Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
mSystems ; 8(1): e0060822, 2023 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-36598241

RESUMEN

A large subset of patients with Angelman syndrome (AS) suffer from concurrent gastrointestinal (GI) issues, including constipation, poor feeding, and reflux. AS is caused by the loss of ubiquitin ligase E3A (UBE3A) gene expression in the brain. Clinical features of AS, which include developmental delays, intellectual disability, microcephaly, and seizures, are primarily due to the deficient expression or function of the maternally inherited UBE3A allele. The association between neurodevelopmental delay and GI disorders is part of the increasing evidence suggesting a link between the brain and the gut microbiome via the microbiota-gut-brain axis. To investigate the associations between colonization of the gut microbiota in AS, we characterized the fecal microbiome in three animal models of AS involving maternal deletions of Ube3A, including mouse, rat, and pig, using 16S rRNA amplicon sequencing. Overall, we identified changes in bacterial abundance across all three animal models of AS. Specific bacterial groups were significantly increased across all animal models, including Lachnospiraceae Incertae sedis, Desulfovibrios sp., and Odoribacter, which have been correlated with neuropsychiatric disorders. Taken together, these findings suggest that specific changes to the local environment in the gut are driven by a Ube3a maternal deletion, unaffected by varying housing conditions, and are prominent and detectable across multiple small and large animal model species. These findings begin to uncover the underlying mechanistic causes of GI disorders in AS patients and provide future therapeutic options for AS patients. IMPORTANCE Angelman syndrome (AS)-associated gastrointestinal (GI) symptoms significantly impact quality of life in patients. In AS models in mouse, rat, and pig, AS animals showed impaired colonization of the gut microbiota compared to wild-type (healthy) control animals. Common changes in AS microbiomes across all three animal models may play a causal effect for GI symptoms and may help to identify ways to treat these comorbidities in patients in the future.


Asunto(s)
Síndrome de Angelman , Enfermedades Gastrointestinales , Microbioma Gastrointestinal , Ratones , Ratas , Animales , Porcinos , Síndrome de Angelman/genética , Microbioma Gastrointestinal/genética , ARN Ribosómico 16S/genética , Calidad de Vida , Modelos Animales de Enfermedad , Ubiquitina-Proteína Ligasas/genética
3.
Hum Mol Genet ; 31(18): 3032-3050, 2022 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-35445702

RESUMEN

Many neurodevelopmental disorders (NDDs) are the result of mutations on the X chromosome. One severe NDD resulting from mutations on the X chromosome is CDKL5 deficiency disorder (CDD). CDD is an epigenetic, X-linked NDD characterized by intellectual disability (ID), pervasive seizures and severe sleep disruption, including recurring hospitalizations. CDD occurs at a 4:1 ratio, with a female bias. CDD is driven by the loss of cyclin-dependent kinase-like 5 (CDKL5), a serine/threonine kinase that is essential for typical brain development, synapse formation and signal transmission. Previous studies focused on male subjects from animal models, likely to avoid the complexity of X mosaicism. For the first time, we report translationally relevant behavioral phenotypes in young adult (8-20 weeks) females and males with robust signal size, including impairments in learning and memory, substantial hyperactivity and increased susceptibility to seizures/reduced seizure thresholds, in both sexes, and in two models of CDD preclinical mice, one with a general loss-of-function mutation and one that is a patient-derived mutation.


Asunto(s)
Quinasas Ciclina-Dependientes , Animales , Cognición , Quinasas Ciclina-Dependientes/deficiencia , Síndromes Epilépticos , Femenino , Humanos , Masculino , Ratones , Proteínas Serina-Treonina Quinasas/genética , Convulsiones/genética , Serina
4.
Neurotherapeutics ; 18(3): 1535-1547, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34528170

RESUMEN

Angelman syndrome (AS) is a rare (~1:15,000) neurodevelopmental disorder characterized by severe developmental delay and intellectual disability, impaired communication skills, and a high prevalence of seizures, sleep disturbances, ataxia, motor deficits, and microcephaly. AS is caused by loss-of-function of the maternally inherited UBE3A gene. UBE3A is located on chromosome 15q11-13 and is biallelically expressed throughout the body but only maternally expressed in the brain due to an RNA antisense transcript that silences the paternal copy. There is currently no cure for AS, but advancements in small molecule drugs and gene therapies offer a promising approach for the treatment of the disorder. Here, we review AS and how loss-of-function of the maternal UBE3A contributes to the disorder. We also discuss the strengths and limitations of current animal models of AS. Furthermore, we examine potential small molecule drug and gene therapies for the treatment of AS and associated challenges faced by the therapeutic design. Finally, gene therapy offers the opportunity for precision medicine in AS and advancements in the treatment of this disorder can serve as a foundation for other single-gene neurodevelopmental disorders.


Asunto(s)
Síndrome de Angelman/genética , Síndrome de Angelman/terapia , Anticonvulsivantes/uso terapéutico , Terapia Genética/métodos , Animales , Productos Biológicos/uso terapéutico , Humanos , Isoxazoles/uso terapéutico , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/terapia , ARN sin Sentido/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Ubiquitina-Proteína Ligasas/genética
5.
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
6.
Genome Med ; 13(1): 69, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33910599

RESUMEN

BACKGROUND: Genes with multiple co-active promoters appear common in brain, yet little is known about functional requirements for these potentially redundant genomic regulatory elements. SCN1A, which encodes the NaV1.1 sodium channel alpha subunit, is one such gene with two co-active promoters. Mutations in SCN1A are associated with epilepsy, including Dravet syndrome (DS). The majority of DS patients harbor coding mutations causing SCN1A haploinsufficiency; however, putative causal non-coding promoter mutations have been identified. METHODS: To determine the functional role of one of these potentially redundant Scn1a promoters, we focused on the non-coding Scn1a 1b regulatory region, previously described as a non-canonical alternative transcriptional start site. We generated a transgenic mouse line with deletion of the extended evolutionarily conserved 1b non-coding interval and characterized changes in gene and protein expression, and assessed seizure activity and alterations in behavior. RESULTS: Mice harboring a deletion of the 1b non-coding interval exhibited surprisingly severe reductions of Scn1a and NaV1.1 expression throughout the brain. This was accompanied by electroencephalographic and thermal-evoked seizures, and behavioral deficits. CONCLUSIONS: This work contributes to functional dissection of the regulatory wiring of a major epilepsy risk gene, SCN1A. We identified the 1b region as a critical disease-relevant regulatory element and provide evidence that non-canonical and seemingly redundant promoters can have essential function.


Asunto(s)
Epilepsia/genética , Regulación de la Expresión Génica , Canal de Sodio Activado por Voltaje NAV1.1/genética , Eliminación de Secuencia/genética , Animales , Atención , Secuencia de Bases , Encéfalo/metabolismo , Encéfalo/patología , Cromatina/metabolismo , Secuencia Conservada/genética , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia/diagnóstico por imagen , Evolución Molecular , Femenino , Células HEK293 , Heterocigoto , Homocigoto , Humanos , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/genética , Ratones Endogámicos C57BL , Neuronas/metabolismo , Prueba de Campo Abierto , Fenotipo , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos/genética , Análisis de Supervivencia , Temperatura , Transactivadores/metabolismo
7.
Neurobiol Learn Mem ; 165: 106874, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-29800646

RESUMEN

Prader-Willi syndrome (PWS) is an imprinted neurodevelopmental disease caused by a loss of paternal genes on chromosome 15q11-q13. It is characterized by cognitive impairments, developmental delay, sleep abnormalities, and hyperphagia often leading to obesity. Clinical research has shown that a lack of expression of SNORD116, a paternally expressed imprinted gene cluster that encodes multiple copies of a small nucleolar RNA (snoRNA) in both humans and mice, is most likely responsible for many PWS symptoms seen in humans. The majority of previous research using PWS preclinical models focused on characterization of the hyperphagic and metabolic phenotypes. However, a crucial understudied clinical phenotype is cognitive impairments and thus we investigated the learning and memory abilities using a model of PWS, with a heterozygous deletion in Snord116. We utilized the novel object recognition task, which doesn't require external motivation, or exhaustive swim training. Automated findings were further confirmed with manual scoring by a highly trained blinded investigator. We discovered deficits in Snord116+/- mutant mice in the novel object recognition, location memory and tone cue fear conditioning assays when compared to age-, sex- matched, littermate control Snord116+/+ mice. Further, we confirmed that despite physical neo-natal developmental delays, Snord116+/- mice had normal exploratory and motor abilities. These results show that the Snord116+/- deletion murine model is a valuable preclinical model for investigating learning and memory impairments in individuals with PWS without common confounding phenotypes.


Asunto(s)
Disfunción Cognitiva/genética , Eliminación de Gen , Síndrome de Prader-Willi/genética , ARN Nucleolar Pequeño/genética , Animales , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Humanos , Ratones , Síndrome de Prader-Willi/complicaciones
8.
Autism Res ; 11(4): 587-601, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29377611

RESUMEN

Mutations in the SHANK3 gene have been discovered in autism spectrum disorder (ASD), and the intellectual disability, Phelan-McDermid Syndrome. This study leveraged a new rat model of Shank3 deficiency to assess complex behavioral phenomena, unique to rats, which display a richer social behavior repertoire than mice. Uniquely detectable emissions of ultrasonic vocalizations (USV) in rats serve as situation-dependent affective signals and accomplish important communicative functions. We report, for the first time, a call and response acoustic playback assay of bidirectional social communication in juvenile Shank3 rats. Interestingly, we found that Shank3-deficient null males did not demonstrate the enhanced social approach behavior typically exhibited following playback of pro-social USV. Concomitantly, we discovered that emission of USV in response to playback was not genotype-dependent and emitted response calls were divergent in meaning. This is the first report of these socially relevant responses using a genetic model of ASD. A comprehensive and empirical analysis of vigorous play during juvenile reciprocal social interactions further revealed fewer bouts and reduced durations of time spent playing by multiple key parameters, including reduced anogenital sniffing and allogrooming. We further discovered that male null Shank3-deficient pups emitted fewer isolation-induced USV than Shank3 wildtype controls. Postnatal whole brain anatomical phenotyping was applied to visualize anatomical substrates that underlie developmental phenotypes. The data presented here lend support for the important role of Shank3 in social communication, the core symptom domain of ASD. By increasing the number of in vivo functional outcome measures, we improved the likelihood for identifying and moving forward with medical interventions. Autism Res 2018, 11: 587-601. © 2018 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Clinically relevant outcomes are required to demonstrate the utility of therapeutics. We introduce findings in a rat model, and assess the impact of mutations in Shank3, an autism risk gene. We found that males with deficient expression of Shank3 did not demonstrate typical responses in a bi-directional social communication test and that social interaction was lower on key parameters. Outcome measures reported herein extend earlier results in mice and capture responses to acoustic calls, which is analogous to measuring receptive and expressive communication.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastornos de los Cromosomas/genética , Comunicación , Modelos Animales de Enfermedad , Modelos Genéticos , Proteínas del Tejido Nervioso/genética , Conducta Social , Factores de Edad , Animales , Deleción Cromosómica , Cromosomas Humanos Par 22/genética , Análisis Mutacional de ADN , Conducta Exploratoria , Eliminación de Gen , Relaciones Interpersonales , Masculino , Fenotipo , Juego e Implementos de Juego , Ratas , Vocalización Animal
9.
Hum Mol Genet ; 26(20): 3995-4010, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29016856

RESUMEN

Maternally derived copy number gains of human chromosome 15q11.2-q13.3 (Dup15q syndrome or Dup15q) cause intellectual disability, epilepsy, developmental delay, hypotonia, speech impairments, and minor dysmorphic features. Dup15q syndrome is one of the most common and penetrant chromosomal abnormalities observed in individuals with autism spectrum disorder (ASD). Although ∼40 genes are located in the 15q11.2-q13.3 region, overexpression of the ubiquitin-protein E3A ligase (UBE3A) gene is thought to be the predominant molecular cause of the phenotypes observed in Dup15q syndrome. The UBE3A gene demonstrates maternal-specific expression in neurons and loss of maternal UBE3A causes Angelman syndrome, a neurodevelopmental disorder with some overlapping neurological features to Dup15q. To directly test the hypothesis that overexpression of UBE3A is an important underlying molecular cause of neurodevelopmental dysfunction, we developed and characterized a mouse overexpressing Ube3a isoform 2 in excitatory neurons. Ube3a isoform 2 is conserved between mouse and human and known to play key roles in neuronal function. Transgenic mice overexpressing Ube3a isoform 2 in excitatory forebrain neurons exhibited increased anxiety-like behaviors, learning impairments, and reduced seizure thresholds. However, these transgenic mice displayed normal social approach, social interactions, and repetitive motor stereotypies that are relevant to ASD. Reduced forebrain, hippocampus, striatum, amygdala, and cortical volume were also observed. Altogether, these findings show neuronal overexpression of Ube3a isoform 2 causes phenotypes translatable to neurodevelopmental disorders.


Asunto(s)
Discapacidad Intelectual/enzimología , Neuronas/enzimología , Ubiquitina-Proteína Ligasas/biosíntesis , Animales , Trastorno de Personalidad Antisocial/genética , Trastorno de Personalidad Antisocial/metabolismo , Ansiedad/genética , Ansiedad/metabolismo , Aberraciones Cromosómicas , Cromosomas Humanos Par 15/enzimología , Cromosomas Humanos Par 15/genética , Cromosomas Humanos Par 15/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Convulsiones/genética , Convulsiones/metabolismo , Ubiquitina-Proteína Ligasas/genética
10.
Nat Neurosci ; 20(8): 1062-1073, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28671691

RESUMEN

The chromatin remodeling gene CHD8 represents a central node in neurodevelopmental gene networks implicated in autism. We examined the impact of germline heterozygous frameshift Chd8 mutation on neurodevelopment in mice. Chd8+/del5 mice displayed normal social interactions with no repetitive behaviors but exhibited cognitive impairment correlated with increased regional brain volume, validating that phenotypes of Chd8+/del5 mice overlap pathology reported in humans with CHD8 mutations. We applied network analysis to characterize neurodevelopmental gene expression, revealing widespread transcriptional changes in Chd8+/del5 mice across pathways disrupted in neurodevelopmental disorders, including neurogenesis, synaptic processes and neuroimmune signaling. We identified a co-expression module with peak expression in early brain development featuring dysregulation of RNA processing, chromatin remodeling and cell-cycle genes enriched for promoter binding by Chd8, and we validated increased neuronal proliferation and developmental splicing perturbation in Chd8+/del5 mice. This integrative analysis offers an initial picture of the consequences of Chd8 haploinsufficiency for brain development.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Haploinsuficiencia/genética , Animales , Encéfalo/metabolismo , Proteínas de Ciclo Celular/genética , Cromatina/metabolismo , Ratones Transgénicos , Mutación/genética , Fenotipo , Factores de Transcripción/genética
11.
Mol Autism ; 8: 26, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28638591

RESUMEN

BACKGROUND: Autism spectrum disorder (ASD) is a clinically and biologically heterogeneous condition characterized by social, repetitive, and sensory behavioral abnormalities. No treatments are approved for the core diagnostic symptoms of ASD. To enable the earliest stages of therapeutic discovery and development for ASD, robust and reproducible behavioral phenotypes and biological markers are essential to establish in preclinical animal models. The goal of this study was to identify electroencephalographic (EEG) and behavioral phenotypes that are replicable between independent cohorts in a mouse model of ASD. The larger goal of our strategy is to empower the preclinical biomedical ASD research field by generating robust and reproducible behavioral and physiological phenotypes in animal models of ASD, for the characterization of mechanistic underpinnings of ASD-relevant phenotypes, and to ensure reliability for the discovery of novel therapeutics. Genetic disruption of the SHANK3 gene, a scaffolding protein involved in the stability of the postsynaptic density in excitatory synapses, is thought to be responsible for a relatively large number of cases of ASD. Therefore, we have thoroughly characterized the robustness of ASD-relevant behavioral phenotypes in two cohorts, and for the first time quantified translational EEG activity in Shank3B null mutant mice. METHODS: In vivo physiology and behavioral assays were conducted in two independently bred and tested full cohorts of Shank3B null mutant (Shank3B KO) and wildtype littermate control (WT) mice. EEG was recorded via wireless implanted telemeters for 7 days of baseline followed by 20 min of recording following pentylenetetrazol (PTZ) challenge. Behaviors relevant to the diagnostic and associated symptoms of ASD were tested on a battery of established behavioral tests. Assays were designed to reproduce and expand on the original behavioral characterization of Shank3B KO mice. Two or more corroborative tests were conducted within each behavioral domain, including social, repetitive, cognitive, anxiety-related, sensory, and motor categories of assays. RESULTS: Relative to WT mice, Shank3B KO mice displayed a dramatic resistance to PTZ seizure induction and an enhancement of gamma band oscillatory EEG activity indicative of enhanced inhibitory tone. These findings replicated in two separate cohorts. Behaviorally, Shank3B KO mice exhibited repetitive grooming, deficits in aspects of reciprocal social interactions and vocalizations, and reduced open field activity, as well as variable deficits in sensory responses, anxiety-related behaviors, learning and memory. CONCLUSIONS: Robust animal models and quantitative, replicable biomarkers of neural dysfunction are needed to decrease risk and enable successful drug discovery and development for ASD and other neurodevelopmental disorders. Complementary to the replicated behavioral phenotypes of the Shank3B mutant mouse is the new identification of a robust, translational in vivo neurophysiological phenotype. Our findings provide strong evidence for robustness and replicability of key translational phenotypes in Shank3B mutant mice and support the usefulness of this mouse model of ASD for therapeutic discovery.


Asunto(s)
Ansiedad/fisiopatología , Trastorno Autístico/fisiopatología , Conducta Animal , Modelos Animales de Enfermedad , Memoria , Proteínas del Tejido Nervioso/genética , Animales , Ansiedad/diagnóstico , Ansiedad/genética , Trastorno Autístico/diagnóstico , Trastorno Autístico/genética , Biomarcadores/análisis , Convulsivantes/administración & dosificación , Electroencefalografía , Femenino , Aseo Animal , Humanos , Relaciones Interpersonales , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Noqueados , Proteínas de Microfilamentos , Proteínas del Tejido Nervioso/deficiencia , Pentilenotetrazol/administración & dosificación , Reproducibilidad de los Resultados , Convulsiones/inducido químicamente , Convulsiones/genética , Convulsiones/fisiopatología
12.
Neuroscience ; 345: 155-165, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-27189882

RESUMEN

SHANK3 is a synaptic scaffolding protein localized in the postsynaptic density and has a crucial role in synaptogenesis and neural physiology. Deletions and point mutations in SHANK3 cause Phelan-McDermid Syndrome (PMS), and have also been implicated in autism spectrum disorder (ASD) and intellectual disabilities, leading to the hypothesis that reduced SHANK3 expression impairs basic brain functions that are important for social communication and cognition. Several mouse models of Shank3 deletions have been generated, varying in the specific domain deleted. Here we report impairments in cognitive function in mice heterozygous for exon 13-16 (coding for the PDZ domain) deletion. The touchscreen pairwise discrimination task was chosen by virtue of its: (a) conceptual and technical similarities to the Cambridge Neuropsychological Test Automated Battery (CANTAB) and NIH Toolbox Cognition Battery used for testing cognitive functions in humans, (b) minimal demand on motor abilities, and (c) capability to measure many aspects of learning and memory and complex cognitive functions, including cognitive flexibility. The similarity between our mouse tasks and human cognitive assays means a high translational validity in future intervention studies using preclinical models. Our study revealed that Shank3B heterozygous mice (+/-) were slower to reach criterion in the pairwise visual discrimination task, and exhibited trends toward making more errors (first trial errors) and more correction errors than wildtype mice (+/+). Open field activity was normal in +/-, ruling out hypo- or hyperactivity as potential confounds in the touchscreen test. Sociability in the three chamber test was also normal in both +/+ and +/-. These results indicate a deficit in discrimination learning in the Shank3B model of PMS and ASD, suggesting that this mouse model is a useful preclinical tool for studying neurobiological mechanisms behind cognitive impairments in PMS and ASD. The current findings are the starting point for our future research in which we will investigate multiple domains of cognition and explore pharmacological interventions.


Asunto(s)
Aprendizaje por Asociación/fisiología , Trastorno Autístico/metabolismo , Trastornos de los Cromosomas/metabolismo , Discapacidades para el Aprendizaje/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Conducta Social , Animales , Trastorno Autístico/psicología , Deleción Cromosómica , Trastornos de los Cromosomas/psicología , Cromosomas Humanos Par 22/metabolismo , Discriminación en Psicología/fisiología , Modelos Animales de Enfermedad , Masculino , Ratones Transgénicos , Proteínas de Microfilamentos , Actividad Motora/fisiología , Proteínas del Tejido Nervioso/genética , Pruebas Neuropsicológicas , Percepción Visual/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...