinical Perspective for the Treatment of Human Diseases. Mol Ther 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Ou L, Przybilla MJ, Tabaran AF, Overn P, O’Sullivan MG, Jiang X, et al. A novel gene editing system to treat both Tay-Sachs and Sandhoff diseases. Gene Ther 2020;27(5):226–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Arnaoutova I, Zhang L, Chen HD, Mansfield BC, Chou JY. Correction of metabolic abnormalities in a mouse model of glycogen storage disease type Ia by CRISPR/Cas9-based gene editing. Mol Ther 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].O’Callaghan B, Hofstra B, Handler HP, Kordasiewicz HB, Cole T, Duvick L, et al. Antisense Oligonucleotide Therapeutic Approach for Suppression of Ataxin-1 Expression: A Safety Assessment. Mol Ther Nucleic Acids 2020;21:1006–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Albrechtsen SS, Born AP, Boesen MS. Nusinersen treatment of spinal muscular atrophy - a systematic review. Dan Med J 2020;67(9). [PubMed] [Google Scholar]
  • [115].Scharner J, Aznarez I. Clinical Applications of Single-Stranded Oligonucleotides: Current Landscape of Approved and In-Development Therapeutics. Mol Ther 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Gheibi-Hayat SM, Jamialahmadi K. Antisense Oligonucleotide (AS-ODN) Technology: Principle, Mechanism and Challenges. Biotechnol Appl Biochem 2020. [DOI] [PubMed] [Google Scholar]
  • [117].Ahonen S, Nitschke S, Grossman TR, Kordasiewicz H, Wang P, Zhao X, et al. Gys1 antisense therapy rescues neuropathological bases of murine Lafora disease. Brain 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Alterman JF, Godinho B, Hassler MR, Ferguson CM, Echeverria D, Sapp E, et al. A divalent siRNA chemical scaffold for potent and sustained modulation of gene expression throughout the central nervous system. Nat Biotechnol 2019;37(8):884–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Silva AC, Lobo DD, Martins IM, Lopes SM, Henriques C, Duarte SP, et al. Antisense oligonucleotide therapeutics in neurodegenerative diseases: the case of polyglutamine disorders. Brain 2020;143(2):407–29. [DOI] [PubMed] [Google Scholar]
  • [120].Caron NS, Southwell AL, Brouwers CC, Cengio LD, Xie Y, Black HF, et al. Potent and sustained huntingtin lowering via AAV5 encoding miRNA preserves striatal volume and cognitive function in a humanized mouse model of Huntington disease. Nucleic Acids Res 2020;48(1):36–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Martier R, Liefhebber JM, Garcia-Osta A, Miniarikova J, Cuadrado-Tejedor M, Espelosin M, et al. Targeting RNA-Mediated Toxicity in C9orf72 ALS and/or FTD by RNAi-Based Gene Therapy. Mol Ther Nucleic Acids 2019;16:26–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Wallace LM, Saad NY, Pyne NK, Fowler AM, Eidahl JO, Domire JS, et al. Pre-clinical Safety and Off-Target Studies to Support Translation of AAV-Mediated RNAi Therapy for FSHD. Mol Ther Methods Clin Dev 2018;8:121–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Mercuri E, Darras BT, Chiriboga CA, Day JW, Campbell C, Connolly AM, et al. Nusinersen versus Sham Control in Later-Onset Spinal Muscular Atrophy. N Engl J Med 2018;378(7):625–35. [DOI] [PubMed] [Google Scholar]
  • [124].Miniarikova J, Zimmer V, Martier R, Brouwers CC, Pythoud C, Richetin K, et al. AAV5-miHTT gene therapy demonstrates suppression of mutant huntingtin aggregation and neuronal dysfunction in a rat model of Huntington’s disease. Gene Ther 2017;24(10):630–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Tang B, Frasinyuk MS, Chikwana VM, Mahalingan KK, Morgan CA, Segvich DM, et al. Discovery and Development of Small-Molecule Inhibitors of Glycogen Synthase. J Med Chem 2020;63(7):3538–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Sanchez-Elexpuru G, Serratosa JM, Sanz P, Sanchez MP. 4-Phenylbutyric acid and metformin decrease sensitivity to pentylenetetrazol-induced seizures in a malin knockout model of Lafora disease. Neuroreport 2017;28(5):268–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Sanz P, Serratosa JM. Neuroinflammation and progressive myoclonus epilepsies: from basic science to therapeutic opportunities. Expert Rev Mol Med 2020;22:e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Bisulli F, Muccioli L, d’Orsi G, Canafoglia L, Freri E, Licchetta L, et al. Treatment with metformin in twelve patients with Lafora disease. Orphanet J Rare Dis 2019;14(1):149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Molla B, Heredia M, Sanz P. Modulators of Neuroinflammation Have a Beneficial Effect in a Lafora Disease Mouse Model. Mol Neurobiol 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Donohue KJ, Gentry MS, Sun RC. The E3 ligase malin plays a pivotal role in promoting nuclear glycogenolysis and histone acetylation. Ann Transl Med 2020;8(5):254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Sun RC, Dukhande VV, Zhou Z, Young LEA, Emanuelle S, Brainson CF, et al. Nuclear Glycogenolysis Modulates Histone Acetylation in Human Non-Small Cell Lung Cancers. Cell Metab 2019;30(5):903–16 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]

Web Reference:

  1. The Lafora Progressive Myoclonus Epilepsy Mutation and Polymorphism Database: http://projects.tcag.ca/lafora/

RESOURCES