Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 176(5): 1143-1157.e13, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30794775

RESUMEN

We tested a newly described molecular memory system, CCR5 signaling, for its role in recovery after stroke and traumatic brain injury (TBI). CCR5 is uniquely expressed in cortical neurons after stroke. Post-stroke neuronal knockdown of CCR5 in pre-motor cortex leads to early recovery of motor control. Recovery is associated with preservation of dendritic spines, new patterns of cortical projections to contralateral pre-motor cortex, and upregulation of CREB and DLK signaling. Administration of a clinically utilized FDA-approved CCR5 antagonist, devised for HIV treatment, produces similar effects on motor recovery post stroke and cognitive decline post TBI. Finally, in a large clinical cohort of stroke patients, carriers for a naturally occurring loss-of-function mutation in CCR5 (CCR5-Δ32) exhibited greater recovery of neurological impairments and cognitive function. In summary, CCR5 is a translational target for neural repair in stroke and TBI and the first reported gene associated with enhanced recovery in human stroke.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Receptores CCR5/metabolismo , Accidente Cerebrovascular/terapia , Anciano , Anciano de 80 o más Años , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Espinas Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Corteza Motora/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Receptores CCR5/fisiología , Rehabilitación de Accidente Cerebrovascular/métodos
2.
Cell ; 173(1): 166-180.e14, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29502969

RESUMEN

Brain-wide fluctuations in local field potential oscillations reflect emergent network-level signals that mediate behavior. Cracking the code whereby these oscillations coordinate in time and space (spatiotemporal dynamics) to represent complex behaviors would provide fundamental insights into how the brain signals emotional pathology. Using machine learning, we discover a spatiotemporal dynamic network that predicts the emergence of major depressive disorder (MDD)-related behavioral dysfunction in mice subjected to chronic social defeat stress. Activity patterns in this network originate in prefrontal cortex and ventral striatum, relay through amygdala and ventral tegmental area, and converge in ventral hippocampus. This network is increased by acute threat, and it is also enhanced in three independent models of MDD vulnerability. Finally, we demonstrate that this vulnerability network is biologically distinct from the networks that encode dysfunction after stress. Thus, these findings reveal a convergent mechanism through which MDD vulnerability is mediated in the brain.


Asunto(s)
Encéfalo/fisiología , Depresión/patología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Depresión/fisiopatología , Modelos Animales de Enfermedad , Estimulación Eléctrica , Electrodos Implantados , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Ketamina/farmacología , Aprendizaje Automático , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenómenos Fisiológicos/efectos de los fármacos , Corteza Prefrontal/fisiología , Estrés Psicológico
3.
Nature ; 606(7912): 146-152, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35614219

RESUMEN

Real-world memories are formed in a particular context and are often not acquired or recalled in isolation1-5. Time is a key variable in the organization of memories, as events that are experienced close in time are more likely to be meaningfully associated, whereas those that are experienced with a longer interval are not1-4. How the brain segregates events that are temporally distinct is unclear. Here we show that a delayed (12-24 h) increase in the expression of C-C chemokine receptor type 5 (CCR5)-an immune receptor that is well known as a co-receptor for HIV infection6,7-after the formation of a contextual memory determines the duration of the temporal window for associating or linking that memory with subsequent memories. This delayed expression of CCR5 in mouse dorsal CA1 neurons results in a decrease in neuronal excitability, which in turn negatively regulates neuronal memory allocation, thus reducing the overlap between dorsal CA1 memory ensembles. Lowering this overlap affects the ability of one memory to trigger the recall of the other, and therefore closes the temporal window for memory linking. Our findings also show that an age-related increase in the neuronal expression of CCR5 and its ligand CCL5 leads to impairments in memory linking in aged mice, which could be reversed with a Ccr5 knockout and a drug approved by the US Food and Drug Administration (FDA) that inhibits this receptor, a result with clinical implications. Altogether, the findings reported here provide insights into the molecular and cellular mechanisms that shape the temporal window for memory linking.


Asunto(s)
Región CA1 Hipocampal , Memoria , Neuronas , Receptores CCR5 , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/fisiología , Memoria/fisiología , Recuerdo Mental/fisiología , Ratones , Neuronas/metabolismo , Receptores CCR5/deficiencia , Receptores CCR5/genética , Receptores CCR5/metabolismo , Factores de Tiempo
4.
Cell ; 135(3): 549-60, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18984165

RESUMEN

We uncovered a role for ERK signaling in GABA release, long-term potentiation (LTP), and learning, and show that disruption of this mechanism accounts for the learning deficits in a mouse model for learning disabilities in neurofibromatosis type I (NF1). Our results demonstrate that neurofibromin modulates ERK/synapsin I-dependent GABA release, which in turn modulates hippocampal LTP and learning. An Nf1 heterozygous null mutation, which results in enhanced ERK and synapsin I phosphorylation, increased GABA release in the hippocampus, and this was reversed by pharmacological downregulation of ERK signaling. Importantly, the learning deficits associated with the Nf1 mutation were rescued by a subthreshold dose of a GABA(A) antagonist. Accordingly, Cre deletions of Nf1 showed that only those deletions involving inhibitory neurons caused hippocampal inhibition, LTP, and learning abnormalities. Importantly, our results also revealed lasting increases in GABA release triggered by learning, indicating that the mechanisms uncovered here are of general importance for learning.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Genes de Neurofibromatosis 1 , Aprendizaje , Potenciación a Largo Plazo , Neurofibromina 1/metabolismo , Transducción de Señal , Ácido gamma-Aminobutírico/metabolismo , Animales , Femenino , Hipocampo/metabolismo , Discapacidades para el Aprendizaje/fisiopatología , Masculino , Ratones , Neurofibromatosis 1/fisiopatología , Neurofibromina 1/genética , Fosforilación , Sinapsinas/metabolismo
5.
Nature ; 534(7605): 115-8, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27251287

RESUMEN

Recent studies suggest that a shared neural ensemble may link distinct memories encoded close in time. According to the memory allocation hypothesis, learning triggers a temporary increase in neuronal excitability that biases the representation of a subsequent memory to the neuronal ensemble encoding the first memory, such that recall of one memory increases the likelihood of recalling the other memory. Here we show in mice that the overlap between the hippocampal CA1 ensembles activated by two distinct contexts acquired within a day is higher than when they are separated by a week. Several findings indicate that this overlap of neuronal ensembles links two contextual memories. First, fear paired with one context is transferred to a neutral context when the two contexts are acquired within a day but not across a week. Second, the first memory strengthens the second memory within a day but not across a week. Older mice, known to have lower CA1 excitability, do not show the overlap between ensembles, the transfer of fear between contexts, or the strengthening of the second memory. Finally, in aged mice, increasing cellular excitability and activating a common ensemble of CA1 neurons during two distinct context exposures rescued the deficit in linking memories. Taken together, these findings demonstrate that contextual memories encoded close in time are linked by directing storage into overlapping ensembles. Alteration of these processes by ageing could affect the temporal structure of memories, thus impairing efficient recall of related information.


Asunto(s)
Región CA1 Hipocampal/citología , Región CA1 Hipocampal/fisiología , Memoria/fisiología , Neuronas/fisiología , Animales , Calcio/análisis , Miedo , Masculino , Recuerdo Mental/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Neurológicos , Factores de Tiempo
6.
Nat Rev Neurosci ; 15(3): 157-69, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24496410

RESUMEN

There is now compelling evidence that the allocation of memory to specific neurons (neuronal allocation) and synapses (synaptic allocation) in a neurocircuit is not random and that instead specific mechanisms, such as increases in neuronal excitability and synaptic tagging and capture, determine the exact sites where memories are stored. We propose an integrated view of these processes, such that neuronal allocation, synaptic tagging and capture, spine clustering and metaplasticity reflect related aspects of memory allocation mechanisms. Importantly, the properties of these mechanisms suggest a set of rules that profoundly affect how memories are stored and recalled.


Asunto(s)
Hipocampo/fisiología , Memoria/fisiología , Modelos Neurológicos , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Animales , Espinas Dendríticas/fisiología , Humanos , Red Nerviosa/fisiología
8.
Proc Natl Acad Sci U S A ; 111(23): 8661-6, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24912150

RESUMEN

The retrosplenial cortex (RSC) is part of a network of interconnected cortical, hippocampal, and thalamic structures harboring spatially modulated neurons. The RSC contains head direction cells and connects to the parahippocampal region and anterior thalamus. Manipulations of the RSC can affect spatial and contextual tasks. A considerable amount of evidence implicates the role of the RSC in spatial navigation, but it is unclear whether this structure actually encodes or stores spatial information. We used a transgenic mouse in which the expression of green fluorescent protein was under the control of the immediate early gene c-fos promoter as well as time-lapse two-photon in vivo imaging to monitor neuronal activation triggered by spatial learning in the Morris water maze. We uncovered a repetitive pattern of cell activation in the RSC consistent with the hypothesis that during spatial learning an experience-dependent memory trace is formed in this structure. In support of this hypothesis, we also report three other observations. First, temporary RSC inactivation disrupts performance in a spatial learning task. Second, we show that overexpressing the transcription factor CREB in the RSC with a viral vector, a manipulation known to enhance memory consolidation in other circuits, results in spatial memory enhancements. Third, silencing the viral CREB-expressing neurons with the allatostatin system occludes the spatial memory enhancement. Taken together, these results indicate that the retrosplenial cortex engages in the formation and storage of memory traces for spatial information.


Asunto(s)
Giro del Cíngulo/fisiología , Hipocampo/fisiología , Memoria/fisiología , Percepción Espacial/fisiología , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Giro del Cíngulo/citología , Giro del Cíngulo/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Aprendizaje por Laberinto/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal/métodos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Neuronas/citología , Neuronas/metabolismo , Neuronas/fisiología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-fos/genética
9.
EMBO J ; 31(5): 1203-16, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22234183

RESUMEN

Memory is essential for our normal daily lives and our sense of self. Ca(2+) influx through the NMDA-type glutamate receptor (NMDAR) and the ensuing activation of the Ca(2+) and calmodulin-dependent protein kinase (CaMKII) are required for memory formation and its physiological correlate, long-term potentiation (LTP). The Ca(2+) influx induces CaMKII binding to the NMDAR to strategically recruit CaMKII to synapses that are undergoing potentiation. We generated mice with two point mutations that impair CaMKII binding to the NMDAR GluN2B subunit. Ca(2+)-triggered postsynaptic accumulation is largely abrogated for CaMKII and destabilized for TARPs, which anchor AMPA-type glutamate receptors (AMPAR). LTP is reduced by 50% and phosphorylation of the AMPAR GluA1 subunit by CaMKII, which enhances AMPAR conductance, impaired. The mutant mice learn the Morris water maze (MWM) as well as WT but show deficiency in recall during the period of early memory consolidation. Accordingly, the activity-driven interaction of CaMKII with the NMDAR is important for recall of MWM memory as early as 24 h, but not 1-2 h, after training potentially due to impaired consolidation.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Memoria , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Mutación Puntual , Unión Proteica , Receptores de N-Metil-D-Aspartato/genética
10.
Learn Mem ; 22(9): 494-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26286658

RESUMEN

The sheer volume and complexity of publications in the biological sciences are straining traditional approaches to research planning. Nowhere is this problem more serious than in molecular and cellular cognition, since in this neuroscience field, researchers routinely use approaches and information from a variety of areas in neuroscience and other biology fields. Additionally, the multilevel integration process characteristic of this field involves the establishment of experimental connections between molecular, electrophysiological, behavioral, and even cognitive data. This multidisciplinary integration process requires strategies and approaches that originate in several different fields, which greatly increases the complexity and demands of this process. Although causal assertions, where phenomenon A is thought to contribute or relate to B, are at the center of this integration process and key to research in biology, there are currently no tools to help scientists keep track of the increasingly more complex network of causal connections they use when making research decisions. Here, we propose the development of semiautomated graphical and interactive tools to help neuroscientists and other biologists, including those working in molecular and cellular cognition, to track, map, and weight causal evidence in research papers. There is a great need for a concerted effort by biologists, computer scientists, and funding institutions to develop maps of causal information that would aid in integration of research findings and in experiment planning.


Asunto(s)
Investigación Biomédica , Cognición/fisiología , Biología Computacional/métodos , Neurociencias/métodos , Proyectos de Investigación , Animales , Curaduría de Datos , Reconocimiento de Normas Patrones Automatizadas/métodos , Edición
11.
J Neurosci ; 34(34): 11180-7, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25143599

RESUMEN

The α isoform of the calcium/calmodulin-dependent protein kinase II (αCaMKII) has been implicated extensively in molecular and cellular mechanisms underlying spatial and contextual learning in a wide variety of species. Germline deletion of Camk2a leads to severe deficits in spatial and contextual learning in mice. However, the temporal and region-specific requirements for αCaMKII have remained largely unexplored. Here, we generated conditional Camk2a mutants to examine the influence of spatially restricted and temporally controlled expression of αCaMKII. Forebrain-specific deletion of the Camk2a gene resulted in severe deficits in water maze and contextual fear learning, whereas mice with deletion restricted to the cerebellum learned normally. Furthermore, we found that temporally controlled deletion of the Camk2a gene in adult mice is as detrimental as germline deletion for learning and synaptic plasticity. Together, we confirm the requirement for αCaMKII in the forebrain, but not the cerebellum, in spatial and contextual learning. Moreover, we highlight the absolute requirement for intact αCaMKII expression at the time of learning.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Condicionamiento Clásico/fisiología , Miedo/fisiología , Aprendizaje por Laberinto/fisiología , Conducta Espacial/fisiología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Condicionamiento Clásico/efectos de los fármacos , Antagonistas de Estrógenos/farmacología , Potenciales Postsinápticos Excitadores/genética , Miedo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hipocampo/citología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Integrasas/genética , Integrasas/metabolismo , Potenciación a Largo Plazo/genética , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Conducta Espacial/efectos de los fármacos , Tamoxifeno/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Hum Brain Mapp ; 36(11): 4566-81, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26304096

RESUMEN

Neurofibromatosis type I (NF1) is a genetic disorder caused by mutations in the neurofibromin 1 gene at locus 17q11.2. Individuals with NF1 have an increased incidence of learning disabilities, attention deficits, and autism spectrum disorders. As a single-gene disorder, NF1 represents a valuable model for understanding gene-brain-behavior relationships. While mouse models have elucidated molecular and cellular mechanisms underlying learning deficits associated with this mutation, little is known about functional brain architecture in human subjects with NF1. To address this question, we used resting state functional connectivity magnetic resonance imaging (rs-fcMRI) to elucidate the intrinsic network structure of 30 NF1 participants compared with 30 healthy demographically matched controls during an eyes-open rs-fcMRI scan. Novel statistical methods were employed to quantify differences in local connectivity (edge strength) and modularity structure, in combination with traditional global graph theory applications. Our findings suggest that individuals with NF1 have reduced anterior-posterior connectivity, weaker bilateral edges, and altered modularity clustering relative to healthy controls. Further, edge strength and modular clustering indices were correlated with IQ and internalizing symptoms. These findings suggest that Ras signaling disruption may lead to abnormal functional brain connectivity; further investigation into the functional consequences of these alterations in both humans and in animal models is warranted.


Asunto(s)
Encéfalo/fisiopatología , Neuroimagen Funcional/métodos , Red Nerviosa/fisiopatología , Neurofibromatosis 1/fisiopatología , Adolescente , Adulto , Niño , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Adulto Joven
13.
Am J Med Genet A ; 167A(8): 1741-6, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25900621

RESUMEN

"The Third International Meeting on Genetic Disorders in the RAS/MAPK Pathway: Towards a Therapeutic Approach" was held at the Renaissance Orlando at SeaWorld Hotel (August 2-4, 2013). Seventy-one physicians and scientists attended the meeting, and parallel meetings were held by patient advocacy groups (CFC International, Costello Syndrome Family Network, NF Network and Noonan Syndrome Foundation). Parent and patient advocates opened the meeting with a panel discussion to set the stage regarding their hopes and expectations for therapeutic advances. In keeping with the theme on therapeutic development, the sessions followed a progression from description of the phenotype and definition of therapeutic endpoints, to definition of genomic changes, to identification of therapeutic targets in the RAS/MAPK pathway, to preclinical drug development and testing, to clinical trials. These proceedings will review the major points of discussion.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Sistema de Señalización de MAP Quinasas , Proteínas ras/metabolismo , Enfermedades Genéticas Congénitas/metabolismo , Humanos
14.
Nat Rev Neurosci ; 10(2): 126-40, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19153576

RESUMEN

Most molecular and cellular studies of cognitive function have focused on either normal or pathological states, but recent research with transgenic mice has started to address the mechanisms of enhanced cognition. These results point to key synaptic and nuclear signalling events that can be manipulated to facilitate the induction or increase the stability of synaptic plasticity, and therefore enhance the acquisition or retention of information. Here, we review these surprising findings and explore their implications to both mechanisms of learning and memory and to ongoing efforts to develop treatments for cognitive disorders. These findings represent the beginning of a fundamental new approach in the study of enhanced cognition.


Asunto(s)
Cognición/fisiología , Biología Molecular/métodos , Plasticidad Neuronal/genética , Neuronas/fisiología , Animales , Calcio/metabolismo , Humanos , Ratones , Ratones Transgénicos , Plasticidad Neuronal/fisiología , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/fisiología
15.
Biomedicines ; 12(1)2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38255309

RESUMEN

There is evidence that viral infections during pre-natal development constitute a risk factor for neuropsychiatric disorders and lead to learning and memory deficits. However, little is known about why viral infections during early post-natal development have a different impact on learning and memory depending on the sex of the subject. We previously showed that early post-natal immune activation induces hippocampal-dependent social memory deficits in a male, but not in a female, mouse model of tuberous sclerosis complex (TSC; Tsc2+/- mice). Here, we explored the impact of a viral-like immune challenge in object memory. We demonstrate that early post-natal immune activation (during the first 2 weeks of life) leads to object memory deficits in female, but not male, mice that are heterozygous for a gene responsible for tuberous sclerosis complex (Tsc2+/- mice), while no effect was observed in wild type (WT) mice. Moreover, we found that the same immune activation in Tsc2+/- adult mice was not able to cause object memory deficits in females, which suggests that the early post-natal development stage constitutes a critical window for the effects of immune challenge on adult memory. Also, our results suggest that mTOR plays a critical role in the observed deficit in object memory in female Tsc2+/- mice. These results, together with previous results published by our laboratory, showing sex-specific memory deficits due to early post-natal immune activation, reinforce the necessity of using both males and females for research studies. This is especially true for studies related to immune activation, since the higher levels of estrogens in females are known to affect inflammation and to provide neuroprotection.

16.
bioRxiv ; 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38585718

RESUMEN

Vascular dementia (VaD) is a white matter ischemic disease and the second-leading cause of dementia, with no direct therapy. Within the lesion site, cell-cell interactions dictate the trajectory towards disease progression or repair. To elucidate the underlying intercellular signaling pathways, a VaD mouse model was developed for transcriptomic and functional studies. The mouse VaD transcriptome was integrated with a human VaD snRNA-Seq dataset. A custom-made database encompassing 4053 human and 2032 mouse ligand-receptor (L-R) interactions identified significantly altered pathways shared between human and mouse VaD. Two intercellular L-R systems, Serpine2-Lrp1 and CD39-A3AR, were selected for mechanistic study as both the ligand and receptor were dysregulated in VaD. Decreased Seprine2 expression enhances OPC differentiation in VaD repair. A clinically relevant drug that reverses the loss of CD39-A3AR function promotes tissue and behavioral recovery in the VaD model. This study presents novel intercellular signaling targets and may open new avenues for VaD therapies.

17.
Nat Commun ; 15(1): 2111, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38454000

RESUMEN

Investigative exploration and foraging leading to food consumption have vital importance, but are not well-understood. Since GABAergic inputs to the lateral and ventrolateral periaqueductal gray (l/vlPAG) control such behaviors, we dissected the role of vgat-expressing GABAergic l/vlPAG cells in exploration, foraging and hunting. Here, we show that in mice vgat l/vlPAG cells encode approach to food and consumption of both live prey and non-prey foods. The activity of these cells is necessary and sufficient for inducing food-seeking leading to subsequent consumption. Activation of vgat l/vlPAG cells produces exploratory foraging and compulsive eating without altering defensive behaviors. Moreover, l/vlPAG vgat cells are bidirectionally interconnected to several feeding, exploration and investigation nodes, including the zona incerta. Remarkably, the vgat l/vlPAG projection to the zona incerta bidirectionally controls approach towards food leading to consumption. These data indicate the PAG is not only a final downstream target of top-down exploration and foraging-related inputs, but that it also influences these behaviors through a bottom-up pathway.


Asunto(s)
Sustancia Gris Periacueductal , Ratones , Animales , Sustancia Gris Periacueductal/fisiología
18.
J Neurosci ; 32(41): 14087-93, 2012 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23055477

RESUMEN

Neurofibromatosis type 1 (NF1) is the most common monogenic disorder in which individuals manifest CNS abnormalities. Affected individuals develop glial neoplasms (optic gliomas, malignant astrocytomas) and neuronal dysfunction (learning disabilities, attention deficits). Nf1 genetically engineered mouse models have revealed the molecular and cellular underpinnings of gliomagenesis, attention deficit, and learning problems with relevance to basic neurobiology. Using NF1 as a model system, these studies have revealed critical roles for the NF1 gene in non-neoplastic cells in the tumor microenvironment, the importance of brain region heterogeneity, novel mechanisms of glial growth regulation, the neurochemical bases for attention deficit and learning abnormalities, and new insights into neural stem cell function. Here we review recent studies, presented at a symposium at the 2012 Society for Neuroscience annual meeting, that highlight unexpected cell biology insights into RAS and cAMP pathway effects on neural progenitor signaling, neuronal function, and oligodendrocyte lineage differentiation.


Asunto(s)
Enfermedades del Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Neurofibromatosis 1/metabolismo , Neurofibromina 1/fisiología , Animales , Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/fisiopatología , Humanos , Neurofibromatosis 1/genética , Neurofibromatosis 1/fisiopatología , Neurofibromina 1/genética , Transducción de Señal/fisiología
19.
Proc Natl Acad Sci U S A ; 107(44): 19026-31, 2010 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-20937865

RESUMEN

Experience-dependent cortical plasticity has been studied by using loss-of-function methods. Here, we take the complementary approach of using a genetic gain-of-function that enhances plasticity. We show that a constitutively active form of H-ras (H-ras(G12V)), expressed presynaptically at excitatory synapses in mice, accelerates and enhances multiple, mechanistically distinct forms of plasticity in the developing visual cortex. In vivo, H-ras(G12V) not only increased the rate of ocular dominance change in response to monocular deprivation (MD), but also accelerated recovery from deprivation by reverse occlusion. In vitro, H-ras(G12V) expression decreased baseline presynaptic release probability and enhanced presynaptically expressed long-term potentiation (LTP). H-ras(G12V) expression also accelerated the increase following MD in the frequency of miniature excitatory potentials, mirroring accelerated plasticity in vivo. These findings demonstrate accelerated neocortical plasticity, which offers an avenue toward future therapies for many neurological and neuropsychiatric disorders.


Asunto(s)
Genes ras , Potenciación a Largo Plazo , Sinapsis/metabolismo , Corteza Visual/metabolismo , Animales , Humanos , Ratones , Ratones Transgénicos , Sinapsis/genética , Corteza Visual/patología
20.
Proc Natl Acad Sci U S A ; 107(29): 13141-6, 2010 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-20624961

RESUMEN

Neurofibromatosis type I (NF1) is one of the most common single-gene causes of learning disabilities. Here, we use behavioral working memory probes and electrophysiological studies in a mouse model of NF1 (Nf1 heterozygous null mutants; Nf1(+/-)) to demonstrate that (i) Neurofibromin regulates prefrontal and striatal inhibitory networks, specifically activity-dependent GABA release and (ii) is required for working memory performance, with inhibition-dependent working memory deficits seen in Nf1(+/-) mice. We find that increased inhibition in medial prefrontal cortex (mPFC) is sufficient to alter persistent activity in a biophysical model of an mPFC microcircuit, suggesting a possible mechanism for Nf1(+/-) working memory deficits. Accordingly, working memory assays applied during functional MRI (fMRI) studies in human subjects with NF1 reveal hypoactivation of corticostriatal networks, which is associated with impaired working memory performance. Collectively, these integrative mouse and human studies reveal molecular and cellular mechanisms contributing to working memory deficits in NF1.


Asunto(s)
Memoria a Corto Plazo/fisiología , Neostriado/metabolismo , Inhibición Neural/fisiología , Neurofibromina 1/metabolismo , Animales , Conducta Animal/fisiología , Simulación por Computador , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Humanos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Ratones , Modelos Biológicos , Neostriado/fisiopatología , Neurofibromatosis 1/fisiopatología , Neurofibromina 1/deficiencia , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Transducción de Señal , Adulto Joven , Ácido gamma-Aminobutírico/metabolismo , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA