Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Neurobiol Dis ; 183: 106163, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37270162

RESUMEN

Intellectual disability (ID) is a neurodevelopmental disorder associated with impaired cognitive and adaptive behaviors and represents a major medical issue. Although ID-patients develop behavioral problems and are diagnosed during childhood, most behavioral studies in rodent models have been conducted in adulthood, missing precocious phenotypes expressed during this critical time-window characterized by intense brain plasticity. Here, we selectively assessed postnatal ontogenesis of behavioral and cognitive processes, as well as postnatal brain development in the male Rsk2-knockout mouse model of the Coffin-Lowry syndrome, an X-linked disorder characterized by ID and neurological abnormalities. While Rsk2-knockout mice were born healthy, a longitudinal MRI study revealed a transient secondary microcephaly and a persistent reduction of hippocampal and cerebellar volumes. Specific behavioral parameters from postnatal day 4 (P4) unveiled delayed acquisition of sensory-motor functions and alterations of spontaneous and cognitive behaviors during adolescence, which together, represent hallmarks of neurodevelopmental disorders. Together, our results suggest for the first time that RSK2, an effector of the MAPK signaling pathways, plays a crucial role in brain and cognitive postnatal development. This study also provides new relevant measures to characterize postnatal cognitive development of mouse models of ID and to design early therapeutic approaches.


Asunto(s)
Síndrome de Coffin-Lowry , Discapacidad Intelectual , Animales , Ratones , Masculino , Discapacidad Intelectual/genética , Encéfalo , Cognición , Síndrome de Coffin-Lowry/genética , Modelos Animales de Enfermedad , Ratones Noqueados
2.
Neuroscience ; 463: 70-85, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33722673

RESUMEN

The mitogen-activated protein kinases (MAPK) are major signaling components of intracellular pathways required for memory consolidation. Mitogen- and stress-activated protein kinases 1 and 2 (MSK1 and MSK2) mediate signal transduction downstream of MAPK. MSKs are activated by Extracellular-signal Regulated Kinase 1/2 (ERK1/2) and p38 MAPK. In turn, they can activate cyclic AMP-response-element-binding protein (CREB), thereby modulating the expression of immediate early genes crucial for the formation of long-term memories. While MSK1 has been previously implicated in certain forms of learning and memory, little is known concerning MSK2. Our goal was to explore the respective contribution of MSK1 and MSK2 in hippocampal synaptic transmission and plasticity and hippocampal-dependent recognition memory. In Msk1- and Msk2-knockout mice, we evaluated object and object-place recognition memory, basal synaptic transmission, paired-pulse facilitation (PPF) and inhibition (PPI), and the capacity to induce and sustain long-term potentiation (LTP) in vivo. We also assessed the level of two proteins downstream in the MAPK/ERK1/2 pathway crucial for long-term memory, CREB and the immediate early gene (IEG) Early growth response 1 (EGR1). Loss of Msk1, but not of Msk2, affected excitatory synaptic transmission at perforant path-to-dentate granule cell synapses, altered short-term presynaptic plasticity, impaired selectively long-term spatial recognition memory, and decreased basal levels of CREB and its activated form. LTP in vivo and LTP-induced CREB phosphorylation and EGR1 expression were unchanged after Msk1 or Msk2 deletion. Our findings demonstrate a dissimilar contribution of MSKs proteins in cognitive processes and suggest that Msk1 loss-of-function only has a deleterious impact on neuronal activity and hippocampal-dependent memory consolidation.


Asunto(s)
Memoria , Proteína Quinasa 11 Activada por Mitógenos , Proteína Quinasa 8 Activada por Mitógenos , Transmisión Sináptica , Animales , Hipocampo/metabolismo , Potenciación a Largo Plazo , Ratones , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
3.
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
4.
Hum Mol Genet ; 29(12): 1950-1968, 2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-31943058

RESUMEN

The link between mutations associated with intellectual disability (ID) and the mechanisms underlying cognitive dysfunctions remains largely unknown. Here, we focused on PAK3, a serine/threonine kinase whose gene mutations cause X-linked ID. We generated a new mutant mouse model bearing the missense R67C mutation of the Pak3 gene (Pak3-R67C), known to cause moderate to severe ID in humans without other clinical signs and investigated hippocampal-dependent memory and adult hippocampal neurogenesis. Adult male Pak3-R67C mice exhibited selective impairments in long-term spatial memory and pattern separation function, suggestive of altered hippocampal neurogenesis. A delayed non-matching to place paradigm testing memory flexibility and proactive interference, reported here as being adult neurogenesis-dependent, revealed a hypersensitivity to high interference in Pak3-R67C mice. Analyzing adult hippocampal neurogenesis in Pak3-R67C mice reveals no alteration in the first steps of adult neurogenesis, but an accelerated death of a population of adult-born neurons during the critical period of 18-28 days after their birth. We then investigated the recruitment of hippocampal adult-born neurons after spatial memory recall. Post-recall activation of mature dentate granule cells in Pak3-R67C mice was unaffected, but a complete failure of activation of young DCX + newborn neurons was found, suggesting they were not recruited during the memory task. Decreased expression of the KCC2b chloride cotransporter and altered dendritic development indicate that young adult-born neurons are not fully functional in Pak3-R67C mice. We suggest that these defects in the dynamics and learning-associated recruitment of newborn hippocampal neurons may contribute to the selective cognitive deficits observed in this mouse model of ID.


Asunto(s)
Disfunción Cognitiva/genética , Discapacidad Intelectual/genética , Quinasas p21 Activadas/genética , Animales , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Proteína Doblecortina , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Discapacidad Intelectual/patología , Masculino , Ratones , Mutación/genética , Neurogénesis/genética , Neuronas/metabolismo , Neuronas/patología
5.
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
6.
Mol Neurobiol ; 56(1): 722-735, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29796989

RESUMEN

Hypothyroidism is a condition that becomes more prevalent with age. Patients with untreated hypothyroidism have consistently reported symptoms of severe cognitive impairments. In patients suffering hypothyroidism, thyroid hormone supplementation offers the prospect to alleviate the cognitive consequences of hypothyroidism; however, the therapeutic value of TH supplementation remains at present uncertain and the link between cellular modifications associated with hypothyroidism and neurodegeneration remains to be elucidated. In the present study, we therefore evaluated the molecular and behavioral consequences of T3 hormone replacement in an animal model of hypothyroidism. We have previously reported that the antithyroid molecule propylthiouracil (PTU) given in the drinking water favors cerebral atrophy, brain neuroinflammation, Aß production, Tau hyperphosphorylation, and altered plasticity-related cell-signaling pathways in the hippocampus in association with hippocampal-dependent spatial memory deficits. In the present study, our aim was to explore, in this model, the effect of hippocampal T3 signaling normalization on various molecular mechanisms involved in learning and memory that goes awry under conditions of hypothyroidism and to evaluate its potential for recovery of hippocampal-dependent memory deficits. We report that T3 supplementation can alleviate hippocampal-dependent memory impairments displayed by hypothyroid rats and normalize key markers of thyroid status in the hippocampus, of neuroinflammation, Aß production, and of cell-signaling pathways known to be involved in synaptic plasticity and memory function. Together, these findings suggest that normalization of hippocampal T3 signaling is sufficient to reverse molecular and cognitive dysfunctions associated with hypothyroidism.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Biomarcadores/metabolismo , Hipocampo/patología , Hipotiroidismo/fisiopatología , Plasticidad Neuronal/efectos de los fármacos , Transducción de Señal , Memoria Espacial/efectos de los fármacos , Hormonas Tiroideas/farmacología , Animales , Ansiedad/complicaciones , Ansiedad/patología , Conducta Animal/efectos de los fármacos , Hipotiroidismo/complicaciones , Hipotiroidismo/metabolismo , Hipotiroidismo/patología , Masculino , Propiltiouracilo/farmacología , Propiltiouracilo/uso terapéutico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/patología , Hormonas Tiroideas/uso terapéutico
7.
Neurobiol Dis ; 115: 69-81, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29627578

RESUMEN

Adult neurogenesis is involved in certain hippocampus-dependent cognitive functions and is linked to psychiatric diseases including intellectual disabilities. The Coffin-Lowry syndrome (CLS) is a developmental disorder caused by mutations in the Rsk2 gene and characterized by intellectual disabilities associated with growth retardation. How RSK2-deficiency leads to cognitive dysfunctions in CLS is however poorly understood. Here, using Rsk2 Knock-Out mice, we characterized the impact of RSK2 deficiency on adult hippocampal neurogenesis in vivo. We report that the absence of RSK2 does not affect basal proliferation, differentiation and survival of dentate gyrus adult-born neurons but alters the maturation progression of young immature newborn neurons. Moreover, when RSK2-deficient mice were submitted to spatial learning, in contrast to wild-type mice, proliferation of adult generated neurons was decreased and no pro-survival effect of learning was observed. Thus, learning failed to recruit a selective population of young newborn neurons in association with deficient long-term memory recall. Given the proposed role of the dentate gyrus and of adult-generated newborn neurons in hippocampal-dependent pattern separation function, we explored this function in a delayed non-matching to place task and in an object-place pattern separation task and report severe deficits in spatial pattern separation in Rsk2-KO mice. Together, this study reveals a previously unknown role for RSK2 in the early stages of maturation and learning-dependent involvement of adult-born dentate gyrus neurons. These alterations associated with a deficit in the ability of RSK2-deficient mice to finely discriminate relatively similar spatial configurations, may contribute to cognitive dysfunction in CLS.


Asunto(s)
Síndrome de Coffin-Lowry/fisiopatología , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Neurogénesis/fisiología , Proteínas Quinasas S6 Ribosómicas 90-kDa/deficiencia , Conducta Espacial/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Síndrome de Coffin-Lowry/genética , Hipocampo/patología , Masculino , Ratones , Ratones Noqueados , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética
8.
Neuromuscul Disord ; 26(11): 775-781, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27614574

RESUMEN

Duchenne muscular dystrophy (DMD) is associated with non-progressive cognitive dysfunction including hippocampal-dependent memory deficits. Loss of the cytoskeleton-associated dystrophin protein in central inhibitory synapses, associated with consequent alterations in GABAergic function and synaptic plasticity, has been proposed as a primary mechanism responsible for cognitive impairments. However, several lines of evidence suggest a multifactorial etiology involving alternative signaling pathways, some of which could affect neuronal survival. To determine whether changes in neuronal density in the hippocampus could contribute to the emergence of memory deficits, we undertook an unbiased stereological estimation of neuron number in the anterodorsal CA1 region of the hippocampus of the dystrophin-deficient mdx mouse model of DMD. We found a significant reduction (~34%) in the number of pyramidal neurons, with a heterogeneous magnitude of genotype differences along the hippocampal antero-posterior axis. This extends previous knowledge of brain morphofunctional alterations induced by dystrophin loss and suggests that putative mechanisms involved in neurogenesis and/or neuron survival might contribute to the emergence of hippocampal-dependent learning and memory deficits in DMD.


Asunto(s)
Región CA1 Hipocampal/patología , Distrofia Muscular de Duchenne/patología , Células Piramidales/patología , Animales , Recuento de Células , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos mdx
9.
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
10.
Mol Autism ; 6: 60, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26527530

RESUMEN

BACKGROUND: The Duchenne and Becker muscular dystrophies (DMD, BMD) show significant comorbid diagnosis for autism, and the genomic sequences encoding the proteins responsible for these diseases, the dystrophin and associated proteins, have been proposed as new candidate risk loci for autism. Dystrophin is expressed not only in muscles but also in central inhibitory synapses in the cerebellum, hippocampus, amygdala, and cerebral cortex, where it contributes to the organization of autism-associated trans-synaptic neurexin-neuroligin complexes and to the clustering of synaptic gamma-aminobutyric acid (GABA)A receptors. While brain defects due to dystrophin loss are associated with deficits in cognitive and executive functions, communication skills and social behavior, only a subpopulation of DMD patients meet the criteria for autism, suggesting that mutations in the dystrophin gene may confer a vulnerability to autism. The loss of dystrophin in the mdx mouse model of DMD has been associated with cognitive and emotional alterations, but social behavior and communication abilities have never been studied in this model. METHODS: Here, we carried out the first in-depth analysis of social behavior and ultrasonic communication in dystrophin-deficient mdx mice, using a range of socially relevant paradigms involving various degrees of executive and cognitive demands, from simple presentation of sexual olfactory stimuli to social choice situations and direct encounters with female and male mice of various genotypes. RESULTS: We identified context-specific alterations in social behavior and ultrasonic vocal communication in mdx mice during direct encounters in novel environments. Social behavior disturbances depended on intruders' genotype and behavior, suggesting alterations in executive functions and adaptive behaviors, and were associated with selective alterations of the development, rate, acoustic properties, and use of the ultrasonic vocal repertoire. CONCLUSIONS: This first evidence that a mutation impeding expression of brain dystrophin affects social behavior and communication sheds new light on critical cognitive, emotional, and conative factors contributing to the development of autistic-like traits in this disease model.

11.
Neurobiol Learn Mem ; 124: 111-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26190833

RESUMEN

Duchenne muscular dystrophy (DMD) is associated with language disabilities and deficits in learning and memory, leading to intellectual disability in a patient subpopulation. Recent studies suggest the presence of broader deficits affecting information processing, short-term memory and executive functions. While the absence of the full-length dystrophin (Dp427) is a common feature in all patients, variable mutation profiles may additionally alter distinct dystrophin-gene products encoded by separate promoters. However, the nature of the cognitive dysfunctions specifically associated with the loss of distinct brain dystrophins is unclear. Here we show that the loss of the full-length brain dystrophin in mdx mice does not modify the perception and sensorimotor gating of auditory inputs, as assessed using auditory brainstem recordings and prepulse inhibition of startle reflex. In contrast, both acquisition and long-term retention of cued and trace fear memories were impaired in mdx mice, suggesting alteration in a functional circuit including the amygdala. Spatial learning in the water maze revealed reduced path efficiency, suggesting qualitative alteration in mdx mice learning strategy. However, spatial working memory performance and cognitive flexibility challenged in various behavioral paradigms in water and radial-arm mazes were unimpaired. The full-length brain dystrophin therefore appears to play a role during acquisition of associative learning as well as in general processes involved in memory consolidation, but no overt involvement in working memory and/or executive functions could be demonstrated in spatial learning tasks.


Asunto(s)
Percepción Auditiva/fisiología , Encéfalo/fisiopatología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Distrofina/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatología , Distrofia Muscular de Duchenne/psicología , Filtrado Sensorial/fisiología , Estimulación Acústica , Animales , Reacción de Prevención/fisiología , Conducta Animal/fisiología , Encéfalo/metabolismo , Condicionamiento Clásico/fisiología , Modelos Animales de Enfermedad , Distrofina/genética , Potenciales Evocados Auditivos del Tronco Encefálico , Función Ejecutiva/fisiología , Miedo/fisiología , Aprendizaje por Laberinto/fisiología , Memoria a Largo Plazo/fisiología , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Reflejo de Sobresalto , Memoria Espacial/fisiología , Navegación Espacial/fisiología
12.
J Neurosci ; 35(19): 7575-86, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25972182

RESUMEN

In search for the mechanisms underlying complex forms of human memory, such as episodic recollection, a primary challenge is to develop adequate animal models amenable to neurobiological investigation. Here, we proposed a novel framework and paradigm that provides means to quantitatively evaluate the ability of rats to form and recollect a combined knowledge of what happened, where it happened, and when or in which context it happened (referred to as episodic-like memory) after a few specific episodes in situations as close as possible to a paradigm we recently developed to study episodic memory in humans. In this task, rats have to remember two odor-drink associations (what happened) encountered in distinct locations (where it happened) within two different multisensory enriched environments (in which context/occasion it happened), each characterized by a particular combination of odors and places. By analyzing licking behavior on each drinking port, we characterized quantitatively individual recollection profiles and showed that rats are able to incidentally form and recollect an accurate, long-term integrated episodic-like memory that can last ≥ 24 d after limited exposure to the episodes. Placing rats in a contextually challenging recollection situation at recall reveals the ability for flexible use of episodic memory as described in humans. We further report that reversible inactivation of the dorsal hippocampus during recall disrupts the animal's capacity to recollect the complete episodic memory. Cellular imaging of c-Fos and Zif268 brain activation reveals that episodic memory recollection recruits a specific, distributed network of hippocampal-prefrontal cortex structures that correlates with the accuracy of the integrated recollection performance.


Asunto(s)
Aprendizaje por Asociación/fisiología , Mapeo Encefálico , Hipocampo/fisiología , Memoria/fisiología , Animales , Conducta de Ingestión de Líquido , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Agonistas de Receptores de GABA-A/farmacología , Hipocampo/efectos de los fármacos , Masculino , Muscimol/farmacología , Odorantes , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Long-Evans , Estadísticas no Paramétricas , Privación de Agua
13.
Biol Aujourdhui ; 209(3): 229-48, 2015.
Artículo en Francés | MEDLINE | ID: mdl-26820830

RESUMEN

A defining characteristic of the brain is its remarkable capacity to undergo activity-dependent functional and structural remodelling via mechanisms of plasticity that form the basis of our capacity to encode and retain memories. The prevailing model of how our brain stores new information about relationships between events or new abstract constructs suggests it resides in activity-driven modifications of synaptic strength and remodelling of neural networks brought about by cellular and molecular changes within the neurons activated during learning. To date, the idea that a form of activity-dependent synaptic plasticity known as long-term potentiation, or LTP, and the associated synaptic growth play a central role in the laying down of memories has received considerable support. Beyond this mechanism of plasticity at the synapse, adult neurogenesis, i.e. the birth and growth of new neurons, is another form of neural plasticity that occurs continuously in defined brain regions such as the dentate gyrus of the hippocampus. Here, based on work in the hippocampus, we review the processes and mechanisms of the generation and selection of new neurons in the adult brain and the accumulating evidence that supports the idea that this form of neural plasticity is essential to store and lead to retrievable hippocampal-dependent memories.


Asunto(s)
Encéfalo/fisiología , Memoria/fisiología , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Adulto , Células Madre Adultas/citología , Células Madre Adultas/fisiología , Animales , Encéfalo/citología , Humanos , Aprendizaje/fisiología , Nicho de Células Madre/fisiología
14.
Hippocampus ; 24(11): 1381-93, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24978200

RESUMEN

The multifactorial causes impacting the risk of developing sporadic forms of Alzheimer's disease (AD) remain to date poorly understood. Epidemiologic studies in humans and research in rodents have suggested that hypothyroidism could participate in the etiology of AD. Recently, we reported that adult-onset hypothyroidism in rats favors ß-amyloid peptide production in the hippocampus. Here, using the same hypothyroidism model with the antithyroid molecule propythiouracyl (PTU), we further explored AD-related features, dysfunctional cell-signaling mechanisms and hippocampal-dependent learning and memory. In vivo MRI revealed a progressive decrease in cerebral volume of PTU-treated rats. In the hippocampus, hypothyroidism resulted in tau hyperphosphorylation and increases in several proinflammatory cytokines. These modifications were associated with impaired spatial memory and reduced hippocampal expression of signaling molecules important for synaptic plasticity and memory, including neurogranin, CaMKII, ERK, GSK3ß, CREB, and expression of the transcription factor EGR1/Zif268. These data strengthen the idea that hypothyroidism represents an important factor influencing the risk of developing sporadic forms of AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Hipocampo/fisiopatología , Hipotiroidismo/fisiopatología , Trastornos de la Memoria/fisiopatología , Memoria Espacial , Enfermedad de Alzheimer/patología , Animales , Ansiedad/patología , Ansiedad/fisiopatología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hipocampo/patología , Hipotiroidismo/patología , Masculino , Trastornos de la Memoria/patología , Actividad Motora/fisiología , Neuroinmunomodulación/fisiología , Tamaño de los Órganos , Fosforilación , Distribución Aleatoria , Ratas Wistar , Proteínas tau/metabolismo
15.
PLoS One ; 9(3): e90056, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24658054

RESUMEN

Melatonin receptor expression exhibits profound developmental changes through poorly understood mechanisms. In mammals, a current model suggests that pubertal reactivation of gonadotrophin-releasing hormone (GnRH) secretion down-regulates MT1 melatonin receptors in pituitary gonadotroph cells, via the induction of early growth response factor-1 (EGR-1). Here we have examined this model by testing the hypotheses that inhibition of Mt1 expression by GnRH occurs directly in gonadotroph cells, can be reversed in adulthood by blockade of GnRH receptors, and requires EGR-1. We first confirmed the endogenous expression of Mt1 mRNA in the αT3-1 gonadotroph cell line. Stimulation of these cells with a GnRH agonist resulted in a rapid increase of Egr-1 mRNA expression, which peaked after 30-60 minutes, and a more prolonged elevation of nuclear EGR-1 immunoreactivity. Moreover, the GnRH agonist significantly decreased Mt1 mRNA. We then treated adult male rats with the GnRH antagonist cetrorelix or saline. After 4 weeks of daily injections, cetrorelix significantly reduced serum LH concentration and testis weight, with histological analysis confirming absence of spermatogenesis. Despite the successful inhibition of GnRH signalling, pituitary Mt1 expression was unchanged. Next we studied the proximal region of the rat Mt1 promoter. Consistent with previous work, over-expression of the transcription factor PITX-1 increased Mt1-luciferase reporter activity; this effect was dependent on the presence of consensus PITX-1 promoter binding regions. Over-expression of EGR-1 inhibited PITX-1-stimulated activity, even following mutation of the consensus EGR-1 binding site. Finally, we studied Egr1-/- mice and observed no difference in pituitary Mt1 expression between Egr1-/- and wild-type litter mates. This work demonstrates that GnRH receptor activation directly down-regulates Mt1 expression in gonadotroph cells. However, pituitary Mt1 expression in adults is unaltered by blockade of GnRH signalling or absence of EGR-1. Our data therefore suggest that melatonin receptor regulation by GnRH is not reversible in adulthood and doesn't require EGR-1.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Receptor de Melatonina MT1/genética , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Regulación de la Expresión Génica , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/farmacología , Antagonistas de Hormonas/farmacología , Masculino , Ratones , Factores de Transcripción Paired Box/metabolismo , Hipófisis/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptor de Melatonina MT1/metabolismo , Receptores LHRH/metabolismo
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.
Brain Struct Funct ; 219(1): 415-30, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23389809

RESUMEN

Over the past few years multiple studies have attempted to uncover molecular signatures of memory reconsolidation when compared to consolidation. In the present study we used immunocytochemical detection of the MAPK/ERK1/2 pathway, to track activated neuronal circuits in the hippocampus and amygdala recruited during the consolidation and reconsolidation of a contextual fear conditioning (CFC) memory. We report selective differences in magnitude and temporal dynamics of activated ERK1/2 signalling in different subregions of these two structures between the post-training and post-retrieval periods, except in the dentate gyrus, where the patterns of activation were similar. We then focused on this brain area to dissect out the patterns of downstream ERK1/2 signalling components, including the phosphorylation of MSK-1 and histone H3 on ser10, along with the induction of the Immediate Early Genes (IEGs) Arc/Arg3.1, c-Fos and Zif268/Egr1 following CFC training and retrieval. We found that the completion of the nucleosomal response as well as the induction of IEGs shorter during the reconsolidation period as compared to consolidation. Our results shed new light on the cellular mechanisms underlying the consolidation and reconsolidation processes engaged following CFC training and retrieval and further extend the notion that memory reconsolidation is not mechanistically a repetition of consolidation. In addition, we provide evidence that the strength of a previously established CFC memory is characterized by distinct patterns of ERK1/2 activation in different hippocampal and amygdalar subfields upon CFC memory recall. Our results emphasize the differences between consolidation and reconsolidation processes in relation to contextual fear memories.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Condicionamiento Clásico/fisiología , Miedo/fisiología , Genes Inmediatos-Precoces/fisiología , Hipocampo/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Recuerdo Mental/fisiología , Animales , Proteínas del Citoesqueleto/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Electrochoque/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factores de Tiempo
18.
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
19.
PLoS One ; 8(8): e72006, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977192

RESUMEN

Compelling evidence points to the existence of independent cellular processes involved in the consolidation and reconsolidation of memory. For instance, a double dissociation has been reported between hippocampal Extracellular-Regulated Kinase-1/2 (ERK1/2) activity being necessary for contextual fear conditioning (CFC) consolidation but not reconsolidation. Conversely, hippocampal expression of the immediate early gene Zif268 is necessary for CFC reconsolidation but not consolidation. Since we previously reported that ERK1/2 controls the transcription of Zif268 in the hippocampus, we examined the precise role of ERK1/2 activity and Zif268 gene expression dosage in CFC memory processing. For this, we first assessed performance of Zif268 homozygous and heterozygous mutant mice in a CFC paradigm. Whereas Zif268-/- mice displayed a deficit of both consolidation and reconsolidation, Zif268+/- mice displayed a selective deficit of reconsolidation only, therefore pointing to the relationship between Zif268 gene expression dosage and CFC memory processing. Zif268 gene expression dosage interfered with the reconsolidation process if and only if CFC memory was relatively recently encoded and directly reactivated. Furthermore, CFC memory strengthening previously reported to involve Zif268 expression in the hippocampus was spared in Zif268+/- mice. Finally, blocking ERK1/2 activity prior to CFC retrieval prevented the deficit of reconsolidation observed in Zif268+/- mice. Collectively, these results highlight a tight relationship between Zif268 gene expression dosage and CFC memory processing. They also suggest that ERK1/2 activity upon CFC memory recall is necessary for its retrieval, a prerequisite for its reactivation and subsequent reconsolidation.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Miedo , Recuerdo Mental , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Estimulación Acústica , Animales , Condicionamiento Psicológico , Reacción Cataléptica de Congelación/fisiología , Dosificación de Gen , Expresión Génica , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...