Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(31): e2407546121, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39042682

RESUMEN

Fragile X syndrome (FXS) is the most common genetic cause of autism spectrum disorder engendered by transcriptional silencing of the fragile X messenger ribonucleoprotein 1 (FMR1) gene. Given the early onset of behavioral and molecular changes, it is imperative to know the optimal timing for therapeutic intervention. Case reports documented benefits of metformin treatment in FXS children between 2 and 14 y old. In this study, we administered metformin from birth to Fmr1-/y mice which corrected up-regulated mitogen-2 activated protein kinase/extracellular signal-regulated kinase and mammalian/mechanistic target of rapamycin complex 1 signaling pathways and specific synaptic mRNA-binding targets of FMRP. Metformin rescued increased number of calls in ultrasonic vocalization and repetitive behavior in Fmr1-/y mice. Our findings demonstrate that in mice, early-in-life metformin intervention is effective in treating FXS pathophysiology.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil , Metformina , Metformina/farmacología , Metformina/uso terapéutico , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/fisiopatología , Síndrome del Cromosoma X Frágil/metabolismo , Animales , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Ratones , Masculino , Ratones Noqueados , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Modelos Animales de Enfermedad , Transducción de Señal/efectos de los fármacos
2.
Brain ; 146(5): 2175-2190, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36315645

RESUMEN

MAPK interacting protein kinases 1 and 2 (Mnk1/2) regulate a plethora of functions, presumably via phosphorylation of their best characterized substrate, eukaryotic translation initiation factor 4E (eIF4E) on Ser209. Here, we show that, whereas deletion of Mnk1/2 (Mnk double knockout) impairs synaptic plasticity and memory in mice, ablation of phospho-eIF4E (Ser209) does not affect these processes, suggesting that Mnk1/2 possess additional downstream effectors in the brain. Translational profiling revealed only a small overlap between the Mnk1/2- and phospho-eIF4E(Ser209)-regulated translatome. We identified the synaptic Ras GTPase activating protein 1 (Syngap1), encoded by a syndromic autism gene, as a downstream target of Mnk1 because Syngap1 immunoprecipitated with Mnk1 and showed reduced phosphorylation (S788) in Mnk double knockout mice. Knockdown of Syngap1 reversed memory deficits in Mnk double knockout mice and pharmacological inhibition of Mnks rescued autism-related phenotypes in Syngap1+/- mice. Thus, Syngap1 is a downstream effector of Mnk1, and the Mnks-Syngap1 axis regulates memory formation and autism-related behaviours.


Asunto(s)
Trastorno Autístico , Factor 4E Eucariótico de Iniciación , Animales , Ratones , Factor 4E Eucariótico de Iniciación/genética , Ratones Noqueados , Fosforilación , Proteínas Activadoras de ras GTPasa/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(36): 18060-18067, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31427534

RESUMEN

Translational control plays a key role in regulation of neuronal activity and behavior. Deletion of the translational repressor 4E-BP2 in mice alters excitatory and inhibitory synaptic functions, engendering autistic-like behaviors. The contribution of 4E-BP2-dependent translational control in excitatory and inhibitory neurons and astrocytic cells to these behaviors remains unknown. To investigate this, we generated cell-type-specific conditional 4E-BP2 knockout mice and tested them for the salient features of autism, including repetitive stereotyped behaviors (self-grooming and marble burying), sociability (3-chamber social and direct social interaction tests), and communication (ultrasonic vocalizations in pups). We found that deletion of 4E-BP2 in GABAergic inhibitory neurons, defined by Gad2, resulted in impairments in social interaction and vocal communication. In contrast, deletion of 4E-BP2 in forebrain glutamatergic excitatory neurons, defined by Camk2a, or in astrocytes, defined by Gfap, failed to cause autistic-like behavioral abnormalities. Taken together, we provide evidence for an inhibitory-cell-specific role of 4E-BP2 in engendering autism-related behaviors.


Asunto(s)
Trastorno Autístico/metabolismo , Conducta Animal , Factores Eucarióticos de Iniciación/deficiencia , Neuronas GABAérgicas/metabolismo , Interneuronas/metabolismo , Biosíntesis de Proteínas , Animales , Astrocitos/metabolismo , Astrocitos/patología , Trastorno Autístico/genética , Trastorno Autístico/patología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Neuronas GABAérgicas/patología , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Interneuronas/patología , Ratones , Ratones Noqueados
4.
Annu Rev Med ; 70: 167-181, 2019 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-30365357

RESUMEN

Fragile X syndrome (FXS) is the most frequent inherited form of intellectual disability and autism spectrum disorder. Loss of the fragile X mental retardation protein, FMRP, engenders molecular, behavioral, and cognitive deficits in FXS patients. Experiments using different animal models advanced our knowledge of the pathophysiology of FXS and led to the discovery of many targets for drug treatments. In this review, we discuss the potential of metformin, an antidiabetic drug approved by the US Food and Drug Administration, to correct core symptoms of FXS and other neurological disorders in humans. We summarize its mechanisms of action in different animal and cellular models and human diseases.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Metformina/uso terapéutico , Enfermedades del Sistema Nervioso/diagnóstico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Animales , Trastorno Autístico/diagnóstico , Trastorno Autístico/tratamiento farmacológico , Modelos Animales de Enfermedad , Síndrome del Cromosoma X Frágil/diagnóstico , Síndrome del Cromosoma X Frágil/genética , Humanos , Ratones , Enfermedad de Parkinson/tratamiento farmacológico , Pronóstico , Serina-Treonina Quinasas TOR/genética , Resultado del Tratamiento
5.
J Neurosci ; 38(8): 2118-2133, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29367404

RESUMEN

The MAPK/ERK (mitogen-activated protein kinases/extracellular signal-regulated kinase) pathway is a cardinal regulator of synaptic plasticity, learning, and memory in the hippocampus. One of major endpoints of this signaling cascade is the 5' mRNA cap binding protein eIF4E (eukaryotic Initiation Factor 4E), which is phosphorylated on Ser 209 by MNK (MAPK-interacting protein kinases) and controls mRNA translation. The precise role of phospho-eIF4E in the brain is yet to be determined. Herein, we demonstrate that ablation of eIF4E phosphorylation in male mice (4Eki mice) does not impair long-term spatial or contextual fear memory, or the late phase of LTP. Using unbiased translational profiling in mouse brain, we show that phospho-eIF4E differentially regulates the translation of a subset of mRNAs linked to inflammation, the extracellular matrix, pituitary hormones, and the serotonin pathway. Consequently, 4Eki male mice display exaggerated inflammatory responses and reduced levels of serotonin, concomitant with depression and anxiety-like behaviors. Remarkably, eIF4E phosphorylation is required for the chronic antidepressant action of the selective serotonin reuptake inhibitor fluoxetine. Finally, we propose a novel phospho-eIF4E-dependent translational control mechanism in the brain, via the GAIT complex (gamma IFN activated inhibitor of translation). In summary, our work proposes a novel translational control mechanism involved in the regulation of inflammation and depression, which could be exploited to design novel therapeutics.SIGNIFICANCE STATEMENT We demonstrate that downstream of the MAPK (mitogen-activated protein kinase) pathway, eukaryotic Initiation Factor 4E (eIF4E) Ser209 phosphorylation is not required for classical forms of hippocampal LTP and memory. We reveal a novel role for eIF4E phosphorylation in inflammatory responses and depression-like behaviors. eIF4E phosphorylation is required for the chronic action of antidepressants, such as fluoxetine in mice. These phenotypes are accompanied by selective translation of extracellular matrix, pituitary hormones, and serotonin pathway genes, in eIF4E phospho-mutant mice. We also describe a previously unidentified translational control mechanism in the brain, whereby eIF4E phosphorylation is required for inhibiting the translation of gamma IFN activated inhibitor of translation element-containing mRNAs. These findings can be used to design novel therapeutics for depression.


Asunto(s)
Depresión/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Inflamación/metabolismo , Biosíntesis de Proteínas/fisiología , Animales , Depresión/fisiopatología , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación
6.
Proc Natl Acad Sci U S A ; 110(8): 3131-6, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23382228

RESUMEN

The multiple memory systems hypothesis posits that dorsal striatum and hippocampus are central nodes in independent memory systems, supporting response-based and place-based learning, respectively. Although our understanding of the function of hippocampus within this framework is relatively well established, the contribution of dorsal striatum is less clear. This in part seems to be due to the heterogeneous nature of dorsal striatum, which receives extensive topographically organized projections from higher cortical areas. Here we quantified neural activity in the intact brain while mice and humans acquired analogous versions of the Morris water maze. We found that dorsomedial striatum and medial prefrontal cortex support the initial acquisition of what is typically considered a hippocampus-dependent spatial learning task. We suggest that the circuit involving dorsomedial striatum and medial prefrontal cortex identified here plays a more task-independent role in early learning than currently thought. Furthermore, our results demonstrate that dorsomedial and dorsolateral striatum serve fundamentally different roles during place learning. The remarkably high degree of anatomical overlap in brain function between mouse and human observed in our study emphasizes the extent of convergence achievable with a well-matched multilevel approach.


Asunto(s)
Cuerpo Estriado/fisiología , Aprendizaje por Laberinto , Corteza Prefrontal/fisiología , Adulto , Animales , Femenino , Humanos , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Adulto Joven
7.
Neurobiol Dis ; 76: 137-158, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25684539

RESUMEN

D1-dopamine receptors (Drd1a) are highly expressed in the deep layers of the cerebral cortex and the striatum. A number of human diseases such as Huntington disease and schizophrenia are known to have cortical pathology involving dopamine receptor expressing neurons. To illuminate their functional role, we exploited a Cre/Lox molecular paradigm to generate Emx-1(tox) MUT mice, a transgenic line in which cortical Drd1a-expressing pyramidal neurons were selectively ablated. Emx-1(tox) MUT mice displayed prominent forelimb dystonia, hyperkinesia, ataxia on rotarod testing, heightened anxiety-like behavior, and age-dependent abnormalities in a test of social interaction. The latter occurred in the context of normal working memory on testing in the Y-maze and for novel object recognition. Some motor and behavioral abnormalities in Emx-1(tox) MUT mice overlapped with those in CamKIIα(tox) MUT transgenic mice, a line in which both striatal and cortical Drd1a-expressing cells were ablated. Although Emx-1(tox) MUT mice had normal striatal anatomy, both Emx-1(tox) MUT and CamKIIα(tox) MUT mice displayed selective neuronal loss in cortical layers V and VI. This study shows that loss of cortical Drd1a-expressing cells is sufficient to produce deficits in multiple motor and behavioral domains, independent of striatal mechanisms. Primary cortical changes in the D1 dopamine receptor compartment are therefore likely to model a number of core clinical features in disorders such as Huntington disease and schizophrenia.


Asunto(s)
Conducta Animal/fisiología , Corteza Cerebral/fisiología , Enfermedad de Huntington/fisiopatología , Células Piramidales/fisiología , Receptores de Dopamina D1/fisiología , Esquizofrenia/fisiopatología , Animales , Ansiedad/genética , Ansiedad/fisiopatología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Femenino , Marcha/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Enfermedad de Huntington/genética , Masculino , Memoria/fisiología , Ratones , Ratones Transgénicos , Actividad Motora/genética , Mutación , Fenotipo , Receptores de Dopamina D1/genética , Esquizofrenia/genética , Conducta Social , Factores de Transcripción/genética , Factores de Transcripción/fisiología
8.
Neurobiol Dis ; 62: 323-37, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24135007

RESUMEN

Progressive cell loss is observed in the striatum, cerebral cortex, thalamus, hypothalamus, subthalamic nucleus and hippocampus in Huntington disease. In the striatum, dopamine-responsive medium spiny neurons are preferentially lost. Clinical features include involuntary movements, gait and orofacial impairments in addition to cognitive deficits and psychosis, anxiety and mood disorders. We utilized the Cre-LoxP system to generate mutant mice with selective postnatal ablation of D1 dopamine receptor-expressing striatal neurons to determine which elements of the complex Huntington disease phenotype relate to loss of this neuronal subpopulation. Mutant mice had reduced body weight, locomotor slowing, reduced rearing, ataxia, a short stride length wide-based erratic gait, impairment in orofacial movements and displayed haloperidol-suppressible tic-like movements. The mutation was associated with an anxiolytic profile. Mutant mice had significant striatal-specific atrophy and astrogliosis. D1-expressing cell number was reduced throughout the rostrocaudal extent of the dorsal striatum consistent with partial destruction of the striatonigral pathway. Additional striatal changes included up-regulated D2 and enkephalin mRNA, and an increased density of D2 and preproenkephalin-expressing projection neurons, and striatal neuropeptide Y and cholinergic interneurons. These data suggest that striatal D1-cell-ablation alone may account for the involuntary movements and locomotor, balance and orofacial deficits seen not only in HD but also in HD phenocopy syndromes with striatal atrophy. Therapeutic strategies would therefore need to target striatal D1 cells to ameliorate deficits especially when the clinical presentation is dominated by a bradykinetic/ataxic phenotype with involuntary movements.


Asunto(s)
Cuerpo Estriado/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/fisiopatología , Receptores de Dopamina D1/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Recuento de Células , Cuerpo Estriado/patología , Discinesias/fisiopatología , Femenino , Marcha/fisiología , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Equilibrio Postural/fisiología , Receptores de Dopamina D1/genética
9.
Neurobiol Dis ; 51: 144-51, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23153818

RESUMEN

Neurobeachin (NBEA), a brain-enriched multidomain scaffolding protein involved in neurotransmitter release and synaptic functioning, has been identified as a candidate gene for autism spectrum disorder (ASD) in four unrelated patients haploinsufficient for NBEA. The aim of this study was to map the behavioral phenotype of Nbea(+/-) mice in order to understand its contribution to the pathogenesis of ASD. ASD-like behavioral variables of Nbea(+/-) mice were related to basal neuronal activity in different brain regions by in situ hybridizations and extracellular field recordings of synaptic plasticity in hippocampal cornu ammonis 1 (CA1) region. Levels of BDNF and phosphorylated cAMP response element-binding protein (CREB) were measured in an attempt to investigate putatively underlying changes in these neuromolecules. Nbea(+/-) mice exhibit several ASD-like features, including changes in self-grooming behavior, social behaviors, conditioned fear responses, and spatial learning and memory, which coincided with enhanced long-term potentiation (LTP) in their CA1 region. The observed alterations in learning and memory and hippocampal LTP are concomitant with decreased expression of the immediate early gene zif268 in dorsomedial striatum and hippocampal CA1 region, increased CREB phosphorylation, and increased hippocampal BDNF expression. These findings indicate that Nbea haploinsufficiency leads to various molecular and cellular changes that affect neuroplasticity and behavioral functions in mice, and could thus underlie the ASD symptomatology in NBEA deficient humans.


Asunto(s)
Trastorno Autístico/genética , Conducta Animal/fisiología , Encéfalo/fisiopatología , Proteínas Portadoras/genética , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/genética , Animales , Niño , Femenino , Haploinsuficiencia , Humanos , Immunoblotting , Hibridación in Situ , Aprendizaje/fisiología , Potenciación a Largo Plazo/fisiología , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Transmisión Sináptica/genética
10.
J Pharmacol Sci ; 121(1): 39-47, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23337398

RESUMEN

Considerable topographic overlap exists between brain opioidergic and dopaminergic neurons. Pharmacological blockade of the dopamine D(1) receptor (Drd1a) reverses several behavioural phenomena elicited by opioids. The present study examines the effects of morphine in adult mutant (MUT) mice expressing the attenuated diphtheria toxin-176 gene in Drd1a-expressing cells, a mutant line shown previously to undergo post-natal striatal atrophy and loss of Drd1a-expression. MUT and wild-type mice were assessed behaviourally following acute administration of 10 mg/kg morphine. Treatment with morphine reduced locomotion and rearing similarly in both genotypes but reduced total grooming only in MUT mice. Morphine-induced Straub tail and stillness were heightened in MUT mice. Chewing and sifting were decreased in MUT mice and these effects were not modified by morphine. Loss of striatal Drd1-positive cells and up-regulated D(2)-expression, as reflected in down-regulated D(1)-like and up-regulated D(2)-like binding, respectively, is not uniform along the cranio-caudal extent in this model but appears to be greater in the caudal striatum. Preferential caudal loss of µ-opioid-expression, a marker for the striosomal compartment, was seen. These data indicate that Drd1a-positive cell loss modifies the exploratory behavioural response elicited by morphine, unmasking novel morphine-induced MUT-specific behaviours and generating a hypersensitivity to morphine for others.


Asunto(s)
Conducta Animal/efectos de los fármacos , Ratones Mutantes/genética , Ratones Mutantes/psicología , Morfina/farmacología , Receptores de Dopamina D1/antagonistas & inhibidores , Animales , Núcleo Caudado/metabolismo , Femenino , Masculino , Ratones , Morfina/administración & dosificación , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores Opioides mu/metabolismo
11.
Curr Res Neurobiol ; 5: 100094, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37416094

RESUMEN

A growing body of evidence has implicated progranulin in neurodevelopment and indicated that aberrant progranulin expression may be involved in neurodevelopmental disease. Specifically, increased progranulin expression in the prefrontal cortex has been suggested to be pathologically relevant in male Fmr1 knockout (Fmr1 KO) mice, a mouse model of Fragile X Syndrome (FXS). Further investigation into the role of progranulin in FXS is warranted to determine if therapies that reduce progranulin expression represent a viable strategy for treating patients with FXS. Several key knowledge gaps remain. The mechanism of increased progranulin expression in Fmr1 KO mice is poorly understood and the extent of progranulin's involvement in FXS-like phenotypes in Fmr1 KO mice has been incompletely explored. To this end, we have performed a thorough characterization of progranulin expression in Fmr1 KO mice. We find that the phenomenon of increased progranulin expression is post-translational and tissue-specific. We also demonstrate for the first time an association between progranulin mRNA and FMRP, suggesting that progranulin mRNA is an FMRP target. Subsequently, we show that progranulin over-expression in Fmr1 wild-type mice causes reduced repetitive behaviour engagement in females and mild hyperactivity in males but is largely insufficient to recapitulate FXS-associated behavioural, morphological, and electrophysiological abnormalities. Lastly, we determine that genetic reduction of progranulin expression on an Fmr1 KO background reduces macroorchidism but does not alter other FXS-associated behaviours or biochemical phenotypes.

12.
Neuron ; 111(19): 3028-3040.e6, 2023 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-37473758

RESUMEN

Dysregulation of protein synthesis is one of the key mechanisms underlying autism spectrum disorder (ASD). However, the role of a major pathway controlling protein synthesis, the integrated stress response (ISR), in ASD remains poorly understood. Here, we demonstrate that the main arm of the ISR, eIF2α phosphorylation (p-eIF2α), is suppressed in excitatory, but not inhibitory, neurons in a mouse model of fragile X syndrome (FXS; Fmr1-/y). We further show that the decrease in p-eIF2α is mediated via activation of mTORC1. Genetic reduction of p-eIF2α only in excitatory neurons is sufficient to increase general protein synthesis and cause autism-like behavior. In Fmr1-/y mice, restoration of p-eIF2α solely in excitatory neurons reverses elevated protein synthesis and rescues autism-related phenotypes. Thus, we reveal a previously unknown causal relationship between excitatory neuron-specific translational control via the ISR pathway, general protein synthesis, and core phenotypes reminiscent of autism in a mouse model of FXS.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Síndrome del Cromosoma X Frágil , Animales , Ratones , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Neuronas/metabolismo , Fenotipo , Ratones Noqueados , Modelos Animales de Enfermedad
13.
Neurobiol Learn Mem ; 95(3): 260-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21130175

RESUMEN

Phosphodiesterase 10A (PDE10A) hydrolyzes both cAMP and cGMP, and is a key element in the regulation of medium spiny neuron (MSN) activity in the striatum. In the present report, we investigated the effects of targeted disruption of PDE10A on spatial learning and memory as well as aversive and appetitive conditioning in C57BL/6J mice. Because of its putative role in motivational processes and reward learning, we also determined the expression of the immediate early gene zif268 in striatum and anterior cingulate cortex. Animals showed decreased response rates in scheduled appetitive operant conditioning, as well as impaired aversive conditioning in a passive avoidance task. Morris water maze performance revealed not-motor related spatial learning and memory deficits. Anxiety and social explorative behavior was not affected in PDE10A-deficient mice. Expression of zif268 was increased in striatum and anterior cingulate cortex, which suggests alterations in the neural connections between striatum and anterior cingulate cortex in PDE10A-deficient mice. The changes in behavior and plasticity in these PDE10A-deficient mice were in accordance with the proposed role of striatal MSNs and corticostriatal connections in evaluative salience attribution.


Asunto(s)
Aprendizaje por Asociación/fisiología , Reacción de Prevención/fisiología , Condicionamiento Operante/fisiología , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Conducta Exploratoria/fisiología , Hidrolasas Diéster Fosfóricas/fisiología , Análisis de Varianza , Animales , Conducta Apetitiva/fisiología , Discriminación en Psicología/fisiología , Giro del Cíngulo/metabolismo , Giro del Cíngulo/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Neostriado/metabolismo , Neostriado/fisiología , Hidrolasas Diéster Fosfóricas/genética , Conducta Social , Estadísticas no Paramétricas
14.
Cell Rep ; 35(4): 109036, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33910008

RESUMEN

Recent studies have demonstrated that selective activation of mammalian target of rapamycin complex 1 (mTORC1) in the cerebellum by deletion of the mTORC1 upstream repressors TSC1 or phosphatase and tensin homolog (PTEN) in Purkinje cells (PCs) causes autism-like features and cognitive deficits. However, the molecular mechanisms by which overactivated mTORC1 in the cerebellum engenders these behaviors remain unknown. The eukaryotic translation initiation factor 4E-binding protein 2 (4E-BP2) is a central translational repressor downstream of mTORC1. Here, we show that mice with selective ablation of 4E-BP2 in PCs display a reduced number of PCs, increased regularity of PC action potential firing, and deficits in motor learning. Surprisingly, although spatial memory is impaired in these mice, they exhibit normal social interaction and show no deficits in repetitive behavior. Our data suggest that, downstream of mTORC1/4E-BP2, there are distinct cerebellar mechanisms independently controlling social behavior and memory formation.


Asunto(s)
Trastorno Autístico/genética , Proteínas Portadoras/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Biosíntesis de Proteínas/genética , Células de Purkinje/metabolismo , Memoria Espacial/fisiología , Animales , Humanos , Ratones
15.
Stem Cell Reports ; 16(7): 1749-1762, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34214487

RESUMEN

Mutations in HPRT1, a gene encoding a rate-limiting enzyme for purine salvage, cause Lesch-Nyhan disease which is characterized by self-injury and motor impairments. We leveraged stem cell and genetic engineering technologies to model the disease in isogenic and patient-derived forebrain and midbrain cell types. Dopaminergic progenitor cells deficient in HPRT showed decreased intensity of all developmental cell-fate markers measured. Metabolic analyses revealed significant loss of all purine derivatives, except hypoxanthine, and impaired glycolysis and oxidative phosphorylation. real-time glucose tracing demonstrated increased shunting to the pentose phosphate pathway for de novo purine synthesis at the expense of ATP production. Purine depletion in dopaminergic progenitor cells resulted in loss of RHEB, impairing mTORC1 activation. These data demonstrate dopaminergic-specific effects of purine salvage deficiency and unexpectedly reveal that dopaminergic progenitor cells are programmed to a high-energy state prior to higher energy demands of terminally differentiated cells.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Metabolismo Energético , Síndrome de Lesch-Nyhan/metabolismo , Síndrome de Lesch-Nyhan/patología , Mesencéfalo/patología , Biomarcadores/metabolismo , Linaje de la Célula , Corteza Cerebral/patología , Glucosa/metabolismo , Glucólisis , Humanos , Hipoxantina Fosforribosiltransferasa/deficiencia , Síndrome de Lesch-Nyhan/enzimología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Células-Madre Neurales/metabolismo , Fosforilación Oxidativa , Vía de Pentosa Fosfato , Purinas/metabolismo
16.
J Muscle Res Cell Motil ; 30(1-2): 73-83, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19404753

RESUMEN

We generated a mouse line with a missense mutation (S248F) in the gene (CHRNA4) encoding the alpha4 subunit of neuronal nicotinic acetylcholine receptor (nAChR). Mutant mice demonstrate brief nicotine induced dystonia that resembles the clinical events seen in patients with the same mutation. Drug-induced dystonia is more pronounced in female mice, thus our aim was to determine if the S248F mutation changed the properties of fast- and slow-twitch muscle fibres from female mutant mice. Reverse transcriptase-PCR confirmed CHRNA4 gene expression in the brain but not skeletal muscles in normal and mutant mice. Ca(2+) and Sr(2+) force activation curves were obtained using skinned muscle fibres prepared from slow-twitch (soleus) and fast-twitch (EDL) muscles. Two significant results were found: (1) the (pCa(50) - pSr(50)) value from EDL fibres was smaller in mutant mice than in wild type (1.01 vs. 1.30), (2) the percentage force produced at pSr 5.5 was larger in mutants than in wild type (5.76 vs. 0.24%). Both results indicate a shift to slow-twitch characteristics in the mutant. This conclusion is supported by the identification of the myosin heavy chain (MHC) isoforms. Mutant EDL fibres expressed MHC I (usually only found in slow-twitch fibres) as well as MHC IIa. Despite the lack of spontaneous dystonic events, our findings suggest that mutant mice may be having subclinical events or the mutation results in a chronic alteration to muscle neural input.


Asunto(s)
Distonía/fisiopatología , Fibras Musculares de Contracción Rápida/fisiología , Fibras Musculares de Contracción Lenta/fisiología , Receptores Nicotínicos/genética , Animales , Encéfalo/metabolismo , Encéfalo/fisiología , Calcio/farmacología , Modelos Animales de Enfermedad , Distonía/inducido químicamente , Distonía/genética , Femenino , Técnicas de Sustitución del Gen , Miembro Posterior/fisiopatología , Ratones , Ratones Endogámicos C57BL , Fibras Musculares de Contracción Rápida/efectos de los fármacos , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Lenta/efectos de los fármacos , Fibras Musculares de Contracción Lenta/metabolismo , Mutación Missense , Cadenas Pesadas de Miosina/metabolismo , Nicotina/toxicidad , Isoformas de Proteínas/metabolismo , Estroncio/farmacología
17.
J Neurosci ; 27(38): 10128-42, 2007 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-17881519

RESUMEN

We generated a mouse line harboring an autosomal-dominant nocturnal frontal lobe epilepsy (ADNFLE) mutation: the alpha4 nicotinic receptor S248F knock-in strain. In this mouse, modest nicotine doses (1-2 mg/kg) elicit a novel behavior termed the dystonic arousal complex (DAC). The DAC includes stereotypical head movements, body jerking, and forelimb dystonia; these behaviors resemble some core features of ADNFLE. A marked Straub tail is an additional component of the DAC. Similar to attacks in ADNFLE, the DAC can be partially suppressed by the sodium channel blocker carbamazepine or by pre-exposure to a very low dose of nicotine (0.1 mg/kg). The DAC is centrally mediated, genetically highly penetrant, and, surprisingly, not associated with overt ictal electrical activity as assessed by (1) epidural or frontal lobe depth-electrode electroencephalography or (2) hippocampal c-fos-regulated gene expression. Heterozygous knock-in mice are partially protected from nicotine-induced seizures. The noncompetitive antagonist mecamylamine does not suppress the DAC, although it suppresses high-dose nicotine-induced wild-type-like seizures. Experiments on agonist-induced 86Rb+ and neurotransmitter efflux from synaptosomes and on alpha4S248Fbeta2 receptors expressed in oocytes confirm that the S248F mutation confers resistance to mecamylamine blockade. Genetic background, gender, and mutant gene expression levels modulate expression of the DAC phenotype in mice. The S248F mouse thus appears to provide a model for the paroxysmal dystonic element of ADNFLE semiology. Our model complements what is seen in other ADNFLE animal models. Together, these mice cover the spectrum of behavioral and electrographic events seen in the human condition.


Asunto(s)
Nivel de Alerta/genética , Modelos Animales de Enfermedad , Trastornos Distónicos/genética , Epilepsia del Lóbulo Frontal/genética , Mutación , Nicotina/toxicidad , Animales , Nivel de Alerta/efectos de los fármacos , Trastornos Distónicos/inducido químicamente , Epilepsia del Lóbulo Frontal/inducido químicamente , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Ratones Transgénicos , Ratas , Especificidad de la Especie , Xenopus
18.
PLoS One ; 12(5): e0176295, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28467439

RESUMEN

Neurons in anterior cingulate cortex (aCC) project to dorsomedial striatum (DMS) as part of a corticostriatal circuit with putative roles in learning and other cognitive functions. In the present study, the spatial-cognitive importance of aCC and DMS was assessed in the hidden-platform version of the Morris water maze (MWM). Brain lesion experiments that focused on areas of connectivity between these regions indicated their involvement in spatial cognition. MWM learning curves were markedly delayed in DMS-lesioned mice in the absence of other major functional impairments, whereas there was a more subtle, but still significant influence of aCC lesions. Lesioned mice displayed impaired abilities to use spatial search strategies, increased thigmotaxic swimming, and decreased searching in the proximity of the escape platform. Additionally, aCC and DMS activity was compared in mice between the early acquisition phase (2 and 3 days of training) and the over-trained high-proficiency phase (after 30 days of training). Neuroplasticity-related expression of the immediate early gene Arc implicated both regions during the goal-directed, early phases of spatial learning. These results suggest the functional involvement of aCC and DMS in processes of spatial cognition that model associative cortex-dependent, human episodic memory abilities.


Asunto(s)
Cognición , Cuerpo Estriado/fisiología , Giro del Cíngulo/fisiología , Conducta Espacial , Animales , Femenino , Ratones , Ratones Endogámicos C57BL
19.
Nat Rev Dis Primers ; 3: 17065, 2017 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-28960184

RESUMEN

Fragile X syndrome (FXS) is the leading inherited form of intellectual disability and autism spectrum disorder, and patients can present with severe behavioural alterations, including hyperactivity, impulsivity and anxiety, in addition to poor language development and seizures. FXS is a trinucleotide repeat disorder, in which >200 repeats of the CGG motif in FMR1 leads to silencing of the gene and the consequent loss of its product, fragile X mental retardation 1 protein (FMRP). FMRP has a central role in gene expression and regulates the translation of potentially hundreds of mRNAs, many of which are involved in the development and maintenance of neuronal synaptic connections. Indeed, disturbances in neuroplasticity is a key finding in FXS animal models, and an imbalance in inhibitory and excitatory neuronal circuits is believed to underlie many of the clinical manifestations of this disorder. Our knowledge of the proteins that are regulated by FMRP is rapidly growing, and this has led to the identification of multiple targets for therapeutic intervention, some of which have already moved into clinical trials or clinical practice.


Asunto(s)
Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil/diagnóstico , Síndrome del Cromosoma X Frágil/fisiopatología , Síndrome del Cromosoma X Frágil/terapia , Humanos
20.
Nat Med ; 23(6): 674-677, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28504725

RESUMEN

Fragile X syndrome (FXS) is the leading monogenic cause of autism spectrum disorders (ASD). Trinucleotide repeat expansions in FMR1 abolish FMRP expression, leading to hyperactivation of ERK and mTOR signaling upstream of mRNA translation. Here we show that metformin, the most widely used drug for type 2 diabetes, rescues core phenotypes in Fmr1-/y mice and selectively normalizes ERK signaling, eIF4E phosphorylation and the expression of MMP-9. Thus, metformin is a potential FXS therapeutic.


Asunto(s)
Conducta Animal/efectos de los fármacos , Factor 4E Eucariótico de Iniciación/efectos de los fármacos , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Hipoglucemiantes/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metformina/farmacología , Conducta Social , Animales , Modelos Animales de Enfermedad , Factor 4E Eucariótico de Iniciación/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/fisiopatología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Expansión de Repetición de Trinucleótido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA