Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Mol Ther ; 32(7): 2130-2149, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38796707

ABSTRACT

Lafora disease is a rare and fatal form of progressive myoclonic epilepsy typically occurring early in adolescence. The disease results from mutations in the EPM2A gene, encoding laforin, or the EPM2B gene, encoding malin. Laforin and malin work together in a complex to control glycogen synthesis and prevent the toxicity produced by misfolded proteins via the ubiquitin-proteasome system. Disruptions in either protein cause alterations in this complex, leading to the formation of Lafora bodies containing abnormal, insoluble, and hyperphosphorylated forms of glycogen. We used the Epm2a-/- knockout mouse model of Lafora disease to apply gene therapy by administering intracerebroventricular injections of a recombinant adeno-associated virus carrying the human EPM2A gene. We evaluated the effects of this treatment through neuropathological studies, behavioral tests, video-electroencephalography, electrophysiological recordings, and proteomic/phosphoproteomic analysis. Gene therapy ameliorated neurological and histopathological alterations, reduced epileptic activity and neuronal hyperexcitability, and decreased the formation of Lafora bodies. Moreover, differential quantitative proteomics and phosphoproteomics revealed beneficial changes in various molecular pathways altered in Lafora disease. Our results represent proof of principle for gene therapy with the coding region of the human EPM2A gene as a treatment for EPM2A-related Lafora disease.


Subject(s)
Dependovirus , Disease Models, Animal , Genetic Therapy , Lafora Disease , Mice, Knockout , Protein Tyrosine Phosphatases, Non-Receptor , Lafora Disease/therapy , Lafora Disease/genetics , Lafora Disease/metabolism , Animals , Genetic Therapy/methods , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Mice , Dependovirus/genetics , Humans , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Carrier Proteins/genetics , Carrier Proteins/metabolism , Electroencephalography , Proteomics/methods
3.
J Biol Chem ; 300(5): 107271, 2024 May.
Article in English | MEDLINE | ID: mdl-38588813

ABSTRACT

Lafora disease (LD) is an autosomal recessive myoclonus epilepsy with onset in the teenage years leading to death within a decade of onset. LD is characterized by the overaccumulation of hyperphosphorylated, poorly branched, insoluble, glycogen-like polymers called Lafora bodies. The disease is caused by mutations in either EPM2A, encoding laforin, a dual specificity phosphatase that dephosphorylates glycogen, or EMP2B, encoding malin, an E3-ubiquitin ligase. While glycogen is a widely accepted laforin substrate, substrates for malin have been difficult to identify partly due to the lack of malin antibodies able to detect malin in vivo. Here we describe a mouse model in which the malin gene is modified at the C-terminus to contain the c-myc tag sequence, making an expression of malin-myc readily detectable. Mass spectrometry analyses of immunoprecipitates using c-myc tag antibodies demonstrate that malin interacts with laforin and several glycogen-metabolizing enzymes. To investigate the role of laforin in these interactions we analyzed two additional mouse models: malin-myc/laforin knockout and malin-myc/LaforinCS, where laforin was either absent or the catalytic Cys was genomically mutated to Ser, respectively. The interaction of malin with partner proteins requires laforin but is not dependent on its catalytic activity or the presence of glycogen. Overall, the results demonstrate that laforin and malin form a complex in vivo, which stabilizes malin and enhances interaction with partner proteins to facilitate normal glycogen metabolism. They also provide insights into the development of LD and the rescue of the disease by the catalytically inactive phosphatase.


Subject(s)
Lafora Disease , Protein Tyrosine Phosphatases, Non-Receptor , Ubiquitin-Protein Ligases , Lafora Disease/metabolism , Lafora Disease/genetics , Lafora Disease/pathology , Animals , Mice , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Humans , Dual-Specificity Phosphatases/metabolism , Dual-Specificity Phosphatases/genetics , Disease Models, Animal , Glycogen/metabolism , Glycogen/genetics
4.
Neurotherapeutics ; 20(6): 1808-1819, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37700152

ABSTRACT

Patients with Lafora disease have a mutation in EPM2A or EPM2B, resulting in dysregulation of glycogen metabolism throughout the body and aberrant glycogen molecules that aggregate into Lafora bodies. Lafora bodies are particularly damaging in the brain, where the aggregation drives seizures with increasing severity and frequency, coupled with neurodegeneration. Previous work employed mouse genetic models to reduce glycogen synthesis by approximately 50%, and this strategy significantly reduced Lafora body formation and disease phenotypes. Therefore, an antisense oligonucleotide (ASO) was developed to reduce glycogen synthesis in the brain by targeting glycogen synthase 1 (Gys1). To test the distribution and efficacy of this drug, the Gys1-ASO was administered to Epm2b-/- mice via intracerebroventricular administration at 4, 7, and 10 months. The mice were then sacrificed at 13 months and their brains analyzed for Gys1 expression, glycogen aggregation, and neuronal excitability. The mice treated with Gys1-ASO exhibited decreased Gys1 protein levels, decreased glycogen aggregation, and reduced epileptiform discharges compared to untreated Epm2b-/- mice. This work provides proof of concept that a Gys1-ASO halts disease progression of EPM2B mutations of Lafora disease.


Subject(s)
Lafora Disease , Humans , Mice , Animals , Lafora Disease/genetics , Lafora Disease/metabolism , Glycogen Synthase/genetics , Disease Models, Animal , Mutation , Oligonucleotides, Antisense/therapeutic use , Glycogen/metabolism , Ubiquitin-Protein Ligases/genetics
5.
Int J Mol Sci ; 24(7)2023 Mar 23.
Article in English | MEDLINE | ID: mdl-37046993

ABSTRACT

Lafora disease (LD) is a neurological disorder characterized by progressive myoclonus epilepsy. The hallmark of the disease is the presence of insoluble forms of glycogen (polyglucosan bodies, or PGBs) in the brain. The accumulation of PGBs is causative of the pathophysiological features of LD. However, despite the efforts made by different groups, the question of why PGBs accumulate in the brain is still unanswered. We have recently demonstrated that, in vivo, astrocytes accumulate most of the PGBs present in the brain, and this could lead to astrocyte dysfunction. To develop a deeper understanding of the defects present in LD astrocytes that lead to LD pathophysiology, we obtained pure primary cultures of astrocytes from LD mice from the postnatal stage under conditions that accumulate PGBs, the hallmark of LD. These cells serve as novel in vitro models for studying PGBs accumulation and related LD dysfunctions. In this sense, the metabolomics of LD astrocytes indicate that they accumulate metabolic intermediates of the upper part of the glycolytic pathway, probably as a consequence of enhanced glucose uptake. In addition, we also demonstrate the feasibility of using the model in the identification of different compounds that may reduce the accumulation of polyglucosan inclusions.


Subject(s)
Lafora Disease , Mice , Animals , Lafora Disease/metabolism , Astrocytes/metabolism , Disease Models, Animal , Glucans/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
6.
Neurobiol Dis ; 177: 105998, 2023 02.
Article in English | MEDLINE | ID: mdl-36638890

ABSTRACT

Laforin and Malin are two proteins that are encoded by the genes EPM2A and EPM2B, respectively. Laforin is a glucan phosphatase and Malin is an E3-ubiquitin ligase, and these two proteins function as a complex. Mutations occurring at the level of one of the two genes lead to the accumulation of an aberrant form of glycogen meant to cluster in polyglucosans that go under the name of Lafora bodies. Individuals affected by the appearance of these polyglucosans, especially at the cerebral level, experience progressive neurodegeneration and several episodes of epilepsy leading to the manifestation of a fatal form of a rare disease called Lafora disease (LD), for which, to date, no treatment is available. Despite the different dysfunctions described for this disease, many molecular aspects still demand elucidation. An effective way to unknot some of the nodes that prevent the achievement of better knowledge of LD is to focus on the substrates that are ubiquitinated by the E3-ubiquitin ligase Malin. Some substrates have already been provided by previous studies based on protein-protein interaction techniques and have been associated with some alterations that mark the disease. In this work, we have used an unbiased alternative approach based on the activity of Malin as an E3-ubiquitin ligase. We report the discovery of novel bonafide substrates of Malin and have characterized one of them more deeply, namely PIP3-dependent Rac exchanger 1 (P-Rex1). The analysis conducted upon this substrate sets the genesis of the delineation of a molecular pathway that leads to altered glucose uptake, which could be one of the origin of the accumulation of the polyglucosans present in the disease.


Subject(s)
Lafora Disease , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Lafora Disease/genetics , Lafora Disease/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Glycogen , Ubiquitins
7.
Int J Mol Sci ; 24(2)2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36674605

ABSTRACT

Lafora disease is a rare, fatal form of progressive myoclonus epilepsy characterized by continuous neurodegeneration with epileptic seizures, characterized by the intracellular accumulation of aberrant polyglucosan granules called Lafora bodies. Several works have provided numerous evidence of molecular and cellular alterations in neural tissue from experimental mouse models deficient in either laforin or malin, two proteins related to the disease. Oxidative stress, alterations in proteostasis, and deregulation of inflammatory signals are some of the molecular alterations underlying this condition in both KO animal models. Lafora bodies appear early in the animal's life, but many of the aforementioned molecular aberrant processes and the consequent neurological symptoms ensue only as animals age. Here, using small RNA-seq and quantitative PCR on brain extracts from laforin and malin KO male mice of different ages, we show that two different microRNA species, miR-155 and miR-146a, are overexpressed in an age-dependent manner. We also observed altered expression of putative target genes for each of the microRNAs studied in brain extracts. These results open the path for a detailed dissection of the molecular consequences of laforin and malin deficiency in brain tissue, as well as the potential role of miR-155 and miR-146a as specific biomarkers of disease progression in LD.


Subject(s)
Lafora Disease , MicroRNAs , Mice , Male , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Lafora Disease/genetics , Lafora Disease/metabolism , Neuroinflammatory Diseases , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Oxidative Stress/genetics , Ubiquitin-Protein Ligases/metabolism
8.
Dis Model Mech ; 16(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36511140

ABSTRACT

Glycogen is the largest cytosolic macromolecule and is kept in solution through a regular system of short branches allowing hydration. This structure was thought to solely require balanced glycogen synthase and branching enzyme activities. Deposition of overlong branched glycogen in the fatal epilepsy Lafora disease (LD) indicated involvement of the LD gene products laforin and the E3 ubiquitin ligase malin in regulating glycogen structure. Laforin binds glycogen, and LD-causing mutations disrupt this binding, laforin-malin interactions and malin's ligase activity, all indicating a critical role for malin. Neither malin's endogenous function nor location had previously been studied due to lack of suitable antibodies. Here, we generated a mouse in which the native malin gene is tagged with the FLAG sequence. We show that the tagged gene expresses physiologically, malin localizes to glycogen, laforin and malin indeed interact, at glycogen, and malin's presence at glycogen depends on laforin. These results, and mice, open the way to understanding unknown mechanisms of glycogen synthesis critical to LD and potentially other much more common diseases due to incompletely understood defects in glycogen metabolism.


Subject(s)
Glycogen , Lafora Disease , Protein Tyrosine Phosphatases, Non-Receptor , Ubiquitin-Protein Ligases , Animals , Mice , Glycogen/metabolism , Lafora Disease/genetics , Lafora Disease/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
9.
Int J Mol Sci ; 23(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35743315

ABSTRACT

Mutations in the EPM2A gene encoding laforin cause Lafora disease (LD), a progressive myoclonic epilepsy characterized by drug-resistant seizures and progressive neurological impairment. To date, rodents are the only available models for studying LD; however, their use for drug screening is limited by regulatory restrictions and high breeding costs. To investigate the role of laforin loss of function in early neurodevelopment, and to screen for possible new compounds for treating the disorder, we developed a zebrafish model of LD. Our results showed the epm2a-/- zebrafish to be a faithful model of LD, exhibiting the main disease features, namely motor impairment and neuronal hyperexcitability with spontaneous seizures. The model also showed increased inflammatory response and apoptotic death, as well as an altered autophagy pathway that occurs early in development and likely contributes to the disease progression. Early administration of trehalose was found to be effective for rescuing motor impairment and neuronal hyperexcitability associated with seizures. Our study adds a new tool for investigating LD and might help to identify new treatment opportunities.


Subject(s)
Lafora Disease , Animals , Lafora Disease/drug therapy , Lafora Disease/genetics , Lafora Disease/metabolism , Mutation , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Seizures , Trehalose/pharmacology , Ubiquitin-Protein Ligases/genetics , Zebrafish/metabolism
10.
Cell Metab ; 33(7): 1404-1417.e9, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34043942

ABSTRACT

Glycosylation defects are a hallmark of many nervous system diseases. However, the molecular and metabolic basis for this pathology is not fully understood. In this study, we found that N-linked protein glycosylation in the brain is metabolically channeled to glucosamine metabolism through glycogenolysis. We discovered that glucosamine is an abundant constituent of brain glycogen, which functions as a glucosamine reservoir for multiple glycoconjugates. We demonstrated the enzymatic incorporation of glucosamine into glycogen by glycogen synthase, and the release by glycogen phosphorylase by biochemical and structural methodologies, in primary astrocytes, and in vivo by isotopic tracing and mass spectrometry. Using two mouse models of glycogen storage diseases, we showed that disruption of brain glycogen metabolism causes global decreases in free pools of UDP-N-acetylglucosamine and N-linked protein glycosylation. These findings revealed fundamental biological roles of brain glycogen in protein glycosylation with direct relevance to multiple human diseases of the central nervous system.


Subject(s)
Brain/metabolism , Glucosamine/metabolism , Glycogen/physiology , Protein Processing, Post-Translational , Animals , Cells, Cultured , Disease Models, Animal , Female , Glycogen/metabolism , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Glycogenolysis/genetics , Glycosylation , Lafora Disease/genetics , Lafora Disease/metabolism , Lafora Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Processing, Post-Translational/genetics
11.
Neurotherapeutics ; 18(2): 1414-1425, 2021 04.
Article in English | MEDLINE | ID: mdl-33830476

ABSTRACT

Many adult and most childhood neurological diseases have a genetic basis. CRISPR/Cas9 biotechnology holds great promise in neurological therapy, pending the clearance of major delivery, efficiency, and specificity hurdles. We applied CRISPR/Cas9 genome editing in its simplest modality, namely inducing gene sequence disruption, to one adult and one pediatric disease. Adult polyglucosan body disease is a neurodegenerative disease resembling amyotrophic lateral sclerosis. Lafora disease is a severe late childhood onset progressive myoclonus epilepsy. The pathogenic insult in both is formation in the brain of glycogen with overlong branches, which precipitates and accumulates into polyglucosan bodies that drive neuroinflammation and neurodegeneration. We packaged Staphylococcus aureus Cas9 and a guide RNA targeting the glycogen synthase gene, Gys1, responsible for brain glycogen branch elongation in AAV9 virus, which we delivered by neonatal intracerebroventricular injection to one mouse model of adult polyglucosan body disease and two mouse models of Lafora disease. This resulted, in all three models, in editing of approximately 17% of Gys1 alleles and a similar extent of reduction of Gys1 mRNA across the brain. The latter led to approximately 50% reductions of GYS1 protein, abnormal glycogen accumulation, and polyglucosan bodies, as well as ameliorations of neuroinflammatory markers in all three models. Our work represents proof of principle for virally delivered CRISPR/Cas9 neurotherapeutics in an adult-onset (adult polyglucosan body) and a childhood-onset (Lafora) neurological diseases.


Subject(s)
Brain/metabolism , Glucans/metabolism , Glycogen Storage Disease/genetics , Glycogen Synthase/genetics , Glycogen/metabolism , Lafora Disease/genetics , Nervous System Diseases/genetics , Neuroinflammatory Diseases/genetics , RNA, Messenger/metabolism , Animals , CRISPR-Cas Systems , Disease Models, Animal , Gene Editing , Glycogen Storage Disease/metabolism , Glycogen Storage Disease/therapy , Lafora Disease/metabolism , Lafora Disease/therapy , Mice , Nervous System Diseases/metabolism , Nervous System Diseases/therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/therapy , Proof of Concept Study
12.
Brain ; 144(8): 2349-2360, 2021 09 04.
Article in English | MEDLINE | ID: mdl-33822008

ABSTRACT

The hallmark of Lafora disease, a fatal neurodegenerative disorder, is the accumulation of intracellular glycogen aggregates called Lafora bodies. Until recently, it was widely believed that brain Lafora bodies were present exclusively in neurons and thus that Lafora disease pathology derived from their accumulation in this cell population. However, recent evidence indicates that Lafora bodies are also present in astrocytes. To define the role of astrocytic Lafora bodies in Lafora disease pathology, we deleted glycogen synthase specifically from astrocytes in a mouse model of the disease (malinKO). Strikingly, blocking glycogen synthesis in astrocytes-thus impeding Lafora bodies accumulation in this cell type-prevented the increase in neurodegeneration markers, autophagy impairment, and metabolic changes characteristic of the malinKO model. Conversely, mice that over-accumulate glycogen in astrocytes showed an increase in these markers. These results unveil the deleterious consequences of the deregulation of glycogen metabolism in astrocytes and change the perspective that Lafora disease is caused solely by alterations in neurons.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Glycogen/metabolism , Lafora Disease/metabolism , Nerve Degeneration/metabolism , Animals , Astrocytes/pathology , Brain/pathology , Disease Models, Animal , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Lafora Disease/genetics , Lafora Disease/pathology , Mice , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
13.
Exp Neurol ; 340: 113656, 2021 06.
Article in English | MEDLINE | ID: mdl-33639210

ABSTRACT

Heat shock response (HSR) is a conserved cytoprotective pathway controlled by the master transcriptional regulator, the heat shock factor 1 (HSF1), that activates the expression of heat shock proteins (HSPs). HSPs, as chaperones, play essential roles in minimizing stress-induced damages and restoring proteostasis. Therefore, compromised HSR is thought to contribute to neurodegenerative disorders. Lafora disease (LD) is a fatal form of neurodegenerative disorder characterized by the accumulation of abnormal glycogen as Lafora bodies in neurons and other tissues. The symptoms of LD include progressive myoclonus epilepsy, dementia, and cognitive deficits. LD is caused by the defects in the gene coding laforin phosphatase or the malin ubiquitin ligase. Laforin and malin are known to work upstream of HSF1 and are essential for the activation of HSR. Herein, we show that mice deficient for laforin or malin show reduced levels of HSF1 and their targets in their brain tissues, suggesting compromised HSR; this could contribute to the neuropathology in LD. Intriguingly, treatment of LD animals with dexamethasone, a synthetic glucocorticoid analogue, partially restored the levels of HSF1 and its targets. Dexamethasone treatment was also able to ameliorate the neuroinflammation and susceptibility to induced seizures in the LD animals. However, dexamethasone treatment did not show a significant effect on Lafora bodies or autophagy defects. Taken together, the present study establishes a role for HSR in seizure susceptibility and neuroinflammation and dexamethasone as a potential antiepileptic agent, suitable for further studies in LD.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Dexamethasone/therapeutic use , Heat Shock Transcription Factors/metabolism , Heat-Shock Response/drug effects , Lafora Disease/metabolism , Seizures/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Disease Models, Animal , Heat Shock Transcription Factors/deficiency , Heat Shock Transcription Factors/genetics , Heat-Shock Response/physiology , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Lafora Disease/drug therapy , Mice , Mice, Knockout , Seizures/drug therapy
14.
J Comp Neurol ; 529(6): 1099-1120, 2021 04 15.
Article in English | MEDLINE | ID: mdl-32785985

ABSTRACT

Lafora disease (LD) is a genetic and fatal form of neurodegenerative disorder characterized by myoclonic epilepsy and cognitive deficits. LD is caused by loss-of-function mutations in the EPM2A or the NHLRC1 gene. A major hallmark of LD is the presence of abnormal glycogen aggregates in neurons and other tissues. Functional studies on the genes have, therefore, mostly focused on glycogen metabolism. The physiological basis of cognitive deficits in LD is thus largely unexplored. Alterations in dendritic spine morphology are known in neurodevelopmental and neuropsychiatric disorders. We, therefore, analyzed the dendritic spine morphologies in pyramidal neurons of the hippocampal and Cortical layer V of the Epm2a or Nhlrc1 knockout mice brain. We found a significant increase in the density, length, and reduction in the width of the dendritic spines in Postnatal day 21 to 12-month-old LD animals. Similar observations were made in the primary cultures of neurons derived from the hippocampi of the embryonic brain, suggesting that the aberrant spine phenotype could be a developmental defect in LD. We also looked at the cognitive and behavioral deficits as a possible readout of the spine abnormalities. The LD animals exhibited hyperactivity, reduced anxiety-like behavior, and deficits in the spatial and nonspatial memory. Such abnormalities were seen in the younger (1-2 months) as well as the older (7-8 months) age groups. Taken together, our results suggest that the dendritic spine abnormalities are primary developmental defects in the LD model and these defects might underlie some of the symptoms, including cognitive deficits, in LD.


Subject(s)
Cerebral Cortex/pathology , Cognitive Dysfunction/pathology , Dendritic Spines/pathology , Hippocampus/pathology , Lafora Disease/pathology , Memory/physiology , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Dendritic Spines/genetics , Dendritic Spines/metabolism , Female , Hippocampus/metabolism , Lafora Disease/genetics , Lafora Disease/metabolism , Male , Mice , Mice, Knockout , Pregnancy , Protein Tyrosine Phosphatases, Non-Receptor/deficiency , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
15.
J Neurochem ; 157(6): 1897-1910, 2021 06.
Article in English | MEDLINE | ID: mdl-32892347

ABSTRACT

Mammalian glycogen chain lengths are subject to complex regulation, including by seven proteins (protein phosphatase-1 regulatory subunit 3, PPP1R3A through PPP1R3G) that target protein phosphatase-1 (PP1) to glycogen to activate the glycogen chain-elongating enzyme glycogen synthase and inactivate the chain-shortening glycogen phosphorylase. Lafora disease is a fatal neurodegenerative epilepsy caused by aggregates of long-chained, and as a result insoluble, glycogen, termed Lafora bodies (LBs). We previously eliminated PPP1R3C from a Lafora disease mouse model and studied the effect on LB formation. In the present work, we eliminate and study the effect of absent PPP1R3D. In the interim, brain cell type levels of all PPP1R3 genes have been published, and brain cell type localization of LBs clarified. Integrating these data we find that PPP1R3C is the major isoform in most tissues including brain. In the brain, PPP1R3C is expressed at 15-fold higher levels than PPP1R3D in astrocytes, the cell type where most LBs form. PPP1R3C deficiency eliminates ~90% of brain LBs. PPP1R3D is quantitatively a minor isoform, but possesses unique MAPK, CaMK2 and 14-3-3 binding domains and appears to have an important functional niche in murine neurons and cardiomyocytes. In neurons, it is expressed equally to PPP1R3C, and its deficiency eliminates ~50% of neuronal LBs. In heart, it is expressed at 25% of PPP1R3C where its deficiency eliminates ~90% of LBs. This work studies the role of a second (PPP1R3D) of seven PP1 subunits that regulate the structure of glycogen, toward better understanding of brain glycogen metabolism generally, and in Lafora disease.


Subject(s)
Disease Models, Animal , Lafora Disease/metabolism , Myocardium/metabolism , Neurons/metabolism , Protein Phosphatase 1/deficiency , Animals , Brain/metabolism , Brain/pathology , Female , Glycogen/metabolism , Humans , Lafora Disease/genetics , Lafora Disease/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Neurons/pathology , Protein Phosphatase 1/genetics
16.
J Biol Chem ; 296: 100150, 2021.
Article in English | MEDLINE | ID: mdl-33277363

ABSTRACT

Malstructured glycogen accumulates over time in Lafora disease (LD) and precipitates into Lafora bodies (LBs), leading to neurodegeneration and intractable fatal epilepsy. Constitutive reduction of glycogen synthase-1 (GYS1) activity prevents murine LD, but the effect of GYS1 reduction later in disease course is unknown. Our goal was to knock out Gys1 in laforin (Epm2a)-deficient LD mice after disease onset to determine whether LD can be halted in midcourse, or even reversed. We generated Epm2a-deficient LD mice with tamoxifen-inducible Cre-mediated Gys1 knockout. Tamoxifen was administered at 4 months and disease progression assessed at 12 months. We verified successful knockout at mRNA and protein levels using droplet digital PCR and Western blots. Glycogen determination and periodic acid-Schiff-diastase staining were used to analyze glycogen and LB accumulation. Immunohistochemistry using astrocytic (glial fibrillary acidic protein) and microglial (ionized calcium-binding adapter molecule 1) markers was performed to investigate neuroinflammation. In the disease-relevant organ, the brain, Gys1 mRNA levels were reduced by 85% and GYS1 protein depleted. Glycogen accumulation was halted at the 4-month level, while LB formation and neuroinflammation were significantly, though incompletely, prevented. Skeletal muscle analysis confirmed that Gys1 knockout inhibits glycogen and LB accumulation. However, tamoxifen-independent Cre recombination precluded determination of disease halting or reversal in this tissue. Our study shows that Gys1 knockdown is a powerful means to prevent LD progression, but this approach did not reduce brain glycogen or LBs to levels below those at the time of intervention. These data suggest that endogenous mechanisms to clear brain LBs are absent or, possibly, compromised in laforin-deficient murine LD.


Subject(s)
Gliosis/prevention & control , Glycogen Synthase/physiology , Inflammation/prevention & control , Lafora Disease/pathology , Muscle, Skeletal/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/deficiency , Animals , Female , Gliosis/metabolism , Gliosis/pathology , Inflammation/metabolism , Inflammation/pathology , Lafora Disease/drug therapy , Lafora Disease/genetics , Lafora Disease/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Selective Estrogen Receptor Modulators/administration & dosage , Tamoxifen/administration & dosage
17.
Int J Mol Sci ; 21(20)2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33092303

ABSTRACT

Mutations in the EPM2A and EPM2B genes, encoding laforin and malin proteins respectively, are responsible for Lafora disease, a fatal form of progressive myoclonus epilepsy with autosomal recessive inheritance. Neuroimaging studies of patients with Lafora disease have shown different degrees of brain atrophy, decreased glucose brain uptake and alterations on different brain metabolites mainly in the frontal cortex, basal ganglia and cerebellum. Mice deficient for laforin and malin present many features similar to those observed in patients, including cognitive, motor, histological and epileptic hallmarks. We describe the neuroimaging features found in two mouse models of Lafora disease. We found altered volumetric values in the cerebral cortex, hippocampus, basal ganglia and cerebellum using magnetic resonance imaging (MRI). Positron emission tomography (PET) of the cerebral cortex, hippocampus and cerebellum of Epm2a-/- mice revealed abnormal glucose uptake, although no alterations in Epm2b-/- mice were observed. Magnetic resonance spectroscopy (MRS) revealed significant changes in the concentration of several brain metabolites, including N-acetylaspartate (NAA), in agreement with previously described findings in patients. These data may provide new insights into disease mechanisms that may be of value for developing new biomarkers for diagnosis, prevention and treatment of Lafora disease using animal models.


Subject(s)
Brain Diseases/metabolism , Brain/abnormalities , Disease Models, Animal , Lafora Disease/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Atrophy , Basal Ganglia/diagnostic imaging , Basal Ganglia/metabolism , Basal Ganglia/pathology , Brain/diagnostic imaging , Brain/metabolism , Brain Diseases/genetics , Brain Diseases/pathology , Cerebellum/diagnostic imaging , Cerebellum/metabolism , Cerebellum/pathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Glucose/metabolism , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Hippocampus/pathology , Humans , Lafora Disease/genetics , Lafora Disease/pathology , Magnetic Resonance Imaging/methods , Mice, Knockout , Mutation , Positron-Emission Tomography/methods , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Ubiquitin-Protein Ligases/genetics
18.
J Vet Intern Med ; 34(4): 1594-1598, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32418279

ABSTRACT

Cortical atrophy has been identified using magnetic resonance imaging (MRI) in humans and dogs with Lafora disease (LD). In humans, proton magnetic resonance spectroscopy (1HMRS) of the brain indicates decreased N-acetyl-aspartate (NAA) relative to other brain metabolites. Brain 1HMRS findings in dogs with LD are lacking. A 6-year-old female Beagle was presented with a history of a single generalized tonic-clonic seizure and episodic reflex myoclonus. Clinical, hematological, and neurological examination findings and 3-Tesla MRI of the brain were unremarkable. Brain 1HMRS with voxel positioning in the thalamus was performed in the affected Beagle. It identified decreased amounts of NAA, glutamate-glutamine complex, and increased total choline and phosphoethanolamine relative to water and total creatine compared with the reference range in healthy control Beagles. A subsequent genetic test confirmed LD. Abnormalities in 1HMRS despite lack of changes with conventional MRI were identified in a dog with LD.


Subject(s)
Dog Diseases/diagnosis , Lafora Disease/veterinary , Proton Magnetic Resonance Spectroscopy , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Brain/metabolism , Choline/metabolism , Dog Diseases/genetics , Dog Diseases/metabolism , Dogs , Ethanolamines/metabolism , Female , Glutamic Acid/metabolism , Glutamine/metabolism , Lafora Disease/diagnosis , Lafora Disease/genetics , Lafora Disease/metabolism , Magnetic Resonance Imaging/veterinary
19.
Mol Neurobiol ; 57(3): 1607-1621, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31808062

ABSTRACT

Lafora disease (LD) is a rare, fatal form of progressive myoclonus epilepsy. The molecular basis of this devastating disease is still poorly understood, and no treatment is available yet, which leads to the death of the patients around 10 years from the onset of the first symptoms. The hallmark of LD is the accumulation of insoluble glycogen-like inclusions in the brain and peripheral tissues, as a consequence of altered glycogen homeostasis. In addition, other determinants in the pathophysiology of LD have been suggested, such as proteostasis impairment, with reduction in autophagy, and oxidative stress, among others. In order to gain a general view of the genes involved in the pathophysiology of LD, in this work, we have performed RNA-Seq transcriptome analyses of whole-brain tissue from two independent mouse models of the disease, namely Epm2a-/- and Epm2b-/- mice, at different times of age. Our results provide strong evidence for three major facts: first, in both models of LD, we found a common set of upregulated genes, most of them encoding mediators of inflammatory response; second, there was a progression with the age in the appearance of these inflammatory markers, starting at 3 months of age; and third, reactive glia was responsible for the expression of these inflammatory genes. These results clearly indicate that neuroinflammation is one of the most important traits to be considered in order to fully understand the pathophysiology of LD, and define reactive glia as novel therapeutic targets in the disease.


Subject(s)
Age Factors , Lafora Disease/metabolism , Myoclonic Epilepsies, Progressive/metabolism , Neuroglia/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Animals , Disease Models, Animal , Disease Progression , Glycogen/metabolism , Inclusion Bodies/metabolism , Lafora Disease/genetics , Mice, Knockout , Myoclonic Epilepsies, Progressive/genetics , Oxidative Stress/physiology
20.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118613, 2020 02.
Article in English | MEDLINE | ID: mdl-31758957

ABSTRACT

Lafora progressive myoclonus epilepsy is a fatal rare neurodegenerative disorder characterized by the accumulation of insoluble abnormal glycogen deposits in the brain and peripheral tissues. Mutations in at least two genes are responsible for the disease: EPM2A, encoding the glucan phosphatase laforin, and EPM2B, encoding the RING-type E3-ubiquitin ligase malin. Both laforin and malin form a functional complex in which laforin recruits the substrates to be ubiquitinated by malin. We and others have described that, in cellular and animal models of this disease, there is an autophagy impairment which leads to the accumulation of dysfunctional mitochondria. In addition, we established that the autophagic defect occurred at the initial steps of autophagosome formation. In this work, we present evidence that in cellular models of the disease there is a decrease in the amount of phosphatidylinositol-3P. This is probably due to defective regulation of the autophagic PI3KC3 complex, in the absence of a functional laforin/malin complex. In fact, we demonstrate that the laforin/malin complex interacts physically and co-localizes intracellularly with core components of the PI3KC3 complex (Beclin1, Vps34 and Vps15), and that this interaction is specific and results in the polyubiquitination of these proteins. In addition, the laforin/malin complex is also able to polyubiquitinate ATG14L and UVRAG. Finally, we show that overexpression of the laforin/malin complex increases PI3KC3 activity. All these results suggest a new role of the laforin/malin complex in the activation of autophagy via regulation of the PI3KC3 complex and explain the defect in autophagy described in Lafora disease.


Subject(s)
Lafora Disease/pathology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Autophagy , Autophagy-Related Proteins/metabolism , Beclin-1/chemistry , Beclin-1/metabolism , Cells, Cultured , Humans , Lafora Disease/metabolism , Microscopy, Fluorescence , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Transcription Factors/chemistry , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL