Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 276
Filtrar
1.
J Neurodev Disord ; 16(1): 18, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637764

RESUMEN

BACKGROUND: Overactivity is prevalent in several rare genetic neurodevelopmental syndromes, including Smith-Magenis syndrome, Angelman syndrome, and tuberous sclerosis complex, although has been predominantly assessed using questionnaire techniques. Threats to the precision and validity of questionnaire data may undermine existing insights into this behaviour. Previous research indicates objective measures, namely actigraphy, can effectively differentiate non-overactive children from those with attention-deficit hyperactivity disorder. This study is the first to examine the sensitivity of actigraphy to overactivity across rare genetic syndromes associated with intellectual disability, through comparisons with typically-developing peers and questionnaire overactivity estimates. METHODS: A secondary analysis of actigraphy data and overactivity estimates from The Activity Questionnaire (TAQ) was conducted for children aged 4-15 years with Smith-Magenis syndrome (N=20), Angelman syndrome (N=26), tuberous sclerosis complex (N=16), and typically-developing children (N=61). Actigraphy data were summarized using the M10 non-parametric circadian rhythm variable, and 24-hour activity profiles were modelled via functional linear modelling. Associations between actigraphy data and TAQ overactivity estimates were explored. Differences in actigraphy-defined activity were also examined between syndrome and typically-developing groups, and between children with high and low TAQ overactivity scores within syndromes. RESULTS: M10 and TAQ overactivity scores were strongly positively correlated for children with Angelman syndrome and Smith-Magenis syndrome. M10 did not substantially differ between the syndrome and typically-developing groups. Higher early morning activity and lower evening activity was observed across all syndrome groups relative to typically-developing peers. High and low TAQ group comparisons revealed syndrome-specific profiles of overactivity, persisting throughout the day in Angelman syndrome, occurring during the early morning and early afternoon in Smith-Magenis syndrome, and manifesting briefly in the evening in tuberous sclerosis complex. DISCUSSION: These findings provide some support for the sensitivity of actigraphy to overactivity in children with rare genetic syndromes, and offer syndrome-specific temporal descriptions of overactivity. The findings advance existing descriptions of overactivity, provided by questionnaire techniques, in children with rare genetic syndromes and have implications for the measurement of overactivity. Future studies should examine the impact of syndrome-related characteristics on actigraphy-defined activity and overactivity estimates from actigraphy and questionnaire techniques.


Asunto(s)
Síndrome de Angelman , Discapacidad Intelectual , Síndrome de Smith-Magenis , Esclerosis Tuberosa , Niño , Humanos , Síndrome de Smith-Magenis/complicaciones , Síndrome de Angelman/complicaciones , Síndrome de Angelman/diagnóstico , Esclerosis Tuberosa/complicaciones , Discapacidad Intelectual/complicaciones
2.
Am J Intellect Dev Disabil ; 129(3): 175-190, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38657964

RESUMEN

Hyperphagia is highly penetrant in Prader-Willi syndrome (PWS) and has increasingly been reported in other neurogenetic conditions (NGC). The Hyperphagia Questionnaire (HQ) was completed by caregivers of 4-8-year-olds with PWS (n = 17), Angelman syndrome (AS; n = 22), Williams syndrome (WS; n = 25), or low-risk controls (LRC; n = 35). All NGC groups were significantly elevated in HQ Total and Behavior scores compared to LRC. Only AS and WS were significantly elevated in the Drive domain, and only PWS in the Severity domain. After controlling for externalizing behavior, HQ Total scores were higher for PWS relative to other groups. Hyperphagic symptoms may not differentiate PWS from other NGCs in early childhood. However, hyperphagic phenotypes may be most severe in PWS. Further investigation of these profiles may inform etiology and syndrome-specific treatments.


Asunto(s)
Síndrome de Angelman , Hiperfagia , Síndrome de Prader-Willi , Humanos , Preescolar , Masculino , Femenino , Síndrome de Prader-Willi/diagnóstico , Niño , Síndrome de Angelman/fisiopatología , Síndrome de Angelman/diagnóstico
3.
Virus Res ; 335: 199191, 2023 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-37541588

RESUMEN

The Ubiquitin-protein ligase E3A, UBE3A, also known as E6-associated protein (E6-AP), is known to play an essential role in regulating the degradation of various proteins by transferring Ub from E2 Ub conjugating enzymes to the substrate proteins. Several studies indicate that UBE3A regulates the stabilities of key viral proteins in the virus-infected cells and, thereby, the infected virus-mediated diseases, even if it were reported that UBE3A participates in non-viral-related human diseases. Furthermore, mutations such as deletions and duplications in the maternally inherited gene in the brain cause human neurodevelopmental disorders such as Angelman syndrome (AS) and autism. It is also known that UBE3A functions as a transcriptional coactivator for the expression of steroid hormone receptors. These reports establish that UBE3A is distinguished by its multitudinous functions that are paramount to viral pathology and human diseases. This review is focused on molecular mechanisms for such intensive participation of UBE3A in disease formation and virus regulation.


Asunto(s)
Síndrome de Angelman , Virosis , Humanos , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Mutación , Encéfalo/patología , Síndrome de Angelman/genética , Síndrome de Angelman/metabolismo , Síndrome de Angelman/patología , Virosis/genética
4.
Neuropharmacology ; 238: 109642, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37392820

RESUMEN

The involvement of the mGlu5 receptors in the pathophysiology of several forms of monogenic autism has been supported by numerous studies following the seminal observation that mGlu5 receptor-dependent long-term depression was enhanced in the hippocampus of mice modeling the fragile-X syndrome (FXS). Surprisingly, there are no studies examining the canonical signal transduction pathway activated by mGlu5 receptors (i.e. polyphosphoinositide - PI - hydrolysis) in mouse models of autism. We have developed a method for in vivo assessment of PI hydrolysis based on systemic injection of lithium chloride followed by treatment with the selective mGlu5 receptor PAM, VU0360172, and measurement of endogenous inositolmonophosphate (InsP) in brain tissue. Here, we report that mGlu5 receptor-mediated PI hydrolysis was blunted in the cerebral cortex, hippocampus, and corpus striatum of Ube3am-/p+ mice modeling Angelman syndrome (AS), and in the cerebral cortex and hippocampus of Fmr1 knockout mice modeling FXS. In vivo mGlu5 receptor-mediated stimulation of Akt on threonine 308 was also blunted in the hippocampus of FXS mice. These changes were associated with a significant increase in cortical and striatal Homer1 levels and striatal mGlu5 receptor and Gαq levels in AS mice, and with a reduction in cortical mGlu5 receptor and hippocampal Gαq levels, and an increase in cortical phospholipase-Cß and hippocampal Homer1 levels in FXS mice. This is the first evidence that the canonical transduction pathway activated by mGlu5 receptors is down-regulated in brain regions of mice modeling monogenic autism.


Asunto(s)
Síndrome de Angelman , Trastorno Autístico , Síndrome del Cromosoma X Frágil , Ratones , Animales , Fosfatos de Fosfatidilinositol/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Hidrólisis , Modelos Animales de Enfermedad , Ratones Noqueados , Síndrome del Cromosoma X Frágil/metabolismo , Proteínas Portadoras , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo
5.
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
6.
Mol Psychiatry ; 28(6): 2382-2397, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36991133

RESUMEN

Angelman syndrome (AS) is a rare genetic neurodevelopmental disorder caused by the maternally inherited loss of function of the UBE3A gene. AS is characterized by a developmental delay, lack of speech, motor dysfunction, epilepsy, autistic features, happy demeanor, and intellectual disability. While the cellular roles of UBE3A are not fully understood, studies suggest that the lack of UBE3A function is associated with elevated levels of reactive oxygen species (ROS). Despite the accumulating evidence emphasizing the importance of ROS during early brain development and its involvement in different neurodevelopmental disorders, up to date, the levels of ROS in AS neural precursor cells (NPCs) and the consequences on AS embryonic neural development have not been elucidated. In this study we show multifaceted mitochondrial aberration in AS brain-derived embryonic NPCs, which exhibit elevated mitochondrial membrane potential (ΔΨm), lower levels of endogenous reduced glutathione, excessive mitochondrial ROS (mROS) levels, and increased apoptosis compared to wild-type (WT) littermates. In addition, we report that glutathione replenishment by glutathione-reduced ethyl ester (GSH-EE) corrects the excessive mROS levels and attenuates the enhanced apoptosis in AS NPCs. Studying the glutathione redox imbalance and mitochondrial abnormalities in embryonic AS NPCs provides an essential insight into the involvement of UBE3A in early neural development, information that can serve as a powerful avenue towards a broader view of AS pathogenesis. Moreover, since mitochondrial dysfunction and elevated ROS levels were associated with other neurodevelopmental disorders, the findings herein suggest some potential shared underlying mechanisms for these disorders as well.


Asunto(s)
Síndrome de Angelman , Células-Madre Neurales , Animales , Síndrome de Angelman/genética , Síndrome de Angelman/patología , Especies Reactivas de Oxígeno , Células-Madre Neurales/patología , Neuronas/patología , Glutatión , Ubiquitina-Proteína Ligasas/genética , Modelos Animales de Enfermedad
7.
Orthopedics ; 46(4): e223-e229, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36779733

RESUMEN

We sought to examine the modern surgical treatment of spinal deformity associated with sister imprinting disorders, Prader-Willi syndrome (PWS) and Angelman syndrome (AS), with emphasis on the specific complications encountered in these patient populations. Fifteen patients with PWS and 5 patients with AS who underwent surgical intervention for spinal deformity between 2000 and 2018 were identified. Postoperative complications were classified using the modified Clavien-Dindo-Sink (CDS) system and further categorized into specific subtypes including excessive drainage, dehiscence, implant failure, infection, and delayed wound healing. Perioperative and final follow-up radiographic data were analyzed. Mean age at surgery was 12.9 years (range, 4-21 years) with mean follow-up of 46.1 months (range, 1-145 months). There were postoperative complications in 17 patients (85%). Ten major complications (CDS ≥ 3) occurred in 9 patients (45%). These included 5 infections requiring reoperation, 1 seroma requiring drainage, 2 severe cervical-thoracic deformities requiring reoperation, 1 implant failure requiring reoperation, and 1 death secondary to fungal sepsis and thromboembolic disease. Eight additional patients (40%) had minor complications (CDS 1 or 2). Eight intraoperative complications occurred in 5 patients (25%), including loss of neuromonitoring signals and cerebrospinal fluid leaks. Surgical intervention for scoliosis in PWS and AS continues to have high complication rates secondary to medical and behavioral comorbidities found in these patient populations. The exact etiology of the high complication rates encountered cannot be definitively stated, but both syndromes frequently present with a number of unique features that may predispose patients to develop surgical complications. [Orthopedics. 2023;46(4):e223-e229.].


Asunto(s)
Síndrome de Angelman , Síndrome de Prader-Willi , Escoliosis , Humanos , Lactante , Síndrome de Prader-Willi/complicaciones , Síndrome de Prader-Willi/cirugía , Escoliosis/cirugía , Complicaciones Posoperatorias/epidemiología
8.
Mol Ther ; 31(7): 2286-2295, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-36805082

RESUMEN

Angelman syndrome (AS) is a rare neurodevelopmental disorder caused by loss of function mutations in maternally expressed UBE3A. No gene-specific treatment is available for patients so far. Although intact and transcriptionally active, paternally inherited UBE3A is silenced by elongation of antisense long noncoding RNA UBE3A-ATS in neurons. Here, we demonstrated that RNA targeting of paternal Ube3a-ATS with a high-fidelity CRISPR-Cas13 (hfCas13x.1) system could restore Ube3a expression to similar levels as that of maternal Ube3a in the cultured mouse neurons. Furthermore, injection into lateral ventricles with neuron-specific hSyn1 promoter-driven hfCas13x.1 packaged in adeno-associated virus (AAV-PHP.eb) could restore paternal Ube3a expression in cortex and hippocampus of neonatal AS mice for up to 4 months after treatment. Behavioral tests showed that expression of paternal Ube3a significantly alleviated AS-related symptoms, including obesity and motor function. Our results suggested that hfCas13x.1-mediated suppression of the Ube3a-ATS lncRNA potentially serves as a promising targeted intervention for AS.


Asunto(s)
Síndrome de Angelman , Animales , Ratones , Síndrome de Angelman/genética , Síndrome de Angelman/terapia , ARN sin Sentido/genética , Obesidad , Ubiquitina-Proteína Ligasas/genética
9.
Mol Ther ; 31(4): 1088-1105, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36641623

RESUMEN

Angelman syndrome (AS) is a neurogenetic disorder caused by the loss of ubiquitin ligase E3A (UBE3A) gene expression in the brain. The UBE3A gene is paternally imprinted in brain neurons. Clinical features of AS are primarily due to the loss of maternally expressed UBE3A in the brain. A healthy copy of paternal UBE3A is present in the brain but is silenced by a long non-coding antisense transcript (UBE3A-ATS). Here, we demonstrate that an artificial transcription factor (ATF-S1K) can silence Ube3a-ATS in an adult mouse model of Angelman syndrome (AS) and restore endogenous physiological expression of paternal Ube3a. A single injection of adeno-associated virus (AAV) expressing ATF-S1K (AAV-S1K) into the tail vein enabled whole-brain transduction and restored UBE3A protein in neurons to ∼25% of wild-type protein. The ATF-S1K treatment was highly specific to the target site with no detectable inflammatory response 5 weeks after AAV-S1K administration. AAV-S1K treatment of AS mice showed behavioral rescue in exploratory locomotion, a task involving gross and fine motor abilities, similar to low ambulation and velocity in AS patients. The specificity and tolerability of a single injection of AAV-S1K therapy for AS demonstrate the use of ATFs as a promising translational approach for AS.


Asunto(s)
Síndrome de Angelman , Animales , Ratones , Síndrome de Angelman/genética , Síndrome de Angelman/terapia , Síndrome de Angelman/metabolismo , Encéfalo/metabolismo , Regulación de la Expresión Génica , Factores de Transcripción/genética , Fenotipo , Ubiquitina-Proteína Ligasas/genética
10.
J Am Acad Child Adolesc Psychiatry ; 62(5): 518-557, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36007813

RESUMEN

OBJECTIVE: To summarize the current state of the literature regarding emotion dysregulation (ED) in syndromic intellectual disabilities (S-IDs) in 6 of the most common forms of S-IDs-Down syndrome, fragile X syndrome (FXS), tuberous sclerosis complex, Williams syndrome, Prader-Willi syndrome, and Angelman syndrome-and to determine future research directions for identification and treatment of ED. METHOD: PubMed bibliographic database was searched from date of inception to May 2021. PRISMA 2020 guidelines were followed with the flowchart, table of included studies, list of excluded studies, and checklist provided. Filters applied included human research and English. Only original research articles were included in the final set, but review articles were used to identify secondary citations of primary studies. All articles were reviewed for appropriateness by 2 authors and summarized. Inclusion criteria were met by 145 articles (Down syndrome = 29, FXS = 55, tuberous sclerosis complex = 11, Williams syndrome = 18, Prader-Willi syndrome = 24, Angelman syndrome = 8). RESULTS: Each syndrome review was summarized separately and further subdivided into articles related to underlying neurobiology, behaviors associated with ED, assessment, and targeted intervention. FXS had the most thorough research base, followed by Down syndrome and Prader-Willi syndrome, with the other syndromes having more limited available research. Very limited research was available regarding intervention for all disorders except FXS. CONCLUSION: Core underlying characteristics of S-IDs appear to place youth at higher risk for ED, but further research is needed to better assess and treat ED in S-IDs. Future studies should have a standard assessment measure of ED, such as the Emotion Dysregulation Inventory, and explore adapting established curricula for ED from the neurotypical and autism spectrum disorder fields.


Asunto(s)
Síndrome de Angelman , Trastorno del Espectro Autista , Síndrome de Down , Síndrome del Cromosoma X Frágil , Discapacidad Intelectual , Síndrome de Prader-Willi , Esclerosis Tuberosa , Síndrome de Williams , Niño , Adolescente , Humanos , Síndrome de Prader-Willi/complicaciones , Síndrome de Prader-Willi/psicología , Síndrome de Angelman/complicaciones , Síndrome de Down/complicaciones , Trastorno del Espectro Autista/complicaciones , Discapacidades del Desarrollo , Esclerosis Tuberosa/complicaciones , Discapacidad Intelectual/etiología , Síndrome del Cromosoma X Frágil/complicaciones , Síndrome del Cromosoma X Frágil/psicología , Síndrome de Williams/complicaciones , Síndrome de Williams/psicología , Emociones
11.
Neurotherapeutics ; 19(4): 1329-1339, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35534672

RESUMEN

The rare genetic neurodevelopmental disease Angelman syndrome (AS) is caused by the loss of function of UBE3A, a ubiquitin ligase. The disease results in a lifetime of severe symptoms, including intellectual disability and motor impairments for which there are no effective treatments. One avenue of treatment for AS is the use of gene therapy to reintroduce a functional copy of the UBE3A gene. Our group had previously shown that recombinant adeno-associated virus (rAAV) expressing mouse Ube3a could rescue deficits in a mouse model of AS. Here, we expand on this work and show that this approach could be successfully replicated in a second AS model using the human UBE3A gene. Furthermore, we address the challenge of limited vector distribution in the brain by developing a novel modified form of UBE3A. This modified protein, termed STUB, was designed with a secretion signal and a cell-penetrating peptide. This allowed transduced cells to act as factories for the production of UBE3A protein that could be taken up by neighboring non-transduced cells, thus increasing the number of neurons receiving the therapeutic protein. Combining this construct with intracerebroventricular injections to maximize rAAV distribution within the brain, we demonstrate that this novel approach improves the recovery of behavioral and electrophysiological deficits in the AS rat model. More importantly, a comparison of rAAV-STUB to a rAAV expressing the normal human UBE3A gene showed that STUB was a more effective therapeutic. These data suggest that rAAV-STUB is a new potential approach for the treatment of AS.


Asunto(s)
Síndrome de Angelman , Péptidos de Penetración Celular , Ubiquitina-Proteína Ligasas , Animales , Humanos , Ratones , Ratas , Síndrome de Angelman/genética , Síndrome de Angelman/terapia , Péptidos de Penetración Celular/genética , Terapia Genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/genética
12.
Eur J Med Genet ; 65(4): 104456, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35218942

RESUMEN

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by loss of expression of the maternally-inherited UBE3A on chromosome 15q11.2. In AS due to a chromosomal deletion that encompasses UBE3A, paternal uniparental disomy of chromosome 15, or imprinting defects (ImpD), the SNRPN locus is unmethylated, while in neurotypical individuals, it is ∼50% methylated. We present the developmental profile of two adults with mild AS assessed using standardized behavioral and neurodevelopmental measures. Both had intellectual disability with unusually advanced verbal communication skills compared to other individuals with AS. Methylation of the SNRPN locus was examined using Methylation Specific Quantitative Melt Analysis (MS-QMA) in different tissues at one time point for participant A (22 years) and two time points for participant B (T1: 22 years, T2: 25 years), and these levels were compared to a typical AS cohort. While participant A showed methylation levels comparable to the typical AS cohort, participant B showed methylation mosaicism in all tissues at both time points and changes in methylation levels from T1 to T2. AS should be considered in individuals with intellectual disability and verbal speech who may not have the typical symptoms of AS.


Asunto(s)
Síndrome de Angelman , Adulto , Síndrome de Angelman/genética , Cromosomas Humanos Par 15/genética , Metilación de ADN , Impresión Genómica , Humanos , Mosaicismo , Disomía Uniparental , Proteínas Nucleares snRNP/genética
13.
Ann Hum Genet ; 86(2): 71-79, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34779508

RESUMEN

Angelman syndrome (AS) (OMIM#105830) is an imprinting disorder caused due to alterations in the maternal chr 15q11-13 region. Majority of cases can be diagnosed by methylation-specific polymerase chain reaction (MS-PCR) of SNRPN gene and by UBE3A sequencing, however, about 10% of cases with AS phenotype remain undiagnosed. Differential diagnoses of AS can be detected by chromosomal microarray (CMA) and clinical exome sequencing (CES). In this study, 30 cases with AS features were evaluated by MS-PCR, CMA, and CES. SNRPN MS-PCR confirmed AS in eight (26%), CMA and CES diagnosed nine (30%) cases. One case was identified with a novel variant c.1125C > T in GABRG3, located at 15q12 region, which is currently not associated with any syndrome. The GABRG3 gene is also speculated to be imprinted, a MS-PCR assay was designed to confirm its differential parental methylation status. This assay identified another case with altered GABRG3 methylation. The two cases with GABRG3 alteration-sequence change and methylation indicate that GABRG3 may be associated with a subtype of AS or a new related syndrome. Performing GABRG3 MS-PCR and sequencing of a larger group of patients with AS phenotype and normal SNPRN and UBE3A status will help in establishing exact genotype-phenotype correlation.


Asunto(s)
Síndrome de Angelman , Receptores de GABA-A , Síndrome de Angelman/diagnóstico , Síndrome de Angelman/genética , Metilación de ADN , Impresión Genómica , Humanos , Fenotipo , Receptores de GABA-A/genética , Proteínas Nucleares snRNP/genética
14.
Cell Rep Med ; 2(8): 100360, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34467244

RESUMEN

Angelman syndrome (AS) is a neurodevelopmental disorder caused by the loss of maternal UBE3A, a ubiquitin protein ligase E3A. Here, we study neurons derived from patients with AS and neurotypical individuals, and reciprocally modulate UBE3A using antisense oligonucleotides. Unbiased proteomics reveal proteins that are regulated by UBE3A in a disease-specific manner, including PEG10, a retrotransposon-derived GAG protein. PEG10 protein increase, but not RNA, is dependent on UBE3A and proteasome function. PEG10 binds to both RNA and ataxia-associated proteins (ATXN2 and ATXN10), localizes to stress granules, and is secreted in extracellular vesicles, modulating vesicle content. Rescue of AS patient-derived neurons by UBE3A reinstatement or PEG10 reduction reveals similarity in transcriptome changes. Overexpression of PEG10 during mouse brain development alters neuronal migration, suggesting that it can affect brain development. These findings imply that PEG10 is a secreted human UBE3A target involved in AS pathophysiology.


Asunto(s)
Síndrome de Angelman/metabolismo , Síndrome de Angelman/fisiopatología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al ADN/metabolismo , Productos del Gen gag/química , Proteínas de Unión al ARN/metabolismo , Retroviridae/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Movimiento Celular , Preescolar , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestructura , Femenino , Humanos , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Dominios Proteicos , Retroelementos/genética , Gránulos de Estrés/metabolismo , Gránulos de Estrés/ultraestructura , Transcriptoma/genética
15.
Expert Opin Investig Drugs ; 30(7): 709-720, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34112038

RESUMEN

Introduction: Angelman syndrome is a rare genetic neurodevelopmental disorder, caused by deficiency or abnormal function of the maternal ubiquitin protein-ligase E3A, known as UBE3A, in the central nervous system. There is no disease-modifying treatment available, but the therapeutic pipeline of Angelman syndrome includes at least 15 different approaches at preclinical or clinical development. In the coming years, several clinical trials will be enrolling patients, which prompted this comprehensive review.Areas covered: We summarize and critically review the different therapeutic approaches. Some approaches attempt to restore the missing or nonfunctional UBE3A protein in the neurons via gene replacement or enzyme replacement therapies. Other therapies aim to induce expression of the normal paternal copy of the UBE3A gene by targeting a long non-coding RNA, the UBE3A-ATS, which interferes with its own expression. Another therapeutic category includes compounds that target molecular pathways and effector proteins known to be involved in Angelman syndrome pathophysiology.Expert opinion: We believe that by 2022-2023, more than five disease-modifying treatments will be simultaneously at clinical testing. However, the are several challenges with regards to safety and efficacy, which need to be addressed. Additionally, there is still a significant unmet need for clinical trial readiness.


Asunto(s)
Síndrome de Angelman/terapia , Terapia Genética/métodos , Ubiquitina-Proteína Ligasas/genética , Síndrome de Angelman/genética , Síndrome de Angelman/fisiopatología , Animales , Terapia de Reemplazo Enzimático/métodos , Humanos
16.
Hum Mol Genet ; 30(12): 1067-1083, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-33856035

RESUMEN

Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by impaired communication skills, ataxia, motor and balance deficits, intellectual disabilities, and seizures. The genetic cause of AS is the neuronal loss of UBE3A expression in the brain. A novel approach, described here, is a stem cell gene therapy which uses lentivector-transduced hematopoietic stem and progenitor cells to deliver functional UBE3A to affected cells. We have demonstrated both the prevention and reversal of AS phenotypes upon transplantation and engraftment of human CD34+ cells transduced with a Ube3a lentivector in a novel immunodeficient Ube3amat-/pat+ IL2rg-/y mouse model of AS. A significant improvement in motor and cognitive behavioral assays as well as normalized delta power measured by electroencephalogram was observed in neonates and adults transplanted with the gene modified cells. Human hematopoietic profiles observed in the lymphoid organs by detection of human immune cells were normal. Expression of UBE3A was detected in the brains of the adult treatment group following immunohistochemical staining illustrating engraftment of the gene-modified cells expressing UBE3A in the brain. As demonstrated with our data, this stem cell gene therapy approach offers a promising treatment strategy for AS, not requiring a critical treatment window.


Asunto(s)
Síndrome de Angelman/terapia , Terapia Genética , Discapacidad Intelectual/terapia , Convulsiones/terapia , Ubiquitina-Proteína Ligasas/genética , Síndrome de Angelman/genética , Síndrome de Angelman/patología , Animales , Antígenos CD34/genética , Ataxia/genética , Ataxia/patología , Encéfalo/metabolismo , Encéfalo/patología , Disfunción Cognitiva/genética , Disfunción Cognitiva/terapia , Modelos Animales de Enfermedad , Electroencefalografía , Regulación de la Expresión Génica/genética , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Humanos , Discapacidad Intelectual/genética , Interleucina-2/genética , Lentivirus/genética , Ratones , Trastornos de la Destreza Motora/genética , Trastornos de la Destreza Motora/patología , Trastornos de la Destreza Motora/terapia , Convulsiones/genética
17.
JBJS Case Connect ; 11(2)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33878039

RESUMEN

CASE: Although scoliosis is common in Angelman syndrome (AS), there are no reports of hip dislocation as a complication of scoliosis surgery. We experienced a case of hip dislocation after posterior spinal instrumentation and fusion from T4 to L5 because of scoliosis progression in an adolescent girl with AS. Postoperatively, posterior dislocation of the left hip occurred; after closed reduction, instability remained. Thus, periacetabular osteotomy was performed (left hip), with good clinical outcomes. CONCLUSION: Especially in cases of abnormal muscle tone, a loss of spinal flexibility because of extensive spinal fixation may result in secondary pathologies of the hip joint.


Asunto(s)
Síndrome de Angelman , Luxación de la Cadera , Escoliosis , Adolescente , Síndrome de Angelman/complicaciones , Síndrome de Angelman/cirugía , Femenino , Luxación de la Cadera/complicaciones , Luxación de la Cadera/cirugía , Humanos , Osteotomía , Escoliosis/complicaciones , Escoliosis/cirugía
18.
Mol Neurobiol ; 58(6): 2543-2557, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33464534

RESUMEN

Angelman syndrome (AS) is a neurogenetic disorder involving ataxia and motor dysfunction, resulting from the absence of the maternally inherited functional Ube3a protein in neurons. Since adenosine A2A receptor (A2AR) blockade relieves synaptic and motor impairments in Parkinson's or Machado-Joseph's diseases, we now tested if A2AR blockade was also effective in attenuating motor deficits in an AS (Ube3am-/p+) mouse model and if this involved correction of synaptic alterations in striatum and cerebellum. Chronic administration of the A2AR antagonist SCH58261 (0.1 mg/kg/day, ip) promoted motor learning of AS mice in the accelerating-rotarod task and rescued the grip strength impairment of AS animals. These motor impairments were accompanied by synaptic alterations in cerebellum and striatum typified by upregulation of synaptophysin and vesicular GABA transporters (vGAT) in the cerebellum of AS mice along with a downregulation of vGAT, vesicular glutamate transporter 1 (vGLUT1) and the dopamine active transporter in AS striatum. Notably, A2AR blockade prevented the synaptic alterations found in AS mice cerebellum as well as the downregulation of striatal vGAT and vGLUT1. This provides the first indications that A2AR blockade may counteract the characteristic motor impairments and synaptic changes of AS, although more studies are needed to unravel the underlying mechanisms.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Síndrome de Angelman/patología , Síndrome de Angelman/fisiopatología , Cerebelo/fisiopatología , Cuerpo Estriado/fisiopatología , Actividad Motora , Receptor de Adenosina A2A/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Fuerza de la Mano , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neurotransmisores/metabolismo , Fosforilación/efectos de los fármacos , Pirimidinas/farmacología , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Triazoles/farmacología , Regulación hacia Arriba/efectos de los fármacos
19.
Brain Dev ; 43(1): 32-44, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32893075

RESUMEN

Angelman Syndrome (AS) is characterized by severe developmental delays including marked speech impairment, movement abnormalities(ataxia, tremor), and unique behaviors such as frequent laughter and is caused by dysfunctional maternal UBE3A gene (maternal 15q11-13 deletions, maternal specific UBE3A mutation, uniparental disomy, and imprinting defect). Intractable epileptic seizures since early childhood with characteristic EEG abnormalities are present in 80-90% patients with AS. Underlying pathophysiology may involve neocortical and thalamocortical hyperexcitability secondary to severe reduction of GABAergic input, as well as dysfunctional synaptic plasticity, deficient synaptogenesis, and neuronal morphological immaturity. The onset of epilepsy is most prevalent between 1 and 3 years of age; however, approximately 25% of patients developed epilepsy before one year of age. Various types of generalized seizures are most prevalent, with most common types are myoclonic and atypical absence.More than 95% of epilepsy patients may have daily seizures at least for a limited time during early childhood, and two-third patients develop disabling seizures. Fever provoked seizures, and frequent occurrence of nonconvulsive status epilepticus are two unique features. Seizures are frequently pharmacoresistant. Considering underlying prominent GABAergic dysfunction, clinicians had used AEDs that target GABAergic signaling such as valproate, phenobarbital, and clonazepam as first-line therapies for AS. However, due to the unfavorable side effect profile of these AEDs, a recent treatment approach involves priority use of levetiracetam, clobazam, topiramate, lamotrigine, ethosuximide, VNS, and carbohydrate-restricted diets. Besides symptomatic management, there has been recent progress to find a curative treatment with the following approaches: 1. Gene/protein replacement therapy (Adeno and lentiviral vector therapy to deliver a gene or secretory protein); 2. Activation of the intact but silent paternal copy of UBE3A (antisense oligonucleotide therapy and artificial transcription factors); and 3. Downstream therapies (OV101/gaboxadol, ketone supplement, novel compounds/peptides, anti-inflammatory/regenerative therapy).


Asunto(s)
Síndrome de Angelman/complicaciones , Síndrome de Angelman/fisiopatología , Epilepsia/fisiopatología , Anticonvulsivantes/uso terapéutico , Niño , Preescolar , Electroencefalografía , Epilepsia/terapia , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Convulsiones/fisiopatología , Convulsiones/terapia , Estado Epiléptico/tratamiento farmacológico , Ubiquitina-Proteína Ligasas/metabolismo , Ácido Valproico/uso terapéutico
20.
Neurobiol Dis ; 148: 105180, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33212289

RESUMEN

BACKGROUND: Angelman syndrome (AS) is a genetic neurodevelopmental disorder caused by the loss of function of the UBE3A protein in the brain. In a previous study, we showed that activity-dependent calcium dynamics in hippocampal CA1 pyramidal neurons of AS mice is compromised, and its normalization rescues the hippocampal-dependent deficits. Therefore, we expected that the expression profiles of calcium-related genes would be altered in AS mice hippocampi. METHODS: We analyzed mRNA sequencing data from AS model mice and WT controls in light of the newly published CaGeDB database of calcium-related genes. We validated our results in two independent RNA sequencing datasets from two additional different AS models: first one, a human neuroblastoma cell line where UBE3A expression was knocked down by siRNA, and the second, an iPSC-derived neurons from AS patient and healthy donor control. FINDINGS: We found signatures of dysregulated calcium-related genes in AS mouse model hippocampus. Additionally, we show that these calcium-related genes function as signatures for AS in other human cellular models of AS, thus strengthening our findings. INTERPRETATION: Our findings suggest the downstream implications and significance of the compromised calcium signaling in Angelman syndrome. Moreover, since AS share similar features with other autism spectrum disorders, we believe that these findings entail meaningful data and approach for other neurodevelopmental disorders, especially those with known alterations of calcium signaling. FUNDING: This work was supported by the Angelman Syndrome Foundation and by the Israel Science Foundation, Grant Number 248/20.


Asunto(s)
Síndrome de Angelman/genética , Señalización del Calcio/genética , Calcio/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Síndrome de Angelman/metabolismo , Animales , Línea Celular Tumoral , Biología Computacional , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Ratones , Neuroblastoma/metabolismo , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Factores Sexuales , Transcriptoma , Ubiquitina-Proteína Ligasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA