Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Eur J Neurosci ; 41(10): 1372-80, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25864922

RESUMEN

The Reelin signaling pathway is implicated in processes controlling synaptic plasticity and hippocampus-dependent learning and memory. A single direct in vivo application of Reelin enhances long-term potentiation, increases dendritic spine density and improves associative and spatial learning and memory. Angelman syndrome (AS) is a neurological disorder that presents with an overall defect in synaptic function, including decreased long-term potentiation, reduced dendritic spine density, and deficits in learning and memory, making it an attractive model in which to examine the ability of Reelin to recover synaptic function and cognitive deficits. In this study, we investigated the effects of Reelin administration on synaptic plasticity and cognitive function in a mouse model of AS and demonstrated that bilateral, intraventricular injections of Reelin recover synaptic function and corresponding hippocampus-dependent associative and spatial learning and memory. Additionally, we describe alteration of the Reelin profile in tissue from both the AS mouse and post-mortem human brain.


Asunto(s)
Síndrome de Angelman/fisiopatología , Síndrome de Angelman/psicología , Moléculas de Adhesión Celular Neuronal/administración & dosificación , Proteínas de la Matriz Extracelular/administración & dosificación , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Proteínas del Tejido Nervioso/administración & dosificación , Serina Endopeptidasas/administración & dosificación , Síndrome de Angelman/tratamiento farmacológico , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Corteza Cerebral/metabolismo , Espinas Dendríticas/efectos de los fármacos , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Células HEK293 , Hipocampo/fisiopatología , Hipocampo/ultraestructura , Humanos , Inyecciones Intraventriculares , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Proteína Reelina , Serina Endopeptidasas/metabolismo , Aprendizaje Espacial/efectos de los fármacos , Memoria Espacial/efectos de los fármacos
2.
J Neurosci ; 31(45): 16241-50, 2011 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22072675

RESUMEN

The protective/neurotoxic role of fractalkine (CX3CL1) and its receptor CX3C chemokine receptor 1 (CX3CR1) signaling in neurodegenerative disease is an intricate and highly debated research topic and it is becoming even more complicated as new studies reveal discordant results. It appears that the CX3CL1/CX3CR1 axis plays a direct role in neurodegeneration and/or neuroprotection depending on the CNS insult. However, all the above studies focused on the role of CX3CL1/CX3CR1 signaling in pathological conditions, ignoring the relevance of CX3CL1/CX3CR1 signaling under physiological conditions. No approach to date has been taken to decipher the significance of defects in CX3CL1/CX3CR1 signaling in physiological condition. In the present study we used CX3CR1⁻/⁻, CX3CR1⁺/⁻, and wild-type mice to investigate the physiological role of CX3CR1 receptor in cognition and synaptic plasticity. Our results demonstrate for the first time that mice lacking the CX3CR1 receptor show contextual fear conditioning and Morris water maze deficits. CX3CR1 deficiency also affects motor learning. Importantly, mice lacking the receptor have a significant impairment in long-term potentiation (LTP). Infusion with IL-1ß receptor antagonist significantly reversed the deficit in cognitive function and impairment in LTP. Our results reveal that under physiological conditions, disruption in CX3CL1 signaling will lead to impairment in cognitive function and synaptic plasticity via increased action of IL-1ß.


Asunto(s)
Trastornos del Conocimiento/patología , Hipocampo/patología , Hipocampo/fisiopatología , Potenciación a Largo Plazo/genética , Receptores de Interleucina-8A/deficiencia , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Biofisica , Bromodesoxiuridina/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Cerebelo/metabolismo , Trastornos del Conocimiento/genética , Condicionamiento Psicológico/fisiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Estimulación Eléctrica , Ensayo de Inmunoadsorción Enzimática/métodos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Conducta Exploratoria/fisiología , Miedo/fisiología , Regulación de la Expresión Génica/genética , Técnicas In Vitro , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Potenciación a Largo Plazo/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Microglía/patología , Actividad Motora/genética , Neurogénesis/genética , Técnicas de Placa-Clamp , Prueba de Desempeño de Rotación con Aceleración Constante
3.
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
4.
J Neurosci ; 30(50): 17068-78, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21159977

RESUMEN

The vast majority of Alzheimer's disease (AD) cases are late onset with progressive synapse loss and neurodegeneration. Although the amyloid hypothesis has generated great insights into the disease mechanism, several lines of evidence indicate that other risk factors might precondition the brain to amyloid toxicity. Here, we show that the deletion of a major lipoprotein receptor, low-density lipoprotein receptor-related protein 1 (LRP1), in forebrain neurons in mice leads to a global defect in brain lipid metabolism characterized by decreased brain levels of cholesterol, sulfatide, galactosylceramide, and triglyceride. These lipid deficits correlate with progressive, age-dependent dendritic spine degeneration, synapse loss, neuroinflammation, memory loss, and eventual neurodegeneration. We further show that the levels of glutamate receptor subunits NMDA receptor 1 and Glu receptor 1 are selectively reduced in LRP1 forebrain knock-out mice and in LRP1 knockdown neurons, which is partially rescued by restoring neuronal cholesterol. Together, these studies support a critical role for LRP1 in maintaining brain lipid homeostasis and associated synaptic and neuronal integrity, and provide important insights into the pathophysiological mechanisms in AD.


Asunto(s)
Metabolismo de los Lípidos/genética , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Degeneración Nerviosa/metabolismo , Neuronas/patología , Prosencéfalo/metabolismo , Sinapsis/patología , Factores de Edad , Amnesia/patología , Animales , Técnicas de Cultivo de Célula , Espinas Dendríticas/patología , Hipocampo/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Ratones Noqueados , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Neuronas/metabolismo , Prosencéfalo/patología , Receptores AMPA/biosíntesis , Receptores de N-Metil-D-Aspartato/biosíntesis , Sinapsis/metabolismo
5.
Autism Res ; 13(3): 397-409, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31961493

RESUMEN

Angelman syndrome (AS) is a rare genetic disorder characterized by severe intellectual disability, seizures, lack of speech, and ataxia. The gene responsible for AS was identified as Ube3a and it encodes for E6AP, an E3 ubiquitin ligase. Currently, there is very little known about E6AP's mechanism of action in vivo or how the lack of this protein in neurons may contribute to the AS phenotype. Elucidating the mechanistic action of E6AP would enhance our understanding of AS and drive current research into new avenues that could lead to novel therapeutic approaches that target E6AP's various functions. To facilitate the study of AS, we have generated a novel rat model in which we deleted the rat Ube3a gene using CRISPR. The AS rat phenotypically mirrors human AS with loss of Ube3a expression in the brain and deficits in motor coordination as well as learning and memory. This model offers a new avenue for the study of AS. Autism Res 2020, 13: 397-409. © 2020 International Society for Autism Research,Wiley Periodicals, Inc. LAY SUMMARY: Angelman syndrome (AS) is a rare genetic disorder characterized by severe intellectual disability, seizures, difficulty speaking, and ataxia. The gene responsible for AS was identified as UBE3A, yet very little is known about its function in vivo or how the lack of this protein in neurons may contribute to the AS phenotype. To facilitate the study of AS, we have generated a novel rat model in which we deleted the rat Ube3a gene using CRISPR. The AS rat mirrors human AS with loss of Ube3a expression in the brain and deficits in motor coordination as well as learning and memory. This model offers a new avenue for the study of AS.


Asunto(s)
Síndrome de Angelman/genética , Síndrome de Angelman/fisiopatología , Eliminación de Gen , Ubiquitina-Proteína Ligasas/genética , Animales , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Humanos , Memoria , Fenotipo , Ratas , Ratas Sprague-Dawley
6.
Learn Mem ; 15(6): 403-11, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18509114

RESUMEN

cAMP is a critical second messenger implicated in synaptic plasticity and memory in the mammalian brain. Substantial evidence links increases in intracellular cAMP to activation of cAMP-dependent protein kinase (PKA) and subsequent phosphorylation of downstream effectors (transcription factors, receptors, protein kinases) necessary for long-term potentiation (LTP) of synaptic strength. However, cAMP may also initiate signaling via a guanine nucleotide exchange protein directly activated by cAMP (Epac). The role of Epac in hippocampal synaptic plasticity is unknown. We found that in area CA1 of mouse hippocampal slices, activation of Epac enhances maintenance of LTP without affecting basal synaptic transmission. The persistence of this form of LTP requires extracellular signal-regulated protein kinase (ERK) and new protein synthesis, but not transcription. Because ERK is involved in translational control of long-lasting plasticity and memory, our data suggest that Epac is a crucial link between cAMP and ERK during some forms of protein synthesis-dependent LTP. Activation of Epac represents a novel signaling pathway for rapid regulation of the stability of enduring forms of LTP and, perhaps, of hippocampus- dependent long-term memories.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Animales , Butadienos/farmacología , Carbazoles/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , AMP Cíclico/fisiología , Dactinomicina/farmacología , Emetina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Femenino , Factores de Intercambio de Guanina Nucleótido/agonistas , Isoproterenol/farmacología , Ratones , Ratones Endogámicos C57BL , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirroles/farmacología , Transmisión Sináptica/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
7.
J Psychopharmacol ; 27(4): 386-95, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23104248

RESUMEN

The lipoprotein receptor ligand Reelin is important for the processes of normal synaptic plasticity, dendritic morphogenesis, and learning and memory. Heterozygous reeler mice (HRM) show many neuroanatomical, biochemical, and behavioral features that are associated with schizophrenia. HRM show subtle morphological defects including reductions in dendritic spine density, altered synaptic plasticity and behavioral deficits in associative learning and memory and pre-pulse inhibition. The present studies test the hypothesis that in vivo elevation of Reelin levels can rescue synaptic and behavioral phenotypes associated with HRM. We demonstrate that a single in vivo injection of Reelin increases GAD67 expression and alters dendritic spine morphology. In parallel we observed enhancement of hippocampal synaptic function and associative learning and memory. Reelin supplementation also increases pre-pulse inhibition. These results suggest that characteristics of HRM, similar to those observed in schizophrenia, are sensitive to Reelin levels and can be modified with Reelin supplementation in male and female adults.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Trastornos Neurológicos de la Marcha/metabolismo , Discapacidades para el Aprendizaje/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal , Esquizofrenia/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/genética , Cruzamientos Genéticos , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Proteínas de la Matriz Extracelular/genética , Femenino , Trastornos Neurológicos de la Marcha/etiología , Glutamato Descarboxilasa/metabolismo , Heterocigoto , Hipocampo/metabolismo , Aprendizaje , Discapacidades para el Aprendizaje/etiología , Masculino , Ratones , Ratones Mutantes Neurológicos , Proteínas del Tejido Nervioso/genética , Inhibición Neural , Neuronas/metabolismo , Proteína Reelina , Esquizofrenia/patología , Esquizofrenia/fisiopatología , Filtrado Sensorial , Serina Endopeptidasas/genética , Transmisión Sináptica
8.
PLoS One ; 6(12): e27221, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22174738

RESUMEN

Angelman syndrome (AS), a genetic disorder occurring in approximately one in every 15,000 births, is characterized by severe mental retardation, seizures, difficulty speaking and ataxia. The gene responsible for AS was discovered to be UBE3A and encodes for E6-AP, an ubiquitin ligase. A unique feature of this gene is that it undergoes maternal imprinting in a neuron-specific manner. In the majority of AS cases, there is a mutation or deletion in the maternally inherited UBE3A gene, although other cases are the result of uniparental disomy or mismethylation of the maternal gene. While most human disorders characterized by severe mental retardation involve abnormalities in brain structure, no gross anatomical changes are associated with AS. However, we have determined that abnormal calcium/calmodulin-dependent protein kinase II (CaMKII) regulation is seen in the maternal UBE3A deletion AS mouse model and is responsible for the major phenotypes. Specifically, there is an increased αCaMKII phosphorylation at the autophosphorylation sites Thr(286) and Thr(305/306), resulting in an overall decrease in CaMKII activity. CaMKII is not produced until after birth, indicating that the deficits associated with AS are not the result of developmental abnormalities. The present studies are focused on exploring the potential to rescue the learning and memory deficits in the adult AS mouse model through the use of an adeno-associated virus (AAV) vector to increase neuronal UBE3A expression. These studies show that increasing the levels of E6-AP in the brain using an exogenous vector can improve the cognitive deficits associated with AS. Specifically, the associative learning deficit was ameliorated in the treated AS mice compared to the control AS mice, indicating that therapeutic intervention may be possible in older AS patients.


Asunto(s)
Síndrome de Angelman/complicaciones , Trastornos del Conocimiento/complicaciones , Dependovirus/metabolismo , Síndrome de Angelman/fisiopatología , Animales , Ansiedad/fisiopatología , Aprendizaje por Asociación/fisiología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Potenciación a Largo Plazo , Aprendizaje por Laberinto/fisiología , Ratones , Actividad Motora/fisiología , Ubiquitina-Proteína Ligasas/metabolismo
9.
Mol Neurodegener ; 1: 12, 2006 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-16984644

RESUMEN

Selenium is an essential micronutrient that function through selenoproteins. Selenium deficiency results in lower concentrations of selenium and selenoproteins. The brain maintains it's selenium better than other tissues under low-selenium conditions. Recently, the selenium-containing protein selenoprotein P (Sepp) has been identified as a possible transporter of selenium. The targeted disruption of the selenoprotein P gene (Sepp1) results in decreased brain selenium concentration and neurological dysfunction, unless selenium intake is excessive However, the effect of selenoprotein P deficiency on the processes of memory formation and synaptic plasticity is unknown. In the present studies Sepp1(-/-) mice and wild type littermate controls (Sepp1(+/+)) fed a high-selenium diet (1 mg Se/kg) were used to characterize activity, motor coordination, and anxiety as well as hippocampus-dependent learning and memory. Normal associative learning, but disrupted spatial learning was observed in Sepp1(-/-) mice. In addition, severe alterations were observed in synaptic transmission, short-term plasticity and long-term potentiation in hippocampus area CA1 synapses of Sepp1(-/-) mice on a 1 mg Se/kg diet and Sepp1(+/+) mice fed a selenium-deficient (0 mg Se/kg) diet. Taken together, these data suggest that selenoprotein P is required for normal synaptic function, either through presence of the protein or delivery of required selenium to the CNS.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA