Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Dis ; 58: 156-68, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23742761

RESUMEN

The Coffin-Lowry syndrome (CLS) is a syndromic form of intellectual disability caused by loss-of-function of the RSK2 serine/threonine kinase encoded by the rsk2 gene. Rsk2 knockout mice, a murine model of CLS, exhibit spatial learning and memory impairments, yet the underlying neural mechanisms are unknown. In the current study, we examined the performance of Rsk2 knockout mice in cued, trace and contextual fear memory paradigms and identified selective deficits in the consolidation and reconsolidation of hippocampal-dependent fear memories as task difficulty and hippocampal demand increase. Electrophysiological, biochemical and electron microscopy analyses were carried out in the dentate gyrus of the hippocampus to explore potential alterations in neuronal functions and structure. In vivo and in vitro electrophysiology revealed impaired synaptic transmission, decreased network excitability and reduced AMPA and NMDA conductance in Rsk2 knockout mice. In the absence of RSK2, standard measures of short-term and long-term potentiation (LTP) were normal, however LTP-induced CREB phosphorylation and expression of the transcription factors EGR1/ZIF268 were reduced and that of the scaffolding protein SHANK3 was blocked, indicating impaired activity-dependent gene regulation. At the structural level, the density of perforated and non-perforated synapses and of multiple spine boutons was not altered, however, a clear enlargement of spine neck width and post-synaptic densities indicates altered synapse ultrastructure. These findings show that RSK2 loss-of-function is associated in the dentate gyrus with multi-level alterations that encompass modifications of glutamate receptor channel properties, synaptic transmission, plasticity-associated gene expression and spine morphology, providing novel insights into the mechanisms contributing to cognitive impairments in CLS.


Asunto(s)
Síndrome de Coffin-Lowry/complicaciones , Síndrome de Coffin-Lowry/genética , Giro Dentado/patología , Miedo , Trastornos de la Memoria/etiología , Mutación/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Transmisión Sináptica/genética , Animales , Condicionamiento Psicológico/fisiología , Señales (Psicología) , Giro Dentado/ultraestructura , Modelos Animales de Enfermedad , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores/genética , Reacción Cataléptica de Congelación/fisiología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , N-Metilaspartato/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Transmisión Sináptica/fisiología , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo
2.
Hippocampus ; 22(3): 631-42, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21425206

RESUMEN

Activity-dependent regulation of Egr1/Zif268, a transcription factor (TF) of the Egr family, is essential for stabilization of dentate gyrus synaptic plasticity and consolidation and reconsolidation of several forms of memory. The gene can be rapidly induced in selective brain circuits after certain types of learning or after recall. Here, we focused on area CA1 and examined regulation of Egr1, Egr2, and Egr3 mRNA and protein, and their DNA binding activity to the Egr response element (ERE) at different times after LTP in vivo and after learning and recall of a fear memory. We found LTP in CA1 leads to rapid induction of the three Egrs, however only Egr1 protein was overexpressed without a co-ordinated change in binding activity, indicating a fundamental difference between CA1 and dentate gyrus LTP. Our investigations in fear memory reveal that both learning and retrieval lead to an increase in binding of constitutively expressed Egr1 and Egr3 to the ERE, but not Egr2. Memory recall was also associated with increased Egr1 protein translation. The nature and temporal dynamics of these changes and tests for interactions between TFs suggest that in addition to ERE-mediated transcription, Egr1 in CA1 may interact with the TF c-Fos to regulate genes via other DNA response elements.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Miedo/fisiología , Memoria/fisiología , Plasticidad Neuronal/fisiología , Sinapsis/metabolismo , Animales , Reacción de Prevención/fisiología , Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Electrochoque , Masculino , Ratas , Ratas Sprague-Dawley , Sinapsis/genética
3.
Proc Natl Acad Sci U S A ; 106(28): 11771-5, 2009 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-19556537

RESUMEN

Zif268 is a transcriptional regulator that plays a crucial role in maintenance of the late phases of hippocampal long-term potentiation (LTP) and consolidation of spatial memories. Because the hippocampal place cell system is essential for long-term spatial memory, we tested the hypothesis that zif268 is required for long-term stability of hippocampal place cell representations by recording CA1 place cells in mice lacking zif268. We found that zif268 gene deletion destabilized the representation of a familiar environment after exposure to a novel environment and impaired the long-term (24 h), but not short-term (1 h), stability of newly formed representations. These impairments could be rescued by repeated exposure to the novel environment, however. These results indicate that zif268 contributes to the long-term stability of spatial representations in CA1 and support the notion that the long-term stability of place cell representations requires transcription-dependent mechanisms similar to those observed in LTP.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/deficiencia , Hipocampo/fisiología , Memoria/fisiología , Células Piramidales/fisiología , Animales , Mapeo Encefálico , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Eliminación de Gen , Potenciación a Largo Plazo/genética , Ratones , Ratones Noqueados , Factores de Tiempo
4.
Neurosci Lett ; 738: 135348, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32891673

RESUMEN

Pea3 proteins belong to a subfamily of the E-twentysix (ETS) domain superfamily of transcription factors, which play various roles during development. Polyoma Enhancer-Activator 3 (Pea3) proteins Pea3, ERM and Er81 are particularly involved in tissues with branching morphogenesis, including kidney, lung, mammary gland and nervous system development. A recent transcriptomic study on novel targets of Pea3 transcription factor revealed various axon guidance and nervous system development related targets, supporting a role of Pea3 proteins in motor neuron connectivity, as well as novel targets in signaling pathways involved in synaptic plasticity. This study focuses on the expression of Pea3 family members in hippocampal neurons, and regulation of putative Pea3 targets in Pea3-overexpressing cell lines and following induction of long-term potentiation or seizure in vivo. We show that Pea3 proteins are expressed in hippocampus in both neuronal and non-neuronal cells, and that Pea3 represses Elk-1 but activates Prkca and Nrcam expression in hippocampal cell lines. We also show that mRNA and protein levels of Pea3 family members are differentially regulated in the dentate gyrus and CA1 region upon MECS stimulation, but not upon LTP induction.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Neuronas/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Transactivadores/genética , Factores de Transcripción/genética , Transcriptoma
5.
Eur J Neurosci ; 30(10): 1923-30, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19895565

RESUMEN

The ability to form long-term memories exists very early during ontogeny; however, the properties of early memory processes, brain structures involved and underlying cellular mechanisms are poorly defined. Here, we examine the role of extracellular signal-regulated kinase (ERK), a member of the mitogen-activated protein kinase/ERK signaling cascade, which is crucial for adult memory, in the consolidation and reconsolidation of an early memory using a conditioned taste aversion paradigm in 3-day-old rat pups. We show that intraperitoneal injection of SL327, the upstream mitogen-activated protein kinase kinase inhibitor, impairs both consolidation and reconsolidation of early memory, leaving short-term memory after acquisition and after reactivation intact. The amnesic effect of SL327 diminishes with increasing delays after acquisition and reactivation. Biochemical analyses revealed ERK hyperphosphorylation in the amygdala but not the hippocampus following acquisition, suggesting functional activation of the amygdala as early as post-natal day 3, although there was no clear evidence for amygdalar ERK activation after reactivation. These results indicate that, despite an immature brain, the basic properties of memory and at least some of the molecular mechanisms and brain structures implicated in aversion memory share a number of similarities with the adult and emerge very early during ontogeny.


Asunto(s)
Envejecimiento , Reacción de Prevención/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Memoria/fisiología , Gusto/fisiología , Aminoacetonitrilo/análogos & derivados , Aminoacetonitrilo/farmacología , Animales , Animales Recién Nacidos , Reacción de Prevención/efectos de los fármacos , Encéfalo/anatomía & histología , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Femenino , Masculino , Memoria/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Gusto/efectos de los fármacos , Factores de Tiempo
6.
Mol Neurobiol ; 56(8): 5815-5834, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30684218

RESUMEN

Current evidence suggests dementia and pathology in Alzheimer's Disease (AD) are both dependent and independent of amyloid processing and can be induced by multiple 'hits' on vital neuronal functions. Type 2 diabetes (T2D) poses the most important risk factor for developing AD after ageing and dysfunctional IR/PI3K/Akt signalling is a major contributor in both diseases. We developed a model of T2D, coupling subdiabetogenic doses of streptozotocin (STZ) with a human junk food (HJF) diet to more closely mimic the human condition. Over 35 weeks, this induced classic signs of T2D (hyperglycemia and insulin dysfunction) and a modest, but stable deficit in spatial recognition memory, with very little long-term modification of proteins in or associated with IR/PI3K/Akt signalling in CA1 of the hippocampus. Intracerebroventricular infusion of soluble amyloid beta 42 (Aß42) to mimic the early preclinical rise in Aß alone induced a more severe, but short-lasting deficits in memory and deregulation of proteins. Infusion of Aß on the T2D phenotype exacerbated and prolonged the memory deficits over approximately 4 months, and induced more severe aberrant regulation of proteins associated with autophagy, inflammation and glucose uptake from the periphery. A mild form of environmental enrichment transiently rescued memory deficits and could reverse the regulation of some, but not all protein changes. Together, these data identify mechanisms by which T2D could create a modest dysfunctional neuronal milieu via multiple and parallel inputs that permits the development of pathological events identified in AD and memory deficits when Aß levels are transiently effective in the brain.


Asunto(s)
Enfermedad de Alzheimer/epidemiología , Enfermedad de Alzheimer/etiología , Diabetes Mellitus Tipo 2/complicaciones , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/administración & dosificación , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Conducta Alimentaria , Hipocampo/patología , Hipocampo/ultraestructura , Humanos , Insulina/sangre , Masculino , Memoria , Trastornos de la Memoria/complicaciones , Modelos Biológicos , Fosforilación , Ratas Sprague-Dawley , Factores de Riesgo , Estreptozocina , Aumento de Peso
7.
Neuron ; 40(4): 695-701, 2003 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-14622575

RESUMEN

Recent research has revived interest in the possibility that previously consolidated memories need to reconsolidate when recalled to return to accessible long-term memory. Evidence suggests that both consolidation and reconsolidation of certain types of memory require protein synthesis, but whether similar molecular mechanisms are involved remains unclear. Here, we explore whether zif268, an activity-dependent inducible immediate early gene (IEG) required for consolidation of new memories, is also recruited for reconsolidation of recognition memory following reactivation. We show that when a consolidated memory for objects is recalled, zif268 mutant mice are impaired in further long-term but not short-term recognition memory. The impairment is specific to reactivation with the previously memorized objects in the relevant context, occurs in delayed recall, and does not recover over several days. These findings indicate that IEG-mediated transcriptional regulation in neurons is one common molecular mechanism for the storage of newly formed and reactivated recognition memories.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Proteínas Inmediatas-Precoces , Aprendizaje/fisiología , Memoria/fisiología , Neuronas/metabolismo , Reconocimiento en Psicología/fisiología , Factores de Transcripción/deficiencia , Animales , Encéfalo/metabolismo , Condicionamiento Psicológico/fisiología , Señales (Psicología) , Proteínas de Unión al ADN/genética , Proteína 1 de la Respuesta de Crecimiento Precoz , Conducta Exploratoria/fisiología , Genes Inmediatos-Precoces/genética , Genes Reguladores/genética , Memoria a Corto Plazo/fisiología , Ratones , Ratones Mutantes , Factores de Transcripción/genética
8.
Eur J Neurosci ; 27(11): 2985-98, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18588538

RESUMEN

The mechanisms underlying the induction of synaptic plasticity and the formation of long-term memory involve activation of cell-signalling cascades and protein modifications such as phosphorylation and dephosphorylation. Based on a protein candidate strategy, studies have identified several protein kinases and their substrates, which show an altered phosphorylation state during the early phases of long-term potentiation (LTP), yet only a limited number of synaptic phosphoproteins are known to be implicated in LTP. To identify new phosphoproteins associated with LTP, we have undertaken a proteomic study of phosphoproteins at different time points following the induction of LTP in the dentate gyrus in vivo (0, 15 and 90 min). For each time point, proteins from the dentate gyrus were separated by two-dimensional gel electrophoresis and stained with Pro-Q Diamond, a fluorescent stain specific for phosphoproteins. Fourteen proteins whose phosphorylation state varied significantly following LTP were identified using matrix-assisted laser desorption ionization/time of flight mass spectrometry and electrospray ionization-Orbitrap tandem mass spectrometry (MS/MS). They are involved in various cellular functions implicated in synaptic plasticity, such as intracellular signalling, axonal growth, exocytosis, protein synthesis and metabolism. Our results highlight new proteins whose phosphorylation or dephosphorylation is associated with LTP induction or maintenance. Further studies focusing on the regulation of specific phosphorylation sites will lead to greater understanding of the individual implications of these proteins in LTP as well as of their molecular interactions.


Asunto(s)
Giro Dentado/metabolismo , Potenciación a Largo Plazo/genética , Fosfoproteínas/análisis , Fosfoproteínas/metabolismo , Proteómica/métodos , Animales , Giro Dentado/efectos de los fármacos , Electroforesis en Gel Bidimensional , Colorantes Fluorescentes , Masculino , Espectrometría de Masas , Fosforilación , Ratas , Ratas Sprague-Dawley , Coloración y Etiquetado/métodos
9.
J Neurosci ; 26(22): 5888-93, 2006 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-16738230

RESUMEN

Activity-dependent synaptic plasticity and neurogenesis are two forms of brain plasticity that can participate in functional remodeling of neural networks during the formation of memories. We examined whether long-term potentiation (LTP) of excitatory synaptic transmission, a well characterized form of synaptic plasticity believed to play a critical role in memory formation, can regulate the rate of neurogenesis in the adult rat dentate gyrus in vivo. We first show that induction of LTP at medial perforant path-granule cell synapses stimulates the proliferation of progenitor cells in the dentate gyrus with a consequential long-term persistence of a larger population of surviving newborn cells. Using protocols to examine the effect of LTP on survival, we next show that LTP induction promotes survival of 1- to 2-week-old dentate granule cells. In no case did LTP appear to affect neuronal differentiation. Finally, we show that LTP induces expression of the plasticity-related transcription factor Zif268 in a substantial fraction of 2-week-old but not 1-week-old neurons, suggesting the prosurvival effect of LTP can be observed in the absence of LTP-mediated Zif268 induction in newborn cells. Our results indicate that electrically induced LTP in the dentate gyrus in vivo provides a cellular/molecular environment that favors both proliferation and survival of adult-generated neurons.


Asunto(s)
Giro Dentado/fisiología , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Regeneración Nerviosa/fisiología , Neuronas/fisiología , Animales , Diferenciación Celular , Giro Dentado/citología , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores/fisiología , Lateralidad Funcional , Masculino , Memoria/fisiología , Red Nerviosa/fisiología , Plasticidad Neuronal , Neuronas/citología , Ratas , Ratas Sprague-Dawley
10.
Neuroscientist ; 13(5): 492-505, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17901258

RESUMEN

A defining characteristic of the brain is its remarkable capacity to undergo activity-dependent functional and morphological remodeling via mechanisms of plasticity that form the basis of our capacity to encode and retain memories. Today, it is generally accepted that the neurobiological substrate of memories resides in activity-driven modifications of synaptic strength and structural remodeling of neural networks activated during learning. Since the discovery of long-term potentiation, the role of synaptic strengthening in learning and memory has been the subject of considerable investigation, and numerous studies have provided new insights into how this form of plasticity can subserve memory function. At the same time, other studies have explored the contribution of synaptic elimination or weakening; synaptogenesis, the growth of new synaptic connections and synapse remodeling; and more recently, neurogenesis, the birth and growth of new neurons in the adult brain. In this review, based on work in the hippocampus, the authors briefly outline recent advances in their understanding of the mechanisms and functional role of these four types of brain plasticity in the context of learning and memory. While they have long been considered as alternative mechanisms of plasticity underlying the storage of long-term memories, recent evidence suggests that they are functionally linked, suggesting the mechanisms underlying plasticity in the brain required for the formation and retention of memories are multifaceted.


Asunto(s)
Encéfalo/fisiología , Memoria/fisiología , Plasticidad Neuronal/fisiología , Sinapsis/fisiología , Animales , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Humanos , Potenciación a Largo Plazo/fisiología
11.
Genes Brain Behav ; 5 Suppl 2: 61-72, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16681801

RESUMEN

The function of mitogen-activated protein kinase (MAPK) in neurons has been the subject of considerable scrunity of late, and recent studies have given new insights into how this signalling cascade can regulate gene expression following cell-surface receptor activation. At the same time, a wealth of experimental data has demonstrated that the MAPK cascade is critically involved in the mechanisms underlying the type of enduring modification of neural networks required for the stability of memories, emphasizing the high level of interest in this signalling molecule. In this review, we briefly outline the main molecular events and mechanisms of the regulation of the MAPK cascade leading to transcriptional activation and summarize recent advances in our understanding of the functional role of this molecular signalling cascade in regulating brain plasticity, memory consolidation and memory reconsolidation.


Asunto(s)
Memoria/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Plasticidad Neuronal/fisiología , Transducción de Señal/fisiología , Activación Transcripcional/fisiología , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fosforilación , Sistemas de Mensajero Secundario/fisiología
12.
J Alzheimers Dis ; 51(4): 1157-73, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26923018

RESUMEN

Although it is well established that insulin/IGF and BDNF signaling are dysfunctionally regulated in Alzheimer's disease, there are very few studies documenting changes in major target proteins in different murine models of the disease. We investigated a panel of proteins in the PI3K-Akt and MAPK/ERK cascades in parietal cortex, dentate gyrus and CA1 in 13-month-old AßPP/PS1 transgenic mice to determine whether amyloid pathology is associated with basal dysregulation of these proteins or following exposure to novelty. The most striking effect we found was that there was little common regulation of proteins either by pathology alone or exposure to novelty across the three structures, suggesting dysfunctional mechanisms that occur simultaneously have important structure specificity. CA1 shared certain dysfunctional regulation of proteins in the MAPK/ERK cascade, but shared dysfunctional regulation of the PI3K/Akt cascade with the dentate gyrus. Changes in ERK/CREB in transgenic mice did not result in coordinated dysfunction of the downstream transcription factor, Egr1, as it was overexpressed in a normal manner following exposure to novelty. In the PI3K-Akt cascade, there was a flagrant increase in the levels of proteins associated with inflammation, such as NFκB, and structure specific regulation of proteins associated with autophagy, such as mTOR and FOXO1 and lack of regulation of Beclin-1. Finally, Beclin-1 was increased by novelty in wild-type mice but deficient in transgenic mice. Results are interpreted in terms of structure-specific dysfunctional regulation of signaling mechanisms associated with Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Presenilina-1/genética
13.
J Neurosci ; 23(12): 5354-60, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12832561

RESUMEN

Consolidation and reconsolidation of long-term memory have been shown to be dependent on the synthesis of new proteins, but the specific molecular mechanisms underlying these events remain to be elucidated. The mitogen-activated protein kinase (MAPK) pathway can trigger genomic responses in neurons, leading to changes in protein synthesis, and several studies have identified its pivotal role in synaptic plasticity and long-term memory formation. In this study, we analyze the involvement of this pathway in the consolidation and reconsolidation of long-term recognition memory, using an object recognition task. We show that inhibition of the MAPK pathway by intracerebroventricular injection of the MEK [MAPK/extracellular signal-regulated kinase (ERK)] inhibitor UO126 blocks consolidation of object recognition memory but does not affect short-term memory. Brain regions of the entorhinal cortex-hippocampal circuitry were analyzed for ERK activation, and it was shown that consolidation of recognition memory was associated with increased phosphorylation of ERK in the dentate gyrus and entorhinal cortex, although total expression of ERK was unchanged. We also report that inhibition of the MAPK pathway blocks reconsolidation of recognition memory, and this was shown to be dependent on reactivation of the memory trace by brief reexposure to the objects. In addition, reconsolidation of memory was associated with an increase in the phosphorylation of ERK in entorhinal cortex and CA1. In summary, our data show that the MAPK kinase pathway is required for both consolidation and reconsolidation of long-term recognition memory, and that this is associated with hyperphosphorylation of ERK in different subregions of the entorhinal cortex-hippocampal circuitry.


Asunto(s)
Hipocampo/fisiología , Memoria/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Red Nerviosa/fisiología , Reconocimiento en Psicología/fisiología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Corteza Entorrinal/enzimología , Corteza Entorrinal/fisiología , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Hipocampo/enzimología , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Memoria/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Red Nerviosa/enzimología , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reconocimiento en Psicología/efectos de los fármacos , Conducta Espacial/efectos de los fármacos , Conducta Espacial/fisiología
14.
Mol Neurobiol ; 27(3): 249-76, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12845151

RESUMEN

The prolongation of life and the rapidly increasing incidence of Alzheimer's disease have brought to the foreground the need for greater understanding of the etiology of the disease and the means to prevent or at least slow down the process. Out of this need the transgenic mouse and the production of synthetic amyloid peptides have been developed in an attempt to create experimental models of Alzheimer's disease that will help our understanding of the cellular and molecular mechanisms by which the pathology leads to memory dysfunction and to test potential therapeutic strategies. Despite 10 or so years of reasonably intensive research with these models, both fall short of producing a viable and faithful model of the complete pathology of Alzheimer's disease and the behavioral consequences are far from modelling the progressive decline in cognitive function. Here we review the advantages and the caveats associated with the two models in terms of the pathology, the associated memory dysfunction, and the effect on synaptic plasticity. Given the more recent advances that have been made in the understanding of the neurobiological changes that occur with the disease and with the consideration of other environmental effects, which have been clearly shown to have an impact on the progression of the disease in humans, we emphasis the advantage of pharmacological or environmental in transgenic mice or rodents injected with synthetic peptides that may prove to be more fruitful in our understanding of the memory deficits associated with the disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/metabolismo , Humanos , Ratones , Ratones Transgénicos
15.
Behav Brain Res ; 142(1-2): 17-30, 2003 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-12798262

RESUMEN

The immediate early genes (IEGs) are activated rapidly and transiently in response to a multitude of stimuli. The zif268 belongs to a category of regulatory IEGs that activate downstream target genes and is considered to be a triggering mechanism to activate the genomic response in neurons. Several studies have shown that zif268 mRNA is upregulated during different forms of associative learning, and following tetanic stimulation that induces long-lasting LTP. To date, there is a general consensus that zif268 activation may constitute a critical mechanism for the encoding of long-lasting memories, however this is based on relatively few studies. Given the fact that zif268 can be activated by a number of different types of stimuli, it becomes important to determine exactly how it may be implicated in memory. Examination of the current literature suggests that zif268 is necessary in the processing of several types of memory, however, it is not entirely clear what aspects of memory zif268 may be implicated in. Here, we review the existing literature and emphasise that understanding the signalling pathways that lead to activation of the IEGs and the downstream targets of these genes will advance our understanding of how functional activation of zif268 may be implicated in processing long-term memories.


Asunto(s)
Proteínas de Unión al ADN/genética , Aprendizaje/fisiología , Memoria/fisiología , Plasticidad Neuronal/genética , Factores de Transcripción/genética , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz , Regulación de la Expresión Génica , Genes Inmediatos-Precoces/genética , Haplorrinos , Hipocampo/fisiología , Proteínas Inmediatas-Precoces/genética , Ratones , ARN Mensajero/genética , Ratas , Sinapsis/fisiología
16.
Prog Mol Biol Transl Sci ; 122: 89-129, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24484699

RESUMEN

The capacity to remember our past experiences and organize our future draws on a number of cognitive processes that allow our brain to form and store neural representations that can be recalled and updated at will. In the brain, these processes require mechanisms of neural plasticity in the activated circuits, brought about by cellular and molecular changes within the neurons activated during learning. At the cellular level, a wealth of experimental data accumulated in recent years provides evidence that signaling from synapses to nucleus and the rapid regulation of the expression of immediate early genes encoding inducible, regulatory transcription factors is a key step in the mechanisms underlying synaptic plasticity and the modification of neural networks required for the laying down of memories. In the activated neurons, these transcriptional events are thought to mediate the activation of selective gene programs and subsequent synthesis of proteins, leading to stable functional and structural remodeling of the activated networks, so that the memory can later be reactivated upon recall. Over the past few decades, novel insights have been gained in identifying key transcriptional regulators that can control the genomic response of synaptically activated neurons. Here, as an example of this approach, we focus on one such activity-dependent transcription factor, Zif268, known to be implicated in neuronal plasticity and memory formation. We summarize current knowledge about the regulation and function of Zif268 in different types of brain plasticity and memory processes.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Memoria/fisiología , Plasticidad Neuronal/fisiología , Animales , Humanos
17.
Philos Trans R Soc Lond B Biol Sci ; 369(1633): 20130159, 2014 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-24298160

RESUMEN

It is well established that Zif268/Egr1, a member of the Egr family of transcription factors, is critical for the consolidation of several forms of memory; however, it is as yet uncertain whether increasing expression of Zif268 in neurons can facilitate memory formation. Here, we used an inducible transgenic mouse model to specifically induce Zif268 overexpression in forebrain neurons and examined the effect on recognition memory and hippocampal synaptic transmission and plasticity. We found that Zif268 overexpression during the establishment of memory for objects did not change the ability to form a long-term memory of objects, but enhanced the capacity to form a long-term memory of the spatial location of objects. This enhancement was paralleled by increased long-term potentiation in the dentate gyrus of the hippocampus and by increased activity-dependent expression of Zif268 and selected Zif268 target genes. These results provide novel evidence that transcriptional mechanisms engaging Zif268 contribute to determining the strength of newly encoded memories.


Asunto(s)
Giro Dentado/fisiología , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Regulación de la Expresión Génica/fisiología , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Percepción Espacial/fisiología , Análisis de Varianza , Animales , Giro Dentado/citología , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Ratones , Ratones Transgénicos , Transmisión Sináptica/fisiología
18.
Neurobiol Aging ; 33(6): 1122.e23-39, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22209410

RESUMEN

Brain-derived neurotrophic factor (BDNF) plays an important role in neuronal plasticity, learning, and memory. Levels of BDNF and its main receptor TrkB (TrkB.TK) have been reported to be decreased while the levels of the truncated TrkB (TrkB.T1) are increased in Alzheimer's disease. We show here that incubation with amyloid-ß increased TrkB.T1 receptor levels and decreased TrkB.TK levels in primary neurons. In vivo, APPswe/PS1dE9 transgenic mice (APdE9) showed an age-dependent relative increase in cortical but not hippocampal TrkB.T1 receptor levels compared with TrkB.TK. To investigate the role of TrkB isoforms in Alzheimer's disease, we crossed AP mice with mice overexpressing the truncated TrkB.T1 receptor (T1) or the full-length TrkB.TK isoform. Overexpression of TrkB.T1 in APdE9 mice exacerbated their spatial memory impairment while the overexpression of TrkB.TK alleviated it. These data suggest that amyloid-ß changes the ratio between TrkB isoforms in favor of the dominant-negative TrkB.T1 isoform both in vitro and in vivo and supports the role of BDNF signaling through TrkB in the pathophysiology and cognitive deficits of Alzheimer's disease.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Trastornos de la Memoria/metabolismo , Presenilina-1/genética , Receptor trkB/antagonistas & inhibidores , Transducción de Señal/genética , Precursor de Proteína beta-Amiloide/biosíntesis , Animales , Células Cultivadas , Femenino , Masculino , Trastornos de la Memoria/genética , Trastornos de la Memoria/psicología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Presenilina-1/biosíntesis , Receptor trkB/biosíntesis , Receptor trkB/genética
19.
Nat Rev Drug Discov ; 11(2): 141-68, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22293568

RESUMEN

Studies of psychiatric disorders have traditionally focused on emotional symptoms such as depression, anxiety and hallucinations. However, poorly controlled cognitive deficits are equally prominent and severely compromise quality of life, including social and professional integration. Consequently, intensive efforts are being made to characterize the cellular and cerebral circuits underpinning cognitive function, define the nature and causes of cognitive impairment in psychiatric disorders and identify more effective treatments. Successful development will depend on rigorous validation in animal models as well as in patients, including measures of real-world cognitive functioning. This article critically discusses these issues, highlighting the challenges and opportunities for improving cognition in individuals suffering from psychiatric disorders.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Trastornos Mentales/tratamiento farmacológico , Nootrópicos/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Cognición/efectos de los fármacos , Cognición/fisiología , Trastornos del Conocimiento/diagnóstico , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Epigénesis Genética/efectos de los fármacos , Predisposición Genética a la Enfermedad/genética , Humanos , Imagen por Resonancia Magnética , Trastornos Mentales/complicaciones , Pruebas Neuropsicológicas , Nootrópicos/farmacología , Desempeño Psicomotor/efectos de los fármacos , Factores de Riesgo , Estrés Psicológico/complicaciones
20.
Front Behav Neurosci ; 4: 177, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21120149

RESUMEN

The idea that an already consolidated memory can become destabilized after recall and requires a process of reconsolidation to maintain it for subsequent use has gained much credence over the past decade. Experimental studies in rodents have shown pharmacological, genetic, or injurious manipulation at the time of memory reactivation can disrupt the already consolidated memory. Despite the force of experimental data showing this phenomenon, a number of questions have remained unanswered and no consensus has emerged as to the conditions under which a memory can be disrupted following reactivation. To date most rodent studies of reconsolidation are based on negatively reinforced memories, in particular fear-associated memories, while the storage and stability of forms of memory that do not rely on explicit reinforcement have been less often studied. In this review, we focus on recognition memory, a paradigm widely used in humans to probe declarative memory. We briefly outline recent advances in our understanding of the processes and brain circuits involved in recognition memory and review the evidence that recognition memory can undergo reconsolidation upon reactivation. We also review recent findings suggesting that some molecular mechanisms underlying consolidation of recognition memory are similarly recruited after recall to ensure memory stability, while others are more specifically engaged in consolidation or reconsolidation. Finally, we provide novel data on the role of Rsk2, a mental retardation gene, and of the transcription factor zif268/egr1 in reconsolidation of object-location memory, and offer suggestions as to how assessing the activation of certain molecular mechanisms following recall in recognition memory may help understand the relative importance of different aspects of remodeling or updating long-lasting memories.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA