Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dis Model Mech ; 16(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36511140

RESUMEN

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.


Asunto(s)
Glucógeno , Enfermedad de Lafora , Proteínas Tirosina Fosfatasas no Receptoras , Ubiquitina-Proteína Ligasas , Animales , Ratones , Glucógeno/metabolismo , Enfermedad de Lafora/genética , Enfermedad de Lafora/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo
2.
Neurotherapeutics ; 19(3): 982-993, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35347645

RESUMEN

Adult polyglucosan body disease (APBD) and Lafora disease (LD) are autosomal recessive glycogen storage neurological disorders. APBD is caused by mutations in the glycogen branching enzyme (GBE1) gene and is characterized by progressive upper and lower motor neuron dysfunction and premature death. LD is a fatal progressive myoclonus epilepsy caused by loss of function mutations in the EPM2A or EPM2B gene. These clinically distinct neurogenetic diseases share a common pathology. This consists of time-dependent formation, precipitation, and accumulation of an abnormal form of glycogen (polyglucosan) into gradually enlarging inclusions, polyglucosan bodies (PBs) in ever-increasing numbers of neurons and astrocytes. The growth and spread of PBs are followed by astrogliosis, microgliosis, and neurodegeneration. The key defect in polyglucosans is that their glucan branches are longer than those of normal glycogen, which prevents them from remaining in solution. Since the lengths of glycogen branches are determined by the enzyme glycogen synthase, we hypothesized that downregulating this enzyme could prevent or hinder the generation of the pathogenic PBs. Here, we pursued an adeno-associated virus vector (AAV) mediated RNA-interference (RNAi) strategy. This approach resulted in approximately 15% reduction of glycogen synthase mRNA and an approximately 40% reduction of PBs across the brain in the APBD and both LD mouse models. This was accompanied by improvements in early neuroinflammatory markers of disease. This work represents proof of principle toward developing a single lifetime dose therapy for two fatal neurological diseases: APBD and LD. The approach is likely applicable to other severe and common diseases of glycogen storage.


Asunto(s)
Enfermedad de Lafora , MicroARNs , Animales , Modelos Animales de Enfermedad , Glucanos , Glucógeno , Enfermedad del Almacenamiento de Glucógeno , Glucógeno Sintasa/genética , Enfermedad de Lafora/genética , Enfermedad de Lafora/patología , Enfermedad de Lafora/terapia , Ratones , Enfermedades del Sistema Nervioso , Enfermedades Neuroinflamatorias
3.
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
4.
Neurotherapeutics ; 18(2): 1414-1425, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33830476

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Glucanos/metabolismo , Enfermedad del Almacenamiento de Glucógeno/genética , Glucógeno Sintasa/genética , Glucógeno/metabolismo , Enfermedad de Lafora/genética , Enfermedades del Sistema Nervioso/genética , Enfermedades Neuroinflamatorias/genética , ARN Mensajero/metabolismo , Animales , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Edición Génica , Enfermedad del Almacenamiento de Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno/terapia , Enfermedad de Lafora/metabolismo , Enfermedad de Lafora/terapia , Ratones , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/terapia , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/terapia , Prueba de Estudio Conceptual
5.
J Neuroimmunol ; 354: 577544, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33756414

RESUMEN

The antioxidant MnTBAP was previously shown to down-regulate the surface expression of CD4 molecule in T cells. This observation obviously holds great potential impact in a number of pathological human conditions, including autoimmunity. Three different single doses of MnTBAP reduced the frequency of CD4high cells. However, the median florescent intensity (MFI) was not different. Initiation of in vivo pharmacotherapy or vehicle control was performed inC57BL/6 mice that were actively immunized for experimental autoimmune encephalomyelitis (EAE). In contrast to published reports, the mean frequency of CD4high cells, and the median fluorescent intensity (MFI) of CD4 was similar in both treatment groups. 25-day survival following active immunization among the MnTBAP treated animals compared to vehicle controls was16.6 ± 6.9 days vs 23.6 ± 2.7 days; (P value <0.05). We conclude that MnTBAP (Sack and Herzog, 2009 (Sack and Herzog, 2009)) does not effectively downregulate CD4 expression in T cells in vivo, probably due to extensive mechanism that distinguishes it from an in vitro model (Harding, 1993 (Harding, 1993)) possesses toxic properties that may limit its clinic use in possible doses that could deliver the immunomodulation through down regulation of CD4 expression, and (Saizawa et al., 1987 (Saizawa et al., 1987)) has limited availability in specific tissues, including the CNS.


Asunto(s)
Antioxidantes/farmacología , Antígenos CD4/biosíntesis , Linfocitos T CD4-Positivos/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/inmunología , Metaloporfirinas/farmacología , Animales , Antígenos CD4/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Regulación hacia Abajo , Ratones , Ratones Endogámicos C57BL
6.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...