Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
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
2.
Epilepsy Res ; 200: 107317, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38341935

ABSTRACT

Lafora disease is a rare and fatal form of progressive myoclonic epilepsy with onset during early adolescence. The disease is caused by mutations in EPM2A, encoding laforin, or EPM2B, encoding malin. Both proteins have functions that affect glycogen metabolism, including glycogen dephosphorylation by laforin and ubiquitination of enzymes involved in glycogen metabolism by malin. Lack of function of laforin or malin results in the accumulation of polyglucosan that forms Lafora bodies in the central nervous system and other tissues. Enzyme replacement therapy through intravenous administration of alglucosidase alfa (Myozyme®) has shown beneficial effects removing polyglucosan aggregates in Pompe disease. We evaluated the effectiveness of intracerebroventricular administration of alglucosidase alfa in the Epm2a-/- knock-out and Epm2aR240X knock-in mouse models of Lafora disease. Seven days after a single intracerebroventricular injection of alglucosidase alfa in 12-month-old Epm2a-/- and Epm2aR240X mice, the number of Lafora bodies was not reduced. Additionally, a prolonged infusion of alglucosidase alfa for 2 or 4 weeks in 6- and 9-month-old Epm2a-/- mice did not result in a reduction in the number of LBs or the amount of glycogen in the brain. These findings hold particular significance in guiding a rational approach to the utilization of novel therapies in Lafora disease.


Subject(s)
Lafora Disease , alpha-Glucosidases , Mice , Animals , Lafora Disease/drug therapy , Lafora Disease/genetics , Mice, Knockout , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Glycogen/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/genetics
3.
Neurobiol Dis ; 181: 106119, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37059210

ABSTRACT

Lafora disease is a rare recessive form of progressive myoclonic epilepsy, usually diagnosed during adolescence. Patients present with myoclonus, neurological deterioration, and generalized tonic-clonic, myoclonic, or absence seizures. Symptoms worsen until death, usually within the first ten years of clinical onset. The primary histopathological hallmark is the formation of aberrant polyglucosan aggregates called Lafora bodies in the brain and other tissues. Lafora disease is caused by mutations in either the EPM2A gene, encoding laforin, or the EPM2B gene, coding for malin. The most frequent EPM2A mutation is R241X, which is also the most prevalent in Spain. The Epm2a-/- and Epm2b-/- mouse models of Lafora disease show neuropathological and behavioral abnormalities similar to those seen in patients, although with a milder phenotype. To obtain a more accurate animal model, we generated the Epm2aR240X knock-in mouse line with the R240X mutation in the Epm2a gene, using genetic engineering based on CRISPR-Cas9 technology. Epm2aR240X mice exhibit most of the alterations reported in patients, including the presence of LBs, neurodegeneration, neuroinflammation, interictal spikes, neuronal hyperexcitability, and cognitive decline, despite the absence of motor impairments. The Epm2aR240X knock-in mouse displays some symptoms that are more severe that those observed in the Epm2a-/- knock-out, including earlier and more pronounced memory loss, increased levels of neuroinflammation, more interictal spikes and increased neuronal hyperexcitability, symptoms that more precisely resemble those observed in patients. This new mouse model can therefore be specifically used to evaluate how new therapies affects these features with greater precision.


Subject(s)
Cognitive Dysfunction , Lafora Disease , Animals , Mice , Cognitive Dysfunction/genetics , Lafora Disease/genetics , Lafora Disease/pathology , Mice, Knockout , Neuroinflammatory Diseases , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Ubiquitin-Protein Ligases/genetics
4.
Neurotherapeutics ; 20(1): 230-244, 2023 01.
Article in English | MEDLINE | ID: mdl-36303102

ABSTRACT

Lafora disease is a fatal form of progressive myoclonic epilepsy caused by mutations in the EPM2A or NHLRC1/EPM2B genes that usually appears during adolescence. The Epm2a-/- and Epm2b-/- knock-out mouse models of the disease develop behavioral and neurological alterations similar to those observed in patients. The aim of this work is to analyze whether early treatment with metformin (from conception to adulthood) ameliorates the formation of Lafora bodies and improves the behavioral and neurological outcomes observed with late treatment (during 2 months at 10 months of age). We also evaluated the benefits of metformin in patients with Lafora disease. To assess neurological improvements due to metformin administration in the two mouse models, we evaluated the effects on pentylenetetrazol sensitivity, posturing, motor coordination and activity, and memory. We also analyzed the effects on Lafora bodies, neurodegeneration, and astrogliosis. Furthermore, we conducted a follow-up study of an initial cohort of 18 patients with Lafora disease, 8 treated with metformin and 10 untreated. Our results indicate that early metformin was more effective than late metformin in Lafora disease mouse models improving neurological alterations of both models such as neuronal hyperexcitability, motor and memory alterations, neurodegeneration, and astrogliosis and decreasing the formation of Lafora bodies. Moreover, patients receiving metformin had a slower progression of the disease. Overall, early treatment improves the outcome seen with late metformin treatment in the two knock-out mouse models of Lafora disease. Metformin-treated patients exhibited an ameliorated course of the disease with slower deterioration of their daily living activities.


Subject(s)
Lafora Disease , Metformin , Animals , Mice , Lafora Disease/drug therapy , Lafora Disease/genetics , Metformin/therapeutic use , Gliosis , Follow-Up Studies , Ubiquitin-Protein Ligases/genetics
5.
bioRxiv ; 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38168354

ABSTRACT

Lafora disease is a rare and fatal form of progressive myoclonic epilepsy typically occurring early in adolescence. Common symptoms include seizures, dementia, and a progressive neurological decline leading to death within 5-15 years from onset. The disease results from mutations transmitted with autosomal recessive inheritance in the EPM2A gene, encoding laforin, a dual-specificity phosphatase, or the EPM2B gene, encoding malin, an E3-ubiquitin ligase. Laforin has glucan phosphatase activity, is an adapter of enzymes involved in glycogen metabolism, is involved in endoplasmic reticulum-stress and protein clearance, and acts as a tumor suppressor protein. 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 can lead to alterations in this complex, leading to the formation of Lafora bodies that contain abnormal, insoluble, and hyperphosphorylated forms of glycogen called polyglucosans. We used the Epm2a -/- knock-out mouse model of Lafora disease to apply a gene replacement therapy by administering intracerebroventricular injections of a recombinant adeno-associated virus carrying the human EPM2A gene. We evaluated the effects of this treatment by means of neuropathological studies, behavioral tests, video-electroencephalography recording, and proteomic/phosphoproteomic analysis. Gene therapy with recombinant adeno-associated virus containing the EPM2A gene ameliorated neurological and histopathological alterations, reduced epileptic activity and neuronal hyperexcitability, and decreased the formation of Lafora bodies. Differential quantitative proteomics and phosphoproteomics revealed beneficial changes in various molecular pathways altered in Lafora disease. Improvements were observed for up to nine months following a single intracerebroventricular injection. In conclusion, gene replacement therapy with human EPM2A gene in the Epm2a -/- knock-out mice shows promise as a potential treatment for Lafora disease.

6.
Int J Mol Sci ; 22(10)2021 May 19.
Article in English | MEDLINE | ID: mdl-34069559

ABSTRACT

Metformin is a drug in the family of biguanide compounds that is widely used in the treatment of type 2 diabetes (T2D). Interestingly, the therapeutic potential of metformin expands its prescribed use as an anti-diabetic drug. In this sense, it has been described that metformin administration has beneficial effects on different neurological conditions. In this work, we review the beneficial effects of this drug as a neuroprotective agent in different neurological diseases, with a special focus on epileptic disorders and Lafora disease, a particular type of progressive myoclonus epilepsy. In addition, we review the different proposed mechanisms of action of metformin to understand its function at the neurological level.


Subject(s)
Central Nervous System/drug effects , Metformin/therapeutic use , Animals , Central Nervous System/metabolism , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Epilepsy/drug therapy , Humans , Hypoglycemic Agents/pharmacology , Lafora Disease/drug therapy , Metformin/metabolism , Metformin/pharmacology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology
7.
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
8.
Neurobiol Dis ; 127: 210-222, 2019 07.
Article in English | MEDLINE | ID: mdl-30831192

ABSTRACT

Autism spectrum disorders are early onset neurodevelopmental disorders characterized by deficits in social communication and restricted repetitive behaviors, yet they are quite heterogeneous in terms of their genetic basis and phenotypic manifestations. Recently, de novo pathogenic mutations in DYRK1A, a chromosome 21 gene associated to neuropathological traits of Down syndrome, have been identified in patients presenting a recognizable syndrome included in the autism spectrum. These mutations produce DYRK1A kinases with partial or complete absence of the catalytic domain, or they represent missense mutations located within this domain. Here, we undertook an extensive biochemical characterization of the DYRK1A missense mutations reported to date and show that most of them, but not all, result in enzymatically dead DYRK1A proteins. We also show that haploinsufficient Dyrk1a+/- mutant mice mirror the neurological traits associated with the human pathology, such as defective social interactions, stereotypic behaviors and epileptic activity. These mutant mice present altered proportions of excitatory and inhibitory neocortical neurons and synapses. Moreover, we provide evidence that alterations in the production of cortical excitatory neurons are contributing to these defects. Indeed, by the end of the neurogenic period, the expression of developmental regulated genes involved in neuron differentiation and/or activity is altered. Therefore, our data indicate that altered neocortical neurogenesis could critically affect the formation of cortical circuits, thereby contributing to the neuropathological changes in DYRK1A haploinsufficiency syndrome.


Subject(s)
Autistic Disorder/metabolism , Haploinsufficiency , Neocortex/metabolism , Nerve Net/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Social Behavior , Animals , Autistic Disorder/genetics , Behavior, Animal/physiology , Male , Mice , Mutation, Missense , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Dyrk Kinases
9.
Int J Mol Sci ; 19(4)2018 Apr 05.
Article in English | MEDLINE | ID: mdl-29621183

ABSTRACT

Patients with dementia present epilepsy more frequently than the general population. Seizures are more common in patients with Alzheimer's disease (AD), dementia with Lewy bodies (LBD), frontotemporal dementia (FTD) and progressive supranuclear palsy (PSP) than in other dementias. Missense mutations in the microtubule associated protein tau (MAPT) gene have been found to cause familial FTD and PSP, while the P301S mutation in MAPT has been associated with early-onset fast progressive dementia and the presence of seizures. Brains of patients with AD, LBD, FTD and PSP show hyperphosphorylated tau aggregates, amyloid-ß plaques and neuropil threads. Increasing evidence suggests the existence of overlapping mechanisms related to the generation of network hyperexcitability and cognitive decline. Neuronal overexpression of tau with various mutations found in FTD with parkinsonism-linked to chromosome 17 (FTDP-17) in mice produces epileptic activity. On the other hand, the use of certain antiepileptic drugs in animal models with AD prevents cognitive impairment. Further efforts should be made to search for plausible common targets for both conditions. Moreover, attempts should also be made to evaluate the use of drugs targeting tau and amyloid-ß as suitable pharmacological interventions in epileptic disorders. The diagnosis of dementia and epilepsy in early stages of those diseases may be helpful for the initiation of treatments that could prevent the generation of epileptic activity and cognitive deterioration.


Subject(s)
Dementia/metabolism , Epilepsy/metabolism , Animals , Brain/metabolism , Brain/pathology , Dementia/pathology , Epilepsy/pathology , Humans , tau Proteins/metabolism
11.
Neuroreport ; 28(5): 268-271, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28181916

ABSTRACT

Lafora disease (LD) is a rare adolescent-onset progressive myoclonic epilepsy caused by loss-of-function mutations either in the EPM2A gene encoding laforin or in the EPM2B gene encoding malin. Mouse models with deletion in the Epm2a or the Epm2b gene show intracellular aggregates of polyglucosans (Lafora bodies) and neurological complications that resemble those observed in patients with LD. In the absence of laforin or malin expression, mice also show different degrees of hyperexcitability, as reflected by an enhanced response to the convulsant drug pentylenetetrazol (PTZ). Malin knockout mice treated with 4-phenylbutyric acid (4-PBA) and metformin showed decreased amounts of Lafora bodies and polyubiquitin protein aggregates in the brain, diminished neurodegeneration, and amelioration of some neurological conditions. In this study, we analyzed the action of 4-PBA and metformin treatments on response to PTZ in a malin knockout model of LD. Both treatments decreased seizure susceptibility, bringing about a reduction in both seizure number and length, and eliminated the mortality induced by PTZ. These results show a neuroprotective role of 4-PBA and metformin and extend the beneficial effects reported in the malin knockout model of LD Video abstract: http://links.lww.com/WNR/A411.


Subject(s)
Anticonvulsants/therapeutic use , Lafora Disease/drug therapy , Lafora Disease/genetics , Metformin/therapeutic use , Phenylbutyrates/therapeutic use , Ubiquitin-Protein Ligases/genetics , Animals , Brain/drug effects , Brain/metabolism , Convulsants/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Lafora Disease/chemically induced , Lafora Disease/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pentylenetetrazole/toxicity , Ubiquitin-Protein Ligases/deficiency
12.
Epilepsia ; 58(3): 467-475, 2017 03.
Article in English | MEDLINE | ID: mdl-28098937

ABSTRACT

OBJECTIVE: To search for new therapies aimed at ameliorating the neurologic symptoms and epilepsy developing in patients with Lafora disease. METHODS: Lafora disease is caused by loss-of-function mutations in either the EPM2A or EPM2B genes. Epm2a-/- and Epm2b-/- mice display neurologic and behavioral abnormalities similar to those found in patients. Selenium is a potent antioxidant and its deficiency has been related to the development of certain diseases, including epilepsy. In this study, we investigated whether sodium selenate treatment improved the neurologic alterations and the hyperexcitability present in the Epm2b-/- mouse model. RESULTS: Sodium selenate ameliorates some of the motor and memory deficits and the sensitivity observed with pentylenetetrazol (PTZ) treatments in Epm2b-/- mice. Neuronal degeneration and gliosis were also diminished after sodium selenate treatment. SIGNIFICANCE: Sodium selenate could be beneficial for ameliorating some symptoms that present in patients with Lafora disease.


Subject(s)
Antioxidants/therapeutic use , Dual-Specificity Phosphatases/deficiency , Lafora Disease/chemically induced , Lafora Disease/drug therapy , Lafora Disease/genetics , Selenic Acid/therapeutic use , Ubiquitin-Protein Ligases/deficiency , Animals , Anxiety/drug therapy , Anxiety/etiology , Convulsants/toxicity , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Exploratory Behavior/drug effects , Glial Fibrillary Acidic Protein/metabolism , Lafora Disease/complications , Memory Disorders/drug therapy , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Movement Disorders/drug therapy , Movement Disorders/etiology , Pentylenetetrazole/toxicity , Phosphopyruvate Hydratase/metabolism , Protein Tyrosine Phosphatases, Non-Receptor , Psychomotor Performance/drug effects , Recognition, Psychology/drug effects , Ubiquitin-Protein Ligases/genetics
13.
J Lipid Res ; 57(3): 422-32, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26764042

ABSTRACT

Flaviviruses, such as the dengue virus and the West Nile virus (WNV), are arthropod-borne viruses that represent a global health problem. The flavivirus lifecycle is intimately connected to cellular lipids. Among the lipids co-opted by flaviviruses, we have focused on SM, an important component of cellular membranes particularly enriched in the nervous system. After infection with the neurotropic WNV, mice deficient in acid sphingomyelinase (ASM), which accumulate high levels of SM in their tissues, displayed exacerbated infection. In addition, WNV multiplication was enhanced in cells from human patients with Niemann-Pick type A, a disease caused by a deficiency of ASM activity resulting in SM accumulation. Furthermore, the addition of SM to cultured cells also increased WNV infection, whereas treatment with pharmacological inhibitors of SM synthesis reduced WNV infection. Confocal microscopy analyses confirmed the association of SM with viral replication sites within infected cells. Our results unveil that SM metabolism regulates flavivirus infection in vivo and propose SM as a suitable target for antiviral design against WNV.


Subject(s)
Host-Pathogen Interactions , Sphingomyelins/metabolism , West Nile virus/physiology , Animals , Brain/metabolism , Brain/virology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Female , Fibroblasts/metabolism , Fibroblasts/virology , Gene Knockout Techniques , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/virology , Male , Mice , Mice, Inbred C57BL , Niemann-Pick Diseases/pathology , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Virus Replication
14.
Mol Neurobiol ; 53(2): 1296-1309, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25627694

ABSTRACT

Lafora disease (LD, OMIM 254780) is a rare fatal neurodegenerative disorder that usually occurs during childhood with generalized tonic-clonic seizures, myoclonus, absences, drop attacks, or visual seizures. Unfortunately, at present, available treatments are only palliatives and no curative drugs are available yet. The hallmark of the disease is the accumulation of insoluble polyglucosan inclusions, called Lafora bodies (LBs), within the neurons but also in heart, muscle, and liver cells. Mouse models lacking functional EPM2A or EPM2B genes (the two major loci related to the disease) recapitulate the Lafora disease phenotype: they accumulate polyglucosan inclusions, show signs of neurodegeneration, and have a dysregulation of protein clearance and endoplasmic reticulum stress response. In this study, we have subjected a mouse model of LD (Epm2b-/-) to different pharmacological interventions aimed to alleviate protein clearance and endoplasmic reticulum stress. We have used two chemical chaperones, trehalose and 4-phenylbutyric acid. In addition, we have used metformin, an activator of AMP-activated protein kinase (AMPK), as it has a recognized neuroprotective role in other neurodegenerative diseases. Here, we show that treatment with 4-phenylbutyric acid or metformin decreases the accumulation of Lafora bodies and polyubiquitin protein aggregates in the brain of treated animals. 4-Phenylbutyric acid and metformin also diminish neurodegeneration (measured in terms of neuronal loss and reactive gliosis) and ameliorate neuropsychological tests of Epm2b-/- mice. As these compounds have good safety records and are already approved for clinical uses on different neurological pathologies, we think that the translation of our results to the clinical practice could be straightforward.


Subject(s)
Brain/pathology , Lafora Disease/drug therapy , Lafora Disease/pathology , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Gliosis/complications , Gliosis/drug therapy , Gliosis/pathology , Glucans/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Lafora Disease/parasitology , Lafora Disease/physiopathology , Metformin/pharmacology , Metformin/therapeutic use , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/complications , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Neuropsychological Tests , Phenylbutyrates/pharmacology , Phenylbutyrates/therapeutic use , Protein Aggregates/drug effects , Trehalose/pharmacology , Trehalose/therapeutic use , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism , Ubiquitinated Proteins/metabolism
15.
Front Neurosci ; 8: 291, 2014.
Article in English | MEDLINE | ID: mdl-25309313

ABSTRACT

Lafora disease is a rare form of inherited progressive myoclonus epilepsy caused by mutations in the EPM2A gene encoding laforin, or in the EPM2B gene, which encodes malin. It is characterized by the presence of polyglucosan inclusion bodies (Lafora bodies) in brain and other tissues. Genetically engineered mice lacking expression of either the laforin (Epm2a(-/-) ) or malin (Epm2b(-/-) ) genes display a number of neurological and behavioral abnormalities that resemble those found in patients suffering from Lafora disease; of these, both Epm2a(-/-) and Epm2b(-/-) mice have shown altered motor activity, impaired motor coordination, episodic memory deficits, and different degrees of spontaneous epileptic activity. In this study, we analyze the sensitivity of Epm2a(-/-) and Epm2b(-/-) mice to the convulsant drug pentylenetetrazol (PTZ), an antagonist of the γ-aminobutyric acid type A (GABAA) receptor, commonly used to induce epileptic tonic-clonic seizures in laboratory animals. PTZ-induced epileptic activity, including myoclonic jerks and tonic-clonic seizures, was analyzed in 2 age groups of mice comprising representative samples of young adult and aged mice, after administration of PTZ at sub-convulsive and convulsive doses. Epm2a(-/-) and Epm2b(-/-) mice showed a lower convulsive threshold after PTZ injections at sub-convulsive doses. A lower convulsive threshold and shorter latencies to develop epileptic seizures were observed after PTZ injections at convulsive doses. Different patterns of generalized seizures and of discharges were observed in Epm2a(-/-) and Epm2b(-/-) mice. Epm2a(-/-) and Epm2b(-/-) mice present an increased sensitivity to the convulsant agent PTZ that may reflect different degrees of increased GABAA receptor-mediated hyperexcitability.

16.
Neurobiol Dis ; 58: 200-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23774255

ABSTRACT

Epileptic seizures are more common in patients with Alzheimer disease than in the general elderly population. Abnormal forms of hyperphosphorylated tau accumulate in Alzheimer disease and other tauopathies. Aggregates of tau are also found in patients with epilepsy and in experimental models of epilepsy. We report here the analysis of epileptic activity and neuropathological correlates of a transgenic line over-expressing human mutant tau, a model of frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17). The FTDP-17 model displays spontaneous epileptic activity and seizures with spike-wave complexes in the EEG, and a higher sensitivity to the GABAA receptor antagonist pentylenetetrazol (PTZ) when compared to age-matched controls, showing a notably increased seizure length and a shorter latency to develop severe seizures. FTDP-17 human tau mutants also display lower convulsive thresholds and higher lethality after PTZ injections. Astrocytosis and activated microglia are prominent in the hippocampus and other brain regions of young FTDP-17 mice where the human mutant tau transgene is expressed, before the appearance of hyperphosphorylated tau aggregates in these structures. FTDP-17 human mutant tau over-expression produces epilepsy and increased GABAA receptor-mediated hyperexcitability in the absence of Aß pathology. Although aggregates of hyperphosphorylated tau have been observed in patients with epilepsy and in different chemically and electrically generated models of epilepsy, the FTDP-17 tau mutant analyzed here is the first model of genetically modified tau that presents with epilepsy. This model may represent a valuable tool to assay novel treatments in order to reduce tau pathology, a potential factor which may be involved in the development of epileptic seizures in dementia and other neurodegenerative diseases.


Subject(s)
Epilepsy/etiology , Frontotemporal Dementia/complications , Frontotemporal Dementia/genetics , tau Proteins/genetics , Animals , Brain/metabolism , Brain/pathology , Calcium-Binding Proteins/metabolism , Convulsants/toxicity , DNA-Binding Proteins , Disease Models, Animal , Electroencephalography , Epilepsy/chemically induced , Frontotemporal Dementia/pathology , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Pentylenetetrazole/toxicity , Polycomb-Group Proteins , Transcription Factors/metabolism , Video Recording
17.
J Neuropathol Exp Neurol ; 71(5): 413-21, 2012 May.
Article in English | MEDLINE | ID: mdl-22487859

ABSTRACT

Lafora disease is a progressive myoclonus epilepsy caused by mutations in the EPM2A gene encoding laforin or in the EPM2B gene encoding malin. It is characterized by the presence of polyglucosan intracellular inclusion bodies (Lafora bodies) in brain and other tissues. Targeted disruption of Epm2a or Epm2b genes in mice produced widespread neuronal degeneration and accumulation of Lafora bodies in neuronal and nonneuronal tissues. Here we analyzed the neurologic alterations produced by disruption of the laforin gene in Epm2a mice and compared them to those in malin-deficient mice. Both Epm2a and Epm2b mice showed altered motor activity, impaired motor coordination, abnormal hind limb clasping, and episodic memory deficits. Epm2a mice also had tonic-clonic seizures, whereas both Epm2a and Epm2b mice had spontaneous single spikes, spike-wave, polyspikes, and polyspike-wave complexes with correlated myoclonic jerks. Neurologic alterations observed in the mutants were comparable and correlated with the accumulation of abundant Lafora bodies in the cerebral cortex, the hippocampus, the basal ganglia, the cerebellum, and the brainstem, suggesting that these inclusions could cause cognitive and behavioral deterioration. Thus, both Epm2a and Epm2b mice exhibit many pathologic aspects seen in patients with Lafora disease and may be valuable for the study of this disorder.


Subject(s)
Carrier Proteins/metabolism , Dual-Specificity Phosphatases/deficiency , Lafora Disease/complications , Nervous System Diseases/etiology , Sequence Deletion/genetics , Age Factors , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Carrier Proteins/genetics , Disease Models, Animal , Electroencephalography , Exons/genetics , Exploratory Behavior/physiology , Hindlimb Suspension/physiology , Lafora Disease/genetics , Lafora Disease/pathology , Mice , Mice, Knockout , Movement/physiology , Nervous System Diseases/genetics , Neuropsychological Tests , Postural Balance/genetics , Protein Tyrosine Phosphatases, Non-Receptor , Psychomotor Performance/physiology , Recognition, Psychology/physiology , Stereotyped Behavior/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Video Recording
18.
Acta Neuropathol ; 117(2): 159-68, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19057915

ABSTRACT

Mutations in the PARK2 gene encoding parkin cause autosomal recessive juvenile parkinsonism, but have also been found in patients diagnosed with certain tauopathies. Conversely, mutations in the MAPT gene encoding tau are present in some types of parkinsonism. In order to investigate the possible relationship between these two proteins, we generated a double mutant mouse that is deficient in PARK2 and that over-expresses the hTauVLW transgene, a mutant form of the tau protein present in FTDP-17. Independent deletion of PARK2 or over-expression of the hTauVLW transgene produces mild phenotypic alterations, while a substantial increase in parkin expression is observed in hTauVLW transgenic mice. However, double mutant mice present memory and exploratory deficits, and accumulation of PHF-1 and AT8 hyperphosphorylated tau epitopes in neurons. These phenomena are coupled with reactive astrocytosis, DNA fragmentation, and variable cerebral atrophy. Here, we show that cortical and hippocampal neurons of double mutant mice develop argyrophilic Gallyas-Braak aggregates of phosphorylated tau from 3 months of age. Their number decreases in old animals. Moreover, numerous phosphorylated tau aggregates were identified with the conformation-dependent Alz-50 antibody and the S-Thioflavin staining. Ventral motor nuclei of the spinal cord also present Alz-50, AT8, and PHF1 hyperphosphorylated tau aggregates when parkin is deleted in mice over-expressing the hTauVLW transgene, begining at early ages. Thus, the combination of PARK2 gene deletion with hTauVLW over-expression in mice produces abnormal hyperphosphorylated tau aggregates, similar to those observed in the brain of patients diagnosed with certain tauopathies. In the light of these changes, these mice may help to understand the molecular processes responsible for these diseases, and they may aid the development of new therapeutic strategies to treat neurodegenerative diseases related to tau and parkin proteins.


Subject(s)
Cerebral Cortex/metabolism , Hippocampus/metabolism , Ubiquitin-Protein Ligases/genetics , tau Proteins/metabolism , Aging/physiology , Animals , Antigens/metabolism , Blotting, Western , Cerebral Cortex/pathology , DNA-Binding Proteins/metabolism , Gene Deletion , Hippocampus/pathology , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Mutation , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/metabolism , Neuropil/metabolism , Phosphorylation , Polycomb-Group Proteins , Spinal Cord/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , tau Proteins/genetics
19.
J Alzheimers Dis ; 13(2): 161-72, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18376058

ABSTRACT

Mutations, haplotypes, and polymorphisms of tau and Park-2 genes constitute risk factors for developing tauopathies. In order to analyze the possible relationship between parkin and tau we generated a double-mutant mouse deficient for Park-2 expression and overexpressing a mutant tau protein (hTauVLW). Mice develop normally, although the median survival rate is considerably reduced with respect to wild type (45%). Aggregates of phosphorylated tau in neurons and reactive gliosis are quite abundant in cortex and hippocampus of these mice. Moreover, while in young transgenic mice the hTauVLW immunostained transgene product is observed in both cell bodies and dendrites, the hTauVLW mutant protein is only detected in the neuronal cell bodies when Park-2 gene is additionally deleted. Moreover, DNA fragmentation was detected by the TUNEL method, and cerebral atrophy is also present in these regions. The levels of phosphorylated tau and Hsp70 are increased in the double-mutant mice, while CHIP expression in hippocampus is lower when the Park-2 gene is deleted. Thus, the combination of Park-2 gene deletion with hTauVLW transgene overexpression in mice produces serious neuropathological effects, which reflect the existence of some relationship between both proteins.


Subject(s)
Lymphocytes, Null/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Animals , Antibodies/immunology , Atrophy/metabolism , Atrophy/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Gene Deletion , Gliosis/immunology , Gliosis/metabolism , Gliosis/pathology , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/ultrastructure , Immunohistochemistry , Mice , Mice, Transgenic , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nerve Degeneration/pathology , Phosphorylation , Point Mutation/genetics , Polymorphism, Genetic/genetics
20.
Behav Brain Res ; 189(2): 350-6, 2008 Jun 03.
Article in English | MEDLINE | ID: mdl-18346797

ABSTRACT

While mutations in the Park-2 gene are the most frequent cause of autosomal-recessive juvenile parkinsonism (AR-JP), they are also present in several forms of tauopathies. Conversely, in some forms of parkinsonism, mutations in the tau gene have also been observed. Deletion of the Park-2 gene and over-expression of mutant tau independently produce mild brain alterations in mice. However, the presence of both mutations simultaneously causes a tau neuropathology, involving reactive astrocytosis, neuron loss in the cortex and hippocampus, and lesions in nigrostriatal and motor neurons. Furthermore, mutant tau over-expression in mice produces important memory impairment. When "parkin" function was abolished in young tau transgenic mice, the memory alterations were exaggerated. Moreover, additional exploratory and motor deficits were observed in older mice, causing the memory alterations to be underestimated. Thus, while memory deficits are more severe in young mice they were somehow attenuated by exploratory impairments in ageing mutants. This double mutant animal will serve as a useful experimental tool to investigate the abnormal processing of hyperphosphorylated tau and its relationship to the development of the cognitive deficits associated with certain neurodegenerative diseases.


Subject(s)
Exploratory Behavior/physiology , Memory Disorders/metabolism , Recognition, Psychology/physiology , Ubiquitin-Protein Ligases/metabolism , tau Proteins/metabolism , Age Factors , Animals , Gene Expression Regulation , Humans , Male , Memory Disorders/genetics , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Psychomotor Performance/physiology , Ubiquitin-Protein Ligases/genetics , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL