Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Chembiochem ; 25(10): e202400184, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38573110

RESUMEN

Genetic aberrations of the maternal UBE3A allele, which encodes the E3 ubiquitin ligase E6AP, are the cause of Angelman syndrome (AS), an imprinting disorder. In most cases, the maternal UBE3A allele is not expressed. Yet, approximately 10 percent of AS individuals harbor distinct point mutations in the maternal allele resulting in the expression of full-length E6AP variants that frequently display compromised ligase activity. In a high-throughput screen, we identified cyanocobalamin, a vitamin B12-derivative, and several alloxazine derivatives as activators of the AS-linked E6AP-F583S variant. Furthermore, we show by cross-linking coupled to mass spectrometry that cobalamins affect the structural dynamics of E6AP-F583S and apply limited proteolysis coupled to mass spectrometry to obtain information about the regions of E6AP that are involved in, or are affected by binding cobalamins and alloxazine derivatives. Our data suggest that dietary supplementation with vitamin B12 can be beneficial for AS individuals.


Asunto(s)
Síndrome de Angelman , Ubiquitina-Proteína Ligasas , Vitamina B 12 , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Síndrome de Angelman/genética , Síndrome de Angelman/tratamiento farmacológico , Síndrome de Angelman/metabolismo , Humanos , Regulación Alostérica/efectos de los fármacos , Vitamina B 12/metabolismo , Vitamina B 12/química , Vitamina B 12/farmacología
2.
Nat Commun ; 14(1): 1167, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36859399

RESUMEN

Angelman syndrome (AS) is a neurogenetic disorder characterized by intellectual disability and atypical behaviors. AS results from loss of expression of the E3 ubiquitin-protein ligase UBE3A from the maternal allele in neurons. Individuals with AS display impaired coordination, poor balance, and gait ataxia. PIEZO2 is a mechanosensitive ion channel essential for coordination and balance. Here, we report that PIEZO2 activity is reduced in Ube3a deficient male and female mouse sensory neurons, a human Merkel cell carcinoma cell line and female human iPSC-derived sensory neurons with UBE3A knock-down, and de-identified stem cell-derived neurons from individuals with AS. We find that loss of UBE3A decreases actin filaments and reduces PIEZO2 expression and function. A linoleic acid (LA)-enriched diet increases PIEZO2 activity, mechano-excitability, and improves gait in male AS mice. Finally, LA supplementation increases PIEZO2 function in stem cell-derived neurons from individuals with AS. We propose a mechanism whereby loss of UBE3A expression reduces PIEZO2 function and identified a fatty acid that enhances channel activity and ameliorates AS-associated mechano-sensory deficits.


Asunto(s)
Síndrome de Angelman , Canales Iónicos , Ácido Linoleico , Animales , Femenino , Humanos , Masculino , Ratones , Alelos , Síndrome de Angelman/tratamiento farmacológico , Síndrome de Angelman/genética , Modelos Animales de Enfermedad , Discapacidad Intelectual , Canales Iónicos/genética , Ácido Linoleico/farmacología
3.
Mol Cell Neurosci ; 120: 103724, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35367589

RESUMEN

We recently generated a novel Angelman syndrome (AS) rat model with a complete Ube3a gene deletion, that recapitulates the loss of UBE3A protein and shows cognitive and EEG deficits. We also recently published the identification of extracellular UBE3A protein within the brain using microdialysis. Here we explored the effects of supplementation of exogenous UBE3A protein to hippocampal slices and intrahippocampal injection of AS rats. We report that the AS rat model demonstrates deficits in hippocampal long-term potentiation (LTP) which can be recovered with the application of exogenous UBE3A protein. Furthermore, injection of recombinant UBE3A protein into the hippocampus of the AS rat can rescue the associative learning and memory deficits seen in the fear conditioning task. These data suggest that extracellular UBE3A protein may play a role in synaptic function, LTP induction and hippocampal-dependent memory formation.


Asunto(s)
Síndrome de Angelman , Síndrome de Angelman/tratamiento farmacológico , Síndrome de Angelman/genética , Síndrome de Angelman/metabolismo , Animales , Suplementos Dietéticos , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Potenciación a Largo Plazo , Ratas , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
Neurobiol Dis ; 76: 77-86, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25684537

RESUMEN

Genetic defects in the UBE3A gene, which encodes for the imprinted E6-AP ubiquitin E3 ligase (UBE3A), is responsible for the occurrence of Angelman syndrome (AS), a neurodegenerative disorder which arises in 1 out of every 12,000-20,000 births. Classical symptoms of AS include delayed development, impaired speech, and epileptic seizures with characteristic electroencephalography (EEG) readings. We have previously reported impaired mitochondrial structure and reduced complex III in the hippocampus and cerebellum in the Ube3a(m-/p+) mice. CoQ10 supplementation restores the electron flow to the mitochondrial respiratory chain (MRC) to ultimately increase mitochondrial antioxidant capacity. A number of recent studies with CoQ10 analogues seem promising in providing therapeutic benefit to patients with a variety of disorders. CoQ10 therapy has been reported to be safe and relatively well-tolerated at doses as high as 3000mg/day in patients with disorders of CoQ10 biosynthesis and MRC disorders. Herein, we report administration of idebenone, a potent CoQ10 analogue, to the Ube3a(m-/p+) mouse model corrects motor coordination and anxiety levels, and also improves the expression of complexes III and IV in hippocampus CA1 and CA2 neurons and cerebellum in these Ube3a(m-/p+) mice. However, treatment with idebenone illustrated no beneficial effects in the reduction of oxidative stress. To our knowledge, this is the first study to suggest an improvement in mitochondrial respiratory chain dysfunction via bioenergetics modulation with a CoQ10 analogue. These findings may further elucidate possible cellular and molecular mechanism(s) and ultimately a clinical therapeutic approach/benefit for patients with Angelman syndrome.


Asunto(s)
Síndrome de Angelman/tratamiento farmacológico , Síndrome de Angelman/metabolismo , Antioxidantes/administración & dosificación , Transporte de Electrón/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ubiquinona/análogos & derivados , Animales , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Actividad Motora/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ubiquinona/administración & dosificación , Ubiquitina-Proteína Ligasas/genética
5.
Nature ; 481(7380): 185-9, 2011 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-22190039

RESUMEN

Angelman syndrome is a severe neurodevelopmental disorder caused by deletion or mutation of the maternal allele of the ubiquitin protein ligase E3A (UBE3A). In neurons, the paternal allele of UBE3A is intact but epigenetically silenced, raising the possibility that Angelman syndrome could be treated by activating this silenced allele to restore functional UBE3A protein. Using an unbiased, high-content screen in primary cortical neurons from mice, we identify twelve topoisomerase I inhibitors and four topoisomerase II inhibitors that unsilence the paternal Ube3a allele. These drugs included topotecan, irinotecan, etoposide and dexrazoxane (ICRF-187). At nanomolar concentrations, topotecan upregulated catalytically active UBE3A in neurons from maternal Ube3a-null mice. Topotecan concomitantly downregulated expression of the Ube3a antisense transcript that overlaps the paternal copy of Ube3a. These results indicate that topotecan unsilences Ube3a in cis by reducing transcription of an imprinted antisense RNA. When administered in vivo, topotecan unsilenced the paternal Ube3a allele in several regions of the nervous system, including neurons in the hippocampus, neocortex, striatum, cerebellum and spinal cord. Paternal expression of Ube3a remained elevated in a subset of spinal cord neurons for at least 12 weeks after cessation of topotecan treatment, indicating that transient topoisomerase inhibition can have enduring effects on gene expression. Although potential off-target effects remain to be investigated, our findings suggest a therapeutic strategy for reactivating the functional but dormant allele of Ube3a in patients with Angelman syndrome.


Asunto(s)
Alelos , Silenciador del Gen/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Inhibidores de Topoisomerasa/farmacología , Ubiquitina-Proteína Ligasas/genética , Síndrome de Angelman/tratamiento farmacológico , Síndrome de Angelman/genética , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Evaluación Preclínica de Medicamentos , Padre , Femenino , Impresión Genómica/efectos de los fármacos , Impresión Genómica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Madres , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Inhibidores de Topoisomerasa/administración & dosificación , Inhibidores de Topoisomerasa/análisis , Inhibidores de Topoisomerasa/farmacocinética , Topotecan/administración & dosificación , Topotecan/farmacocinética , Topotecan/farmacología , Ubiquitina-Proteína Ligasas/deficiencia
7.
Am J Med Genet A ; 155A(12): 2956-63, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22002941

RESUMEN

Angelman syndrome (AS) is due to deficient ubiquitin protein ligase 3a, the gene for which (UBE3A) maps to chromosome 15q11-q13 and is imprinted such that only the maternally inherited gene is expressed. The paternally inherited UBE3A gene is silenced, a process mediated by an antisense transcript. We conducted a trial using methylation-promoting dietary supplements (betaine, metafolin, creatine, and vitamin B(12) ) in an attempt to reduce antisense transcript production, increase UBE3A expression, and ameliorate the symptoms of AS. Neuropsychological evaluations, biochemical testing, and assessment of DNA methylation were performed at the beginning and at the end of 1 year of supplementation. The primary outcome measures were changes in the level of developmental function (cognitive, motor, and language) as measured using standardized instruments. The secondary outcomes measures were changes in biochemical parameters and global DNA methylation. These data were compared to those of a control group from a previous randomized double-blind trial using folic acid and betaine. There were no statistically significant changes in the developmental performance of children treated with supplements. There were no unexpected changes in biochemical parameters and no change in site-specific DNA methylation when comparing samples from before and after treatment. There were 10 adverse events that resulted in study withdrawal of 7 participants (worsening of seizures, onset, or worsening of sleep problems, constipation, and anorexia). Supplementation with betaine, metafolin, creatine, and vitamin B(12) appears safe but ineffective in decreasing the severity of AS.


Asunto(s)
Síndrome de Angelman/tratamiento farmacológico , Metilación de ADN/efectos de los fármacos , Suplementos Dietéticos , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Psicometría , Resultado del Tratamiento
8.
Am J Med Genet A ; 152A(8): 1994-2001, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20635355

RESUMEN

Angelman syndrome (AS) is caused by reduced or absent expression of the maternally inherited ubiquitin protein ligase 3A gene (UBE3A), which maps to chromosome 15q11-q13. UBE3A is subject to genomic imprinting in neurons in most regions of the brain. Expression of UBE3A from the maternal chromosome is essential to prevent AS, because the paternally inherited gene is not expressed, probably mediated by antisense UBE3A RNA. We hypothesized that increasing methylation might reduce expression of the antisense UBE3A RNA, thereby increasing UBE3A expression from the paternal gene and ameliorating the clinical phenotype. We conducted a trial using two dietary supplements, betaine and folic acid to promote global levels of methylation and attempt to activate the paternally inherited UBE3A gene. We performed a number of investigations at regular intervals including general clinical and developmental evaluations, biochemical determinations on blood and urine, and electroencephalographic studies. We report herein the data on 48 children with AS who were enrolled in a double-blind placebo-controlled protocol using betaine and folic acid for 1 year. There were no statistically significant changes between treated and untreated children; however, in a small subset of patients we observed some positive trends.


Asunto(s)
Síndrome de Angelman/tratamiento farmacológico , Betaína/uso terapéutico , Ácido Fólico/uso terapéutico , Adolescente , Síndrome de Angelman/genética , Síndrome de Angelman/patología , Niño , Preescolar , Cromosomas Humanos Par 15/genética , Metilación de ADN , Método Doble Ciego , Combinación de Medicamentos , Femenino , Impresión Genómica , Humanos , Lactante , Lipotrópicos/uso terapéutico , Masculino , Fenotipo , Placebos , Ubiquitina-Proteína Ligasas/genética , Complejo Vitamínico B/uso terapéutico
9.
Epilepsia ; 46(12): 1860-70, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16393151

RESUMEN

PURPOSE: gamma-Aminobutyric acid receptor (GABA(A)r) subunit beta3-deficient mice model Angelman syndrome by displaying impaired learning, abnormal EEG with interictal spikes and slowing, myoclonus, and convulsions. The beta3-subunit deficiency causes a failure of intrathalamic reticular nucleus inhibition, leading to abnormally synchronized thalamocortical oscillations. We postulated that this pathophysiology underlies the abnormal cortical EEG and triggers interictal spikes and seizures, but extrathalamic regions also contribute to interictal spikes and seizures, so that the EEG slowing should reveal an absence-like response profile, whereas spikes and seizures have dual responsiveness to absence and partial-seizure drugs. METHODS: Recording electrodes were implanted over the parietal cortices of wild-type, heterozygotes, and homozygous null mice. In each experiment, EEG was recorded for 45 min, either drug or vehicle administered, and EEG recorded for another 3 h. Each EEG was scored for slow-wave activity, interictal spikes, and seizures by a reader blinded to treatments. RESULTS: Interictal spiking and percentage of time in EEG slowing in heterozygotes were increased by the proabsence drug baclofen (GABA(B)-receptor agonist), whereas CGP 35348 (GABA(B)-receptor antagonist) had the opposite effect. The antiabsence drug ethosuximide markedly suppressed EEG slowing and interictal spiking in heterozygote and null mice. Broad-spectrum clonazepam and valproate were more effective on interictal spiking than on EEG slowing, and fosphenytoin suppressed only interictal spiking. CONCLUSIONS: The results suggest that this model of Angelman syndrome, although not expressing typical absence seizures, is characterized by hypersynchronous thalamocortical oscillations that possess absence-like pharmacologic responsiveness and promote EEG slowing, interictal spikes, and convulsive seizures.


Asunto(s)
Síndrome de Angelman/fisiopatología , Corteza Cerebral/fisiopatología , Electroencefalografía/efectos de los fármacos , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/fisiología , Tálamo/fisiopatología , Síndrome de Angelman/tratamiento farmacológico , Síndrome de Angelman/metabolismo , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Baclofeno/farmacología , Corteza Cerebral/efectos de los fármacos , Modelos Animales de Enfermedad , Electrodos Implantados , Electroencefalografía/estadística & datos numéricos , Etosuximida/farmacología , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , Vías Nerviosas/fisiopatología , Compuestos Organofosforados/farmacología , Lóbulo Parietal/fisiopatología , Receptores de GABA-A/deficiencia , Tálamo/efectos de los fármacos
10.
Epilepsia ; 43(1): 103-4, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11879394

RESUMEN

PURPOSE: We examined whether a dietary supplement containing omega-3 polyunsaturated fatty acids (n-3 PUFAs) can alleviate and/or reduce the frequency of epileptic seizures in patients with central nervous system (CNS) diseases treated with anticonvulsive drugs (ACDs). METHODS: A special spread containing 65% n-3 PUFAs was added to the daily diet. The patients consumed 5 g of this spread at every breakfast for 6 months. RESULTS: Five patients completed the study. In all of them, a marked reduction in both frequency and strength of the epileptic seizures was recorded. CONCLUSIONS: Incorporation of the dietary supplement containing n-3 PUFAs may be beneficial in suppression of some cases of epileptic seizures.


Asunto(s)
Ácidos Grasos Omega-3/administración & dosificación , Espasmos Infantiles/dietoterapia , Adolescente , Adulto , Síndrome de Angelman/dietoterapia , Síndrome de Angelman/tratamiento farmacológico , Anticonvulsivantes/uso terapéutico , Parálisis Cerebral/dietoterapia , Parálisis Cerebral/tratamiento farmacológico , Niño , Femenino , Humanos , Lactante , Masculino , Espasmos Infantiles/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA