Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Mol Psychiatry ; 26(12): 7280-7295, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34561615

RESUMEN

Despite the central role of Rho GTPases in neuronal development, their functions in adult hippocampal neurogenesis remain poorly explored. Here, by using a retrovirus-based loss-of-function approach in vivo, we show that the atypical Rho GTPase Rnd2 is crucial for survival, positioning, somatodendritic morphogenesis, and functional maturation of adult-born dentate granule neurons. Interestingly, most of these functions are specific to granule neurons generated during adulthood since the deletion of Rnd2 in neonatally-born granule neurons only affects dendritogenesis. In addition, suppression of Rnd2 in adult-born dentate granule neurons increases anxiety-like behavior whereas its deletion in pups has no such effect, a finding supporting the adult neurogenesis hypothesis of anxiety disorders. Thus, our results are in line with the view that adult neurogenesis is not a simple continuation of earlier processes from development, and establish a causal relationship between Rnd2 expression and anxiety.


Asunto(s)
Ansiedad , Giro Dentado , Neurogénesis , Proteínas de Unión al GTP rho/metabolismo , Animales , Ansiedad/genética , Giro Dentado/metabolismo , Ratones , Neuronas/metabolismo , Proteínas de Unión al GTP rho/genética
2.
Cereb Cortex ; 29(8): 3527-3539, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-30215686

RESUMEN

In nonhuman mammals and in particular in rodents, most granule neurons of the dentate gyrus (DG) are generated during development and yet little is known about their properties compared with adult-born neurons. Although it is generally admitted that these populations are morphologically indistinguishable once mature, a detailed analysis of developmentally born neurons is lacking. Here, we used in vivo electroporation to label dentate granule cells (DGCs) generated in mouse embryos (E14.5) or in neonates (P0) and followed their morphological development up to 6 months after birth. By comparison with mature retrovirus-labeled DGCs born at weaning (P21) or young adult (P84) stages, we provide the evidence that perinatally born neurons, especially embryonically born cells, are morphologically distinct from later-born neurons and are thus easily distinguishable. In addition, our data indicate that semilunar and hilar GCs, 2 populations in ectopic location, are generated during the embryonic and the neonatal periods, respectively. Thus, our findings provide new insights into the development of the different populations of GCs in the DG and open new questions regarding their function in the brain.


Asunto(s)
Giro Dentado/embriología , Neuronas/citología , Animales , Animales Recién Nacidos , Cuerpo Celular , Dendritas/patología , Giro Dentado/citología , Giro Dentado/crecimiento & desarrollo , Electroporación , Embrión de Mamíferos , Proteínas Fluorescentes Verdes , Hipocampo/citología , Hipocampo/embriología , Hipocampo/crecimiento & desarrollo , Ratones
3.
Int J Mol Sci ; 21(19)2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-33019657

RESUMEN

The calcium/calmodulin-dependent protein kinase II (CaMKII) is a ubiquitous and central player in Ca2+ signaling that is best known for its functions in the brain. In particular, the α isoform of CaMKII has been the subject of intense research and it has been established as a central regulator of neuronal plasticity. In contrast, little attention has been paid to CaMKIIß, the other predominant brain isoform that interacts directly with the actin cytoskeleton, and the functions of CaMKIIß in this organ remain largely unexplored. However, recently, the perturbation of CaMKIIß expression has been associated with multiple neuropsychiatric and neurodevelopmental diseases, highlighting CAMK2B as a gene of interest. Herein, after highlighting the main structural and expression differences between the α and ß isoforms, we will review the specific functions of CaMKIIß, as described so far, in neuronal development and plasticity, as well as its potential implication in brain diseases.


Asunto(s)
Encéfalo/enzimología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Trastornos Mentales/genética , Trastornos del Neurodesarrollo/genética , Plasticidad Neuronal/fisiología , Neuronas/enzimología , Animales , Encéfalo/fisiopatología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/química , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Movimiento Celular , Regulación de la Expresión Génica , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Memoria/fisiología , Trastornos Mentales/enzimología , Trastornos Mentales/fisiopatología , Mutación , Trastornos del Neurodesarrollo/enzimología , Trastornos del Neurodesarrollo/fisiopatología , Neuronas/ultraestructura , Transducción de Señal , Sinapsis/enzimología , Sinapsis/ultraestructura
4.
Mol Psychiatry ; 23(11): 2209-2226, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29712998

RESUMEN

Perturbation of CaMKIIß expression has been associated with multiple neuropsychiatric diseases, highlighting CaMKIIß as a gene of interest. Yet, in contrast to CaMKIIα, the specific functions of CaMKIIß in the brain remain poorly explored. Here, we reveal a novel function for this CaMKII isoform in vivo during neuronal development. By using in utero electroporation, we show that CaMKIIß is an important regulator of radial migration of projection neurons during cerebral cortex development. Knockdown of CaMKIIß causes accelerated migration of nascent pyramidal neurons, whereas overexpression of CaMKIIß inhibits migration, demonstrating that precise regulation of CaMKIIß expression is required for correct neuronal migration. More precisely, CaMKIIß controls the multipolar-bipolar transition in the intermediate zone and locomotion in the cortical plate through its actin-binding and -bundling activities. In addition, our data indicate that a fine-tuned balance between CaMKIIß and cofilin activities is necessary to ensure proper migration of cortical neurons. Thus, our findings define a novel isoform-specific function for CaMKIIß, demonstrating that CaMKIIß has a major biological function in the developing brain.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Movimiento Celular/fisiología , Corteza Cerebral/fisiología , Neurogénesis/fisiología , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Corteza Cerebral/metabolismo , Embrión de Mamíferos/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Proteínas de Microfilamentos/genética , Trastornos del Neurodesarrollo/genética , Neurogénesis/genética , Neuronas/metabolismo , Cultivo Primario de Células , Isoformas de Proteínas/metabolismo , Células Piramidales/metabolismo
5.
Genes Dev ; 25(9): 930-45, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21536733

RESUMEN

Proneural genes such as Ascl1 are known to promote cell cycle exit and neuronal differentiation when expressed in neural progenitor cells. The mechanisms by which proneural genes activate neurogenesis--and, in particular, the genes that they regulate--however, are mostly unknown. We performed a genome-wide characterization of the transcriptional targets of Ascl1 in the embryonic brain and in neural stem cell cultures by location analysis and expression profiling of embryos overexpressing or mutant for Ascl1. The wide range of molecular and cellular functions represented among these targets suggests that Ascl1 directly controls the specification of neural progenitors as well as the later steps of neuronal differentiation and neurite outgrowth. Surprisingly, Ascl1 also regulates the expression of a large number of genes involved in cell cycle progression, including canonical cell cycle regulators and oncogenic transcription factors. Mutational analysis in the embryonic brain and manipulation of Ascl1 activity in neural stem cell cultures revealed that Ascl1 is indeed required for normal proliferation of neural progenitors. This study identified a novel and unexpected activity of the proneural gene Ascl1, and revealed a direct molecular link between the phase of expansion of neural progenitors and the subsequent phases of cell cycle exit and neuronal differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Telencéfalo/citología , Telencéfalo/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Estudio de Asociación del Genoma Completo , Ratones , Embarazo
6.
Sci Rep ; 14(1): 5022, 2024 02 29.
Artículo en Inglés | MEDLINE | ID: mdl-38424161

RESUMEN

The dentate gyrus (DG) of the hippocampus is a mosaic of dentate granule neurons (DGNs) accumulated throughout life. While many studies focused on the morpho-functional properties of adult-born DGNs, much less is known about DGNs generated during development, and in particular those born during embryogenesis. One of the main reasons for this gap is the lack of methods available to specifically label and manipulate embryonically-born DGNs. Here, we have assessed the relevance of the PenkCre mouse line as a genetic model to target this embryonically-born population. In young animals, PenkCre expression allows to tag neurons in the DG with positional, morphological and electrophysiological properties characteristic of DGNs born during the embryonic period. In addition, PenkCre+ cells in the DG are distributed in both blades along the entire septo-temporal axis. This model thus offers new possibilities to explore the functions of this underexplored population of embryonically-born DGNs.


Asunto(s)
Giro Dentado , Neuronas , Animales , Ratones , Giro Dentado/fisiología , Neuronas/fisiología , Hipocampo , Neurogénesis/fisiología
7.
Cereb Cortex ; 21(7): 1695-702, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21127017

RESUMEN

Vascular-specific growth factor angiopoietin-2 (Ang2) is mainly involved during vascular network setup. Recently, Ang2 was suggested to play a role in adult neurogenesis, affecting migration and differentiation of adult neuroblasts in vitro. However, to date, no data have reported an effect of Ang2 on neurogenesis during embryonic development. As we detected Ang2 expression in the developing cerebral cortex at embryonic day E14.5 and E16.5, we used in utero electroporation to knock down Ang2 expression in neuronal progenitors located in the cortical ventricular zone (VZ) to examine the role of Ang2 in cortical embryonic neurogenesis. Using this strategy, we showed that radial migration from the VZ toward the cortical plate of Ang2-knocked down neurons is altered as well as their morphology. In parallel, we observed a perturbation of intermediate progenitor population and the surrounding vasculature. Taken together, our results show for the first time that, in addition to its role during brain vasculature setup, Ang2 is also involved in embryonic cortical neurogenesis and especially in the radial migration of projection neurons.


Asunto(s)
Angiopoyetina 2/fisiología , Neurogénesis/fisiología , Telencéfalo/embriología , Telencéfalo/crecimiento & desarrollo , Angiopoyetina 2/genética , Animales , Animales Recién Nacidos , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Femenino , Técnicas de Silenciamiento del Gen/métodos , Ratones , Neurogénesis/genética , Neuronas/citología , Neuronas/fisiología , Embarazo , Telencéfalo/irrigación sanguínea
8.
Prog Neurobiol ; 219: 102364, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36244613

RESUMEN

Hippocampal adult neurogenesis is involved in many memory processes from learning, to remembering and forgetting. However, whether or not the stimulation of adult neurogenesis is a sufficient condition to improve memory performance remains unclear. Here, we developed and validated, using ex-vivo electrophysiology, a chemogenetic approach that combines selective tagging and activation of discrete adult-born neuron populations. Then we demonstrated that, in rats, this activation can improve accuracy and strength of remote memory. These results show that stimulation of adult-born neuron activity can counteract the natural fading of memory traces that occurs with the passage of time. This opens up new avenues for treating memory problems that may arise over time.


Asunto(s)
Memoria a Largo Plazo , Neurogénesis , Ratas , Animales , Neurogénesis/fisiología , Memoria a Largo Plazo/fisiología , Memoria/fisiología , Hipocampo/fisiología , Aprendizaje/fisiología
9.
Small GTPases ; 12(5-6): 336-357, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33054516

RESUMEN

Rnd proteins constitute a subfamily of Rho GTPases represented in mammals by Rnd1, Rnd2 and Rnd3. Despite their GTPase structure, their specific feature is the inability to hydrolyse GTP-bound nucleotide. This aspect makes them atypical among Rho GTPases. Rnds are regulated for their expression at the transcriptional or post-transcriptional levels and they are activated through post-translational modifications and interactions with other proteins. Rnd proteins are mainly involved in the regulation of the actin cytoskeleton and cell proliferation. Whereas Rnd3 is ubiquitously expressed, Rnd1 and 2 are tissue-specific. Increasing data has described their important role during development and diseases. Herein, we describe their involvement in physiological and pathological conditions with a focus on the neuronal and vascular systems, and summarize their implications in tumorigenesis.


Asunto(s)
Neoplasias/fisiopatología , Enfermedades del Sistema Nervioso/fisiopatología , Enfermedades Vasculares/fisiopatología , Proteínas de Unión al GTP rho/metabolismo , Humanos , Neoplasias/enzimología , Enfermedades del Sistema Nervioso/enzimología , Enfermedades Vasculares/enzimología
10.
Nat Commun ; 12(1): 1778, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33741954

RESUMEN

Memory reconsolidation, the process by which memories are again stabilized after being reactivated, has strengthened the idea that memory stabilization is a highly plastic process. To date, the molecular and cellular bases of reconsolidation have been extensively investigated particularly within the hippocampus. However, the role of adult neurogenesis in memory reconsolidation is unclear. Here, we combined functional imaging, retroviral and chemogenetic approaches in rats to tag and manipulate different populations of rat adult-born neurons. We find that both mature and immature adult-born neurons are activated by remote memory retrieval. However, only specific silencing of the adult-born neurons immature during learning impairs remote memory retrieval-induced reconsolidation. Hence, our findings show that adult-born neurons immature during learning are required for the maintenance and update of remote memory reconsolidation.


Asunto(s)
Aprendizaje/fisiología , Consolidación de la Memoria/fisiología , Memoria a Largo Plazo/fisiología , Neuronas/fisiología , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Hipocampo/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Microscopía Confocal , Neuronas/metabolismo , Biosíntesis de Proteínas/genética , Biosíntesis de Proteínas/fisiología , Ratas Sprague-Dawley , Factores de Tiempo
11.
Methods Mol Biol ; 2047: 513-523, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31552674

RESUMEN

In utero electroporation is a rapid and powerful technique to study the development of many brain regions. This approach presents several advantages over other methods to study specific steps of brain development in vivo, from proliferation to synaptic integration. Here, we describe in detail the individual steps necessary to carry out the technique. We also highlight the variations that can be implemented to target different cerebral structures and to study specific steps of development.


Asunto(s)
Encéfalo/embriología , Electroporación/métodos , Útero/embriología , Animales , Embrión de Mamíferos/metabolismo , Femenino , Ratones
12.
Biochem Biophys Res Commun ; 377(2): 400-406, 2008 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-18848521

RESUMEN

This study demonstrates that a prolyl hydroxylase inhibitor, FG-0041, is able, in combination with the ROCK inhibitor, Y-27632, to initiate differentiation of mesenchymal stem cells (MSCs) into neuron-like cells. FG-0041/Y-27632 co-treatment provokes morphological changes into neuron-like cells, increases neuronal marker expression and provokes modifications of cell cycle-related gene expression consistent with a cell cycle arrest of MSC, three events showing the engagement of MSC towards the neuronal lineage. Moreover, as we observed in our previous studies with cobalt chloride and desferroxamine, the activation of HIF-1 by this prolyl hydroxylase inhibitor is potentiated by Y-27632 which could explain at least in part the effect of this co-treatment on MSC neuronal differentiation. In addition, we show that this co-treatment enhances neurite outgrowth and tyrosine hydroxylase expression in PC12 cells. Altogether, these results evidence that concomitant inhibition of prolyl hydroxylases and ROCK represents a relevant protocol to initiate neuronal differentiation.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/efectos de los fármacos , Neuronas/citología , Fenantrolinas/farmacología , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Quinasas Asociadas a rho/antagonistas & inhibidores , Amidas/farmacología , Animales , Diferenciación Celular/genética , Linaje de la Célula , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Factor 1 Inducible por Hipoxia/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/enzimología , Neuritas/enzimología , Neuritas/fisiología , Neuronas/enzimología , Células PC12 , Piridinas/farmacología , Ratas
13.
Behav Brain Res ; 178(2): 235-43, 2007 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-17250902

RESUMEN

Excitotoxic lesion of the striatum provides a useful model for evaluating the excitotoxic processes involved in neurological disorders, in particular stroke diseases. The behavioural outcome after such injury is however poorly described. We have therefore investigated the potential behavioural deficits induced by a NMDA-induced excitotoxic unilateral lesion of the lateral part of the striatum, by comparison with a PBS striatal injection (sham procedure), and non-operated mice behaviour. Three groups of male adult Swiss mice were constituted: unilateral NMDA (20 nmol striatal NMDA injection), sham (striatal PBS injection), and control (healthy non-operated mice). From 14 to 29 days post-surgery, sensorimotor and mnesic tests were performed in all groups. After euthanasia, immunohistochemical stainings (NeuN and GFAP) were performed in order to assess the size of the lesion. Straight runway and passive avoidance performances revealed mild deficits related to the excitotoxic NMDA-induced lesion as compared to the sham procedure. Moreover, accelerated rotarod and Morris water maze acquisition performances also revealed deficits related to the surgery, i.e. observed in sham-operated as compared to control mice. NeuN staining revealed no striatal lesion in the sham and non-operated groups in contrast to the NMDA-injected group in which the volume of infarcted striatum was 2.4+/-0.3mm3. GFAP staining revealed a glial reaction in the lesioned striatum of NMDA animals and at the PBS injection site in sham animals. These results suggest that NMDA-induced excitotoxic lesion induces subtle long-term behavioural deficits in mice. Moreover, this study shows the importance of the sham group to investigate the behavioural deficits after excitotoxic lesion models in mice.


Asunto(s)
Reacción de Prevención/fisiología , Lateralidad Funcional/fisiología , Aprendizaje por Laberinto/fisiología , Actividad Motora/fisiología , Neostriado/fisiología , Análisis de Varianza , Animales , Lesiones Encefálicas/inducido químicamente , Estudios de Seguimiento , Masculino , Ratones , Destreza Motora/fisiología , N-Metilaspartato , Neostriado/lesiones , Neurotoxinas , Prueba de Desempeño de Rotación con Aceleración Constante , Estadísticas no Paramétricas , Factores de Tiempo
14.
Elife ; 62017 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-28682239

RESUMEN

The amyloid precursor protein (APP) harbors physiological roles at synapses and is central to Alzheimer's disease (AD) pathogenesis. Evidence suggests that APP intracellular domain (AICD) could regulate synapse function, but the underlying molecular mechanisms remain unknown. We addressed AICD actions at synapses, per se, combining in vivo AICD expression, ex vivo AICD delivery or APP knock-down by in utero electroporation of shRNAs with whole-cell electrophysiology. We report a critical physiological role of AICD in controlling GluN2B-containing NMDA receptors (NMDARs) at immature excitatory synapses, via a transcription-dependent mechanism. We further show that AICD increase in mature neurons, as reported in AD, alters synaptic NMDAR composition to an immature-like GluN2B-rich profile. This disrupts synaptic signal integration, via over-activation of SK channels, and synapse plasticity, phenotypes rescued by GluN2B antagonism. We provide a new physiological role for AICD, which becomes pathological upon AICD increase in mature neurons. Thus, AICD could contribute to AD synaptic failure.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/patología , Neurogénesis/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Ratones , Dominios Proteicos , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
15.
Clin Cancer Res ; 11(3): 993-9, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15709164

RESUMEN

PURPOSE: Expression of erythropoietin (Epo) and its receptor (Epo-R) has been shown in various normal and neoplastic nonhematopoietic tissues. This study, in non-small cell lung carcinoma, was designed to investigate the previously unreported expression of Epo and Epo-R as well as hypoxia-inducible factor-1alpha (HIF-1alpha), which is known to control Epo expression. EXPERIMENTAL DESIGN: Samples from lung squamous cell carcinomas (n = 17) and adenocarcinomas (n = 12) were obtained from patients undergoing curative surgery. mRNA transcripts of Epo, Epo-R, soluble Epo-R (sEpo-R), HIF-1alpha, and factor inhibiting HIF-1 (FIH-1) were evaluated by reverse transcription-PCR, whereas localization of Epo, Epo-R, and HIF-1alpha was assessed by immunohistochemistry. RESULTS: Epo, Epo-R, sEpo-R, HIF-1alpha, and FIH-1 transcripts were detected by reverse transcription-PCR in all samples tested, but with heterogeneous levels of expression for Epo, Epo-R, and sEpo-R. Coordinated levels of mRNA were observed for HIF-1alpha and FIH-1.Epo was detected in carcinomatous cells by immunohistochemistry in 50% of samples and Epo-R was detected in 96% of samples. Co-expression of Epo and Epo-R was observed on contiguous sections from 50% of tumors. HIF-1alpha was immunolocalized in 80% of non-small cell lung carcinomas. CONCLUSION: Epo-R was expressed in almost all samples and Epo was expressed in one half of samples on immunohistochemistry and in 100% of samples by mRNA detection, suggesting a potential paracrine and/or autocrine role of endogenous Epo in non-small cell lung carcinoma. The detection of stabilized HIF-1alpha suggests a possible role in Epo expression. Moreover, in the light of these results, the potential interactions between therapeutic recombinant Epo and the putative neoplastic Epo/Epo-R signaling pathways must be considered.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Eritropoyetina/genética , Neoplasias Pulmonares/patología , Receptores de Eritropoyetina/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Eritropoyetina/análisis , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Inmunohistoquímica , Antígeno Ki-67/análisis , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Oxigenasas de Función Mixta , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Eritropoyetina/análisis , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Transcripción/análisis , Factores de Transcripción/genética
16.
Curr Protoc Neurosci ; 77: 2.26.1-2.26.18, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27696363

RESUMEN

Brain electroporation is a rapid and powerful approach to study neuronal development. In particular, this technique has become a method of choice for studying the process of radial migration of projection neurons in the embryonic cerebral cortex. This method has considerably helped to describe in detail the different steps of radial migration and to characterize the molecular mechanisms controlling this process. Delineating the complexities of neuronal migration is critical to our understanding not only of normal cerebral cortex formation but also of neurodevelopmental disorders resulting from neuronal migration defects. Here, we describe in detail the protocols to perform in utero or ex vivo electroporation of progenitor cells in the ventricular zone of the cerebral cortex with the aim of studying the process of radial migration of projection neurons during embryonic development. © 2016 by John Wiley & Sons, Inc.


Asunto(s)
Movimiento Celular/fisiología , Corteza Cerebral/citología , Neuronas/citología , Animales , Corteza Cerebral/fisiología , Electroporación , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Neurogénesis/fisiología , Embarazo , Células Madre/citología
17.
Front Neurosci ; 9: 19, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25705175

RESUMEN

The mammalian cerebral cortex contains a high variety of neuronal subtypes that acquire precise spatial locations and form long or short-range connections to establish functional neuronal circuits. During embryonic development, cortical projection neurons are generated in the areas lining the lateral ventricles and they subsequently undergo radial migration to reach the position of their final maturation within the cortical plate. The control of the neuroblast migratory behavior and the coordination of the migration process with other neurogenic events such as cell cycle exit, differentiation and final maturation are crucial to normal brain development. Among the key regulators of cortical neuron migration, the small GTP binding proteins of the Rho family and the atypical Rnd members play important roles in integrating intracellular signaling pathways into changes in cytoskeletal dynamics and motility behavior. Here we review the role of Rnd proteins during cortical neuronal migration and we discuss both the upstream mechanisms that regulate Rnd protein activity and the downstream molecular pathways that mediate Rnd effects on cell cytoskeleton.

18.
Methods Mol Biol ; 1082: 285-93, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24048941

RESUMEN

In utero electroporation is a rapid and powerful technique to study the development of many brain regions. This approach presents several advantages over other methods to study specific steps of brain development in vivo, from proliferation to synaptic integration. Here, we describe in detail the individual steps necessary to carry out the technique. We also highlight the variations that can be implemented to target different cerebral structures and to study specific steps of development.


Asunto(s)
Encéfalo/embriología , Electroporación/métodos , Animales , Encéfalo/citología , Encéfalo/cirugía , ADN/metabolismo , Ratones
19.
Front Cell Neurosci ; 8: 445, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25610373

RESUMEN

The cerebral cortex is the site of higher human cognitive and motor functions. Histologically, it is organized into six horizontal layers, each containing unique populations of molecularly and functionally distinct excitatory projection neurons and inhibitory interneurons. The stereotyped cellular distribution of cortical neurons is crucial for the formation of functional neural circuits and it is predominantly established during embryonic development. Cortical neuron development is a multiphasic process characterized by sequential steps of neural progenitor proliferation, cell cycle exit, neuroblast migration and neuronal differentiation. This series of events requires an extensive and dynamic remodeling of the cell cytoskeleton at each step of the process. As major regulators of the cytoskeleton, the family of small Rho GTPases has been shown to play essential functions in cerebral cortex development. Here we review in vivo findings that support the contribution of Rho GTPases to cortical projection neuron development and we address their involvement in the etiology of cerebral cortex malformations.

20.
Nat Commun ; 5: 3405, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24572910

RESUMEN

A transcriptional programme initiated by the proneural factors Neurog2 and Ascl1 controls successive steps of neurogenesis in the embryonic cerebral cortex. Previous work has shown that proneural factors also confer a migratory behaviour to cortical neurons by inducing the expression of the small GTP-binding proteins such as Rnd2 and Rnd3. However, the directionality of radial migration suggests that migrating neurons also respond to extracellular signal-regulated pathways. Here we show that the Plexin B2 receptor interacts physically and functionally with Rnd3 and stimulates RhoA activity in migrating cortical neurons. Plexin B2 competes with p190RhoGAP for binding to Rnd3, thus blocking the Rnd3-mediated inhibition of RhoA and also recruits RhoGEFs to directly stimulate RhoA activity. Thus, an interaction between the cell-extrinsic Plexin signalling pathway and the cell-intrinsic Ascl1-Rnd3 pathway determines the level of RhoA activity appropriate for cortical neuron migration.


Asunto(s)
Movimiento Celular , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Western Blotting , Células COS , Línea Celular Tumoral , Corteza Cerebral/citología , Corteza Cerebral/embriología , Chlorocebus aethiops , Transferencia Resonante de Energía de Fluorescencia , Hibridación in Situ , Ratones , Microscopía Confocal , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Unión Proteica , Interferencia de ARN , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA