Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Epilepsy Res ; 200: 107317, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38341935

RESUMEN

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.


Asunto(s)
Enfermedad de Lafora , alfa-Glucosidasas , Ratones , Animales , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/genética , Ratones Noqueados , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Glucógeno/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/genética
2.
Neurotherapeutics ; 20(1): 230-244, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36303102

RESUMEN

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.


Asunto(s)
Enfermedad de Lafora , Metformina , Animales , Ratones , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/genética , Metformina/uso terapéutico , Gliosis , Estudios de Seguimiento , Ubiquitina-Proteína Ligasas/genética
3.
J Med Case Rep ; 16(1): 360, 2022 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-36192771

RESUMEN

BACKGROUND: Lafora disease is a rare genetic disorder involving glycogen metabolism disorder. It is inherited by autosomal recessive pattern presenting as a progressive myoclonus epilepsy and neurologic deterioration beginning in adolescence. It is characterized by Lafora bodies in tissues such as brain, skin, muscle, and liver. CASE PRESENTATION: We report a rare case of Lafora disease in a 16-year-old Albanian girl who presented at a tertiary health care center with generalized tonic-clonic seizures, eyelid twitches, hallucinations, headache, and cognitive dysfunction. She was initially treated for generalized epilepsy and received an antiepileptic drug. However, owing to resistance of seizures to this antiepileptic drug, a second drug was introduced. However, seizures continued despite compliance with therapy, and general neurological status began to deteriorate. The child began to have hallucinations and decline of cognitive function. She developed dysarthria and unsteady gait. When admitted to the hospital, blood tests and imaging examinations were planned. The blood tests were unremarkable. There was no relevant family history and no consanguinity. Electroencephalography showed multifocal discharges in both hemispheres, and brain magnetic resonance imaging revealed no abnormality. Axillary skin biopsy revealed inclusion bodies in apocrine glands. Consequently, the child was referred to an advanced center for genetic testing, which also confirmed diagnosis of Lafora disease with a positive mutation on NHLRC1 gene. CONCLUSIONS:  Even though rare as a condition, Lafora disease should be considered on differential diagnosis in progressive and drug-refractory epilepsy in adolescents, especially when followed by cognitive decline.


Asunto(s)
Enfermedad de Lafora , Adolescente , Anticonvulsivantes/uso terapéutico , Niño , Femenino , Glucógeno , Alucinaciones , Humanos , Enfermedad de Lafora/diagnóstico , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/genética , Convulsiones/etiología , Ubiquitina-Proteína Ligasas
4.
Rev Neurol ; 75(6): 159-163, 2022 09 16.
Artículo en Español | MEDLINE | ID: mdl-36098450

RESUMEN

INTRODUCTION: Mioclonic progressive epilepsy (MPE) includes a clinical and genetical heterogeneous group of neuro-degenerative disorders that associate spontaneous and action-induced myoclonus as well as progressive cognitive impairment. Lafora`s disease is a subtype of MPE with autosomical recessive inheritance due to a mutation in EPM2A or EPM2B genes. Seizures, especially myoclonus, are often refractary to antiepileptic drugs (AD). CASE REPORT: In this article we report a patient with Lafora´s disease diagnosis, previously resistant to several AD tested with good and sustained response to zonisamide. Indeed, we describe a brief review about the efficacy of zonisamida in MPE. CONCLUSION: Zonisamide may be considered as a good therapeutic alternative in MPE.


TITLE: Eficacia de la zonisamida en un caso de enfermedad de Lafora y breve revisión en la epilepsia mioclónica progresiva.Introducción. La epilepsia mioclónica progresiva constituye un grupo complejo de enfermedades neurodegenerativas clínica y genéticamente heterogéneas que asocian mioclonías espontáneas o inducidas por la acción y el deterioro neurológico progresivo. Dentro de estas entidades se encuentra la enfermedad de Lafora, una patología autosómica recesiva causada por mutación en el gen responsable de la síntesis de una proteína llamada laforina (EPM2A) o el gen responsable de la síntesis de la proteína malina (EPM2B o NHLRC1). Son entidades cuyas crisis, en especial las mioclonías, son frecuentemente resistentes a los fármacos anticrisis epilépticas. Caso clínico. Presentamos el caso de una paciente con diagnóstico de enfermedad de Lafora que, tras varios regímenes terapéuticos ineficaces, presentó buena respuesta a la introducción de la zonisamida, con una respuesta favorable mantenida en el tiempo. Asimismo, hacemos una breve revisión de la eficacia de la zonisamida en cuadros de epilepsia mioclónica progresiva. Conclusión. La zonisamida puede ser una buena alternativa en el tratamiento de cuadros con epilepsia mioclónica progresiva.


Asunto(s)
Enfermedad de Lafora , Epilepsias Mioclónicas Progresivas , Mioclonía , Anticonvulsivantes/uso terapéutico , Humanos , Enfermedad de Lafora/diagnóstico , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/genética , Epilepsias Mioclónicas Progresivas/tratamiento farmacológico , Epilepsias Mioclónicas Progresivas/genética , Zonisamida/uso terapéutico
5.
Int J Mol Sci ; 23(12)2022 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-35743315

RESUMEN

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.


Asunto(s)
Enfermedad de Lafora , Animales , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/genética , Enfermedad de Lafora/metabolismo , Mutación , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Convulsiones , Trehalosa/farmacología , Ubiquitina-Proteína Ligasas/genética , Pez Cebra/metabolismo
6.
Int J Mol Sci ; 22(10)2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-34069559

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Metformina/uso terapéutico , Animales , Sistema Nervioso Central/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Modelos Animales de Enfermedad , Epilepsia/tratamiento farmacológico , Humanos , Hipoglucemiantes/farmacología , Enfermedad de Lafora/tratamiento farmacológico , Metformina/metabolismo , Metformina/farmacología , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología
7.
Brain ; 144(10): 2985-2993, 2021 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-33993268

RESUMEN

Lafora disease is a fatal progressive myoclonus epilepsy. At root, it is due to constant acquisition of branches that are too long in a subgroup of glycogen molecules, leading them to precipitate and accumulate into Lafora bodies, which drive a neuroinflammatory response and neurodegeneration. As a potential therapy, we aimed to downregulate glycogen synthase, the enzyme responsible for glycogen branch elongation, in mouse models of the disease. We synthesized an antisense oligonucleotide (Gys1-ASO) that targets the mRNA of the brain-expressed glycogen synthase 1 gene (Gys1). We administered Gys1-ASO by intracerebroventricular injection and analysed the pathological hallmarks of Lafora disease, namely glycogen accumulation, Lafora body formation, and neuroinflammation. Gys1-ASO prevented Lafora body formation in young mice that had not yet formed them. In older mice that already exhibited Lafora bodies, Gys1-ASO inhibited further accumulation, markedly preventing large Lafora bodies characteristic of advanced disease. Inhibition of Lafora body formation was associated with prevention of astrogliosis and strong trends towards correction of dysregulated expression of disease immune and neuroinflammatory markers. Lafora disease manifests gradually in previously healthy teenagers. Our work provides proof of principle that an antisense oligonucleotide targeting the GYS1 mRNA could prevent, and halt progression of, this catastrophic epilepsy.


Asunto(s)
Glucógeno Sintasa/administración & dosificación , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/patología , Oligorribonucleótidos Antisentido/administración & dosificación , Animales , Femenino , Inyecciones Intraventriculares , Enfermedad de Lafora/genética , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética
8.
Exp Neurol ; 340: 113656, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33639210

RESUMEN

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.


Asunto(s)
Antiinflamatorios/uso terapéutico , Dexametasona/uso terapéutico , Factores de Transcripción del Choque Térmico/metabolismo , Respuesta al Choque Térmico/efectos de los fármacos , Enfermedad de Lafora/metabolismo , Convulsiones/metabolismo , Animales , Antiinflamatorios/farmacología , Dexametasona/farmacología , Modelos Animales de Enfermedad , Factores de Transcripción del Choque Térmico/deficiencia , Factores de Transcripción del Choque Térmico/genética , Respuesta al Choque Térmico/fisiología , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Enfermedad de Lafora/tratamiento farmacológico , Ratones , Ratones Noqueados , Convulsiones/tratamiento farmacológico
9.
J Biol Chem ; 296: 100150, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33277363

RESUMEN

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.


Asunto(s)
Gliosis/prevención & control , Glucógeno Sintasa/fisiología , Inflamación/prevención & control , Enfermedad de Lafora/patología , Músculo Esquelético/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/deficiencia , Animales , Femenino , Gliosis/metabolismo , Gliosis/patología , Inflamación/metabolismo , Inflamación/patología , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/genética , Enfermedad de Lafora/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/patología , Moduladores Selectivos de los Receptores de Estrógeno/administración & dosificación , Tamoxifeno/administración & dosificación
11.
BMJ Case Rep ; 13(12)2020 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-33370974

RESUMEN

Progressive myoclonic epilepsy (PME) is a progressive neurological disorder. Unfortunately, until now, no definitive curative treatment exists; however, it is of utmost importance to identify patients with PME. The underlying aetiology can be pinpointed if methodological clinical evaluation is performed, followed by subsequent genetic testing. We report a case of PME that was diagnosed as Lafora body disease. This case emphasises that, suspecting and identifying PME is important so as to start appropriate treatment and reduce the probability of morbidity and prognosticate the family.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Disfunción Cognitiva/etiología , Enfermedad de Lafora/diagnóstico , Adolescente , Biopsia , Encéfalo/diagnóstico por imagen , Disfunción Cognitiva/tratamiento farmacológico , Progresión de la Enfermedad , Quimioterapia Combinada , Electroencefalografía , Resultado Fatal , Humanos , Enfermedad de Lafora/complicaciones , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/patología , Imagen por Resonancia Magnética , Masculino , Piel/patología
12.
Artículo en Alemán | MEDLINE | ID: mdl-33086413

RESUMEN

Lafora disease is an autosomal recessive lysosomal storage disorder leading to an accumulation of toxic glycogen bodies into the cells of the central nervous system and other tissues. In the progressive form of myoclonic epilepsy, clinical signs typically start around 7 years of age. Causal therapy is impossible, however, in the early stages the symptoms may at least be alleviated by modern antiepileptic drugs. In the case reported here, an approximately 7-year-old Beagle presented with daytime-dependent fasciculations, focal and generalized myoclonus ranging up to a brief tonic-clonic seizure. The signs could be triggered and augmented by stress, sounds and light. Histologic examination was performed on biopsy samples of skin, liver, muscle and nervous tissue to test for the clinical diagnosis of Lafora disease. Sarcoplasmic PAS-positive pla®ue deposits typical of Lafora bodies were detected in the muscle biopsies but not in any of the other specimens. Initial treatment with phenobarbital and imepitoin was unsuccessful. However, treatment with levetiracetam significantly alleviated the clinical signs. At time of writing this publication, 2 years following the diagnosis, the now 9-year-old dog shows occasional, stress-related increase in fokal myoclonic seizures. Episodes of collapse or tonic-clonic seizures did not occur to any further extent.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Enfermedades de los Perros , Enfermedad de Lafora , Animales , Enfermedades de los Perros/diagnóstico , Enfermedades de los Perros/tratamiento farmacológico , Perros , Enfermedad de Lafora/diagnóstico , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/veterinaria , Levetiracetam/uso terapéutico
13.
Mol Pharm ; 16(9): 3791-3801, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31329461

RESUMEN

Lafora disease (LD) is a fatal juvenile epilepsy characterized by the accumulation of aberrant glucan aggregates called Lafora bodies (LBs). Delivery of protein-based therapeutics to the central nervous system (CNS) for the clearance of LBs remains a unique challenge in the field. Recently, a humanized antigen-binding fragment (hFab) derived from a murine systemic lupus erythematosus DNA autoantibody (3E10) has been shown to mediate cell penetration and proposed as a broadly applicable carrier to mediate cellular targeting and uptake. We report studies on the efficacy and CNS delivery of VAL-0417, an antibody-enzyme fusion composed of the 3E10 hFab and human pancreatic α-amylase, in a mouse model of LD. An enzyme-linked immunosorbent assay has been developed to detect VAL-0417 post-treatment as a measure of delivery efficacy. We demonstrate the robust and sensitive detection of the fusion protein in multiple tissue types. Using this method, we measured biodistribution in different methods of delivery. We found that intracerebroventricular administration provided robust CNS delivery when compared to intrathecal administration. These data define critical steps in the translational pipeline of VAL-0417 for the treatment of LD.


Asunto(s)
Encéfalo/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Enfermedad de Lafora/tratamiento farmacológico , alfa-Amilasas Pancreáticas/genética , alfa-Amilasas Pancreáticas/farmacocinética , Animales , Fusión Artificial Génica/métodos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Portadores de Fármacos/metabolismo , Ensayo de Inmunoadsorción Enzimática , Glucanos/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Plásmidos/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , Distribución Tisular , Resultado del Tratamiento
14.
Orphanet J Rare Dis ; 14(1): 149, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31227012

RESUMEN

BACKGROUND: Lafora disease (LD) is a rare, lethal, progressive myoclonus epilepsy for which no targeted therapy is currently available. Studies on a mouse model of LD showed a good response to metformin, a drug with a well known neuroprotective effect. For this reason, in 2016, the European Medicines Agency granted orphan designation to metformin for the treatment of LD. However, no clinical data is available thus far. METHODS: We retrospectively collected data on LD patients treated with metformin referred to three Italian epilepsy centres. RESULTS: Twelve patients with genetically confirmed LD (6 EPM2A, 6 NHLRC1) at middle/late stages of disease were treated with add-on metformin for a mean period of 18 months (range: 6-36). Metformin was titrated to a mean maintenance dose of 1167 mg/day (range: 500-2000 mg). In four patients dosing was limited by gastrointestinal side-effects. No serious adverse events occurred. Three patients had a clinical response, which was temporary in two, characterized by a reduction of seizure frequency and global clinical improvement. CONCLUSIONS: Metformin was overall safe in our small cohort of LD patients. Even though the clinical outcome was poor, this may be related to the advanced stage of disease in our cases and we cannot exclude a role of metformin in slowing down LD progression. Therefore, on the grounds of the preclinical data, we believe that treatment with metformin may be attempted as early as possible in the course of LD.


Asunto(s)
Enfermedad de Lafora/tratamiento farmacológico , Metformina/uso terapéutico , Adolescente , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedad de Lafora/genética , Masculino , Mutación/genética , Epilepsias Mioclónicas Progresivas/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , Estudios Retrospectivos , Ubiquitina-Proteína Ligasas/genética
16.
Neuroreport ; 28(5): 268-271, 2017 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-28181916

RESUMEN

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.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/genética , Metformina/uso terapéutico , Fenilbutiratos/uso terapéutico , Ubiquitina-Proteína Ligasas/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Convulsivantes/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Enfermedad de Lafora/inducido químicamente , Enfermedad de Lafora/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pentilenotetrazol/toxicidad , Ubiquitina-Proteína Ligasas/deficiencia
17.
Epilepsia ; 58(3): 467-475, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28098937

RESUMEN

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.


Asunto(s)
Antioxidantes/uso terapéutico , Fosfatasas de Especificidad Dual/deficiencia , Enfermedad de Lafora/inducido químicamente , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/genética , Ácido Selénico/uso terapéutico , Ubiquitina-Proteína Ligasas/deficiencia , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/etiología , Convulsivantes/toxicidad , Modelos Animales de Enfermedad , Fosfatasas de Especificidad Dual/genética , Conducta Exploratoria/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Enfermedad de Lafora/complicaciones , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos del Movimiento/tratamiento farmacológico , Trastornos del Movimiento/etiología , Pentilenotetrazol/toxicidad , Fosfopiruvato Hidratasa/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras , Desempeño Psicomotor/efectos de los fármacos , Reconocimiento en Psicología/efectos de los fármacos , Ubiquitina-Proteína Ligasas/genética
18.
Epileptic Disord ; 18(S2): 38-62, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27702709

RESUMEN

Lafora disease (LD) is an autosomal recessive progressive myoclonus epilepsy due to mutations in the EPM2A (laforin) and EPM2B (malin) genes, with no substantial genotype-phenotype differences between the two. Founder effects and recurrent mutations are common, and mostly isolated to specific ethnic groups and/or geographical locations. Pathologically, LD is characterized by distinctive polyglucosans, which are formations of abnormal glycogen. Polyglucosans, or Lafora bodies (LB) are typically found in the brain, periportal hepatocytes of the liver, skeletal and cardiac myocytes, and in the eccrine duct and apocrine myoepithelial cells of sweat glands. Mouse models of the disease and other naturally occurring animal models have similar pathology and phenotype. Hypotheses of LB formation remain controversial, with compelling evidence and caveats for each hypothesis. However, it is clear that the laforin and malin functions regulating glycogen structure are key. With the exception of a few missense mutations LD is clinically homogeneous, with onset in adolescence. Symptoms begin with seizures, and neurological decline follows soon after. The disease course is progressive and fatal, with death occurring within 10 years of onset. Antiepileptic drugs are mostly non-effective, with none having a major influence on the progression of cognitive and behavioral symptoms. Diagnosis and genetic counseling are important aspects of LD, and social support is essential in disease management. Future therapeutics for LD will revolve around the pathogenesics of the disease. Currently, efforts at identifying compounds or approaches to reduce brain glycogen synthesis appear to be highly promising.


Asunto(s)
Enfermedad de Lafora , Animales , Modelos Animales de Enfermedad , Asesoramiento Genético , Glucógeno/metabolismo , Humanos , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/genética , Enfermedad de Lafora/metabolismo , Enfermedad de Lafora/fisiopatología , Ratones , Proteínas Tirosina Fosfatasas no Receptoras/genética
19.
Epilepsy Behav ; 62: 132-5, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27459034

RESUMEN

Lafora disease (LD) is a fatal intractable adolescence-onset progressive myoclonus epilepsy. Recently, two single-case studies reported drastic reductions in seizures and myoclonus with the AMPA antagonist perampanel and improved activities of daily living. We proceeded to study the effect of perampanel on 10 patients with genetically confirmed LD with disease duration ranging from 2 to 27years. Open-label perampanel was added to ongoing medications to a mean dose of 6.7mg/day. Seizures, myoclonus, functional disability, and cognition scores were measured for the third and ninth months following initiation and compared with those of the month prior to the start of therapy. Three patients withdrew because of inefficacy or side effects. Four had significant reduction in seizures of greater than 74% from baseline. Seven had major improvement in myoclonus with group-adjusted sum score of myoclonus intensity reduced from 7.01 at baseline to 5.67 and 5.18 at 3 and 9months, respectively. There was no significant improvement in disability and cognition. While not universal, perampanel adjunctive therapy appears to confer particular benefit not commonly seen with other antiepileptic drugs on seizures and myoclonus in LD. Improvement in the extremely disabling myoclonus of LD is a major benefit in this devastating disease.


Asunto(s)
Actividades Cotidianas , Anticonvulsivantes/uso terapéutico , Enfermedad de Lafora/tratamiento farmacológico , Mioclonía/tratamiento farmacológico , Piridonas/uso terapéutico , Adolescente , Adulto , Anticonvulsivantes/efectos adversos , Femenino , Humanos , Masculino , Nitrilos , Piridonas/efectos adversos , Adulto Joven
20.
Mol Neurobiol ; 53(2): 1296-1309, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25627694

RESUMEN

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.


Asunto(s)
Encéfalo/patología , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/patología , Animales , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Gliosis/complicaciones , Gliosis/tratamiento farmacológico , Gliosis/patología , Glucanos/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/patología , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/metabolismo , Enfermedad de Lafora/parasitología , Enfermedad de Lafora/fisiopatología , Metformina/farmacología , Metformina/uso terapéutico , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/complicaciones , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/patología , Pruebas Neuropsicológicas , Fenilbutiratos/farmacología , Fenilbutiratos/uso terapéutico , Agregado de Proteínas/efectos de los fármacos , Trehalosa/farmacología , Trehalosa/uso terapéutico , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Ubiquitinadas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...