Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Neurosci ; 34(34): 11274-87, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25143608

RESUMEN

Neuron-glial related cell adhesion molecule (NrCAM) is a regulator of axon growth and repellent guidance, and has been implicated in autism spectrum disorders. Here a novel postsynaptic role for NrCAM in Semaphorin3F (Sema3F)-induced dendritic spine remodeling was identified in pyramidal neurons of the primary visual cortex (V1). NrCAM localized to dendritic spines of star pyramidal cells in postnatal V1, where it was coexpressed with Sema3F. NrCAM deletion in mice resulted in elevated spine densities on apical dendrites of star pyramidal cells at both postnatal and adult stages, and electron microscopy revealed increased numbers of asymmetric synapses in layer 4 of V1. Whole-cell recordings in cortical slices from NrCAM-null mice revealed increased frequency of mEPSCs in star pyramidal neurons. Recombinant Sema3F-Fc protein induced spine retraction on apical dendrites of wild-type, but not NrCAM-null cortical neurons in culture, while re-expression of NrCAM rescued the spine retraction response. NrCAM formed a complex in brain with Sema3F receptor subunits Neuropilin-2 (Npn-2) and PlexinA3 (PlexA3) through an Npn-2-binding sequence (TARNER) in the extracellular Ig1 domain. A trans heterozygous genetic interaction test demonstrated that Sema3F and NrCAM pathways interacted in vivo to regulate spine density in star pyramidal neurons. These findings reveal NrCAM as a novel postnatal regulator of dendritic spine density in cortical pyramidal neurons, and an integral component of the Sema3F receptor complex. The results implicate NrCAM as a contributor to excitatory/inhibitory balance in neocortical circuits.


Asunto(s)
Encéfalo/citología , Moléculas de Adhesión Celular/fisiología , Espinas Dendríticas/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/ultraestructura , Factores de Edad , Animales , Animales Recién Nacidos , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Moléculas de Adhesión Celular/deficiencia , Células Cultivadas , Chlorocebus aethiops , Espinas Dendríticas/ultraestructura , Embrión de Mamíferos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Antagonistas del GABA/farmacología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/deficiencia , Bloqueadores de los Canales de Sodio/farmacología , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/ultraestructura
2.
PLoS One ; 8(9): e73000, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24023801

RESUMEN

NrCAM (Neuron-glial related cell adhesion molecule), a member of the L1 family of cell adhesion molecules, reversibly binds ankyrin and regulates axon growth, but it has not been studied for a role in retinotopic mapping. During development of retino-collicular topography, NrCAM was expressed uniformly in retinal ganglion cells (RGCs) along both mediolateral and anteroposterior retinal axes, and was localized on RGC axons within the optic tract and superior colliculus (SC). Anterograde tracing of RGC axons in NrCAM null mutant mice at P10, when the map resembles its mature form, revealed laterally displaced ectopic termination zones (eTZs) of axons from the temporal retina, indicating defective mediolateral topography, which is governed by ephrinB/EphBs. Axon tracing at P2 revealed that interstitial branch orientation of ventral-temporal RGC axons in NrCAM null mice was compromised in the medial direction, likely accounting for displacement of eTZs. A similar retinocollicular targeting defect in EphB mutant mice suggested that NrCAM and EphB interact to regulate mediolateral retino-collicular targeting. We found that EphB2 tyrosine kinase but not an EphB2 kinase dead mutant, phosphorylated NrCAM at a conserved tyrosine residue in the FIGQY ankyrin binding motif, perturbing ankyrin recruitment in NrCAM transfected HEK293 cells. Furthermore, the phosphorylation of NrCAM at FIGQY in SC was decreased in EphB1/3 and EphB1/2/3 null mice compared to WT, while phospho-FIGQY of NrCAM in SC was increased in EphB2 constitutively active (F620D/F620D) mice. These results demonstrate that NrCAM contributes to mediolateral retinocollicular axon targeting by regulating RGC branch orientation through a likely mechanism in which ephrinB/EphB phosphorylates NrCAM to modulate linkage to the actin cytoskeleton.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Retina/metabolismo , Colículos Superiores/metabolismo , Animales , Axones/metabolismo , Moléculas de Adhesión Celular/genética , Línea Celular , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Ratones Noqueados , Células Ganglionares de la Retina/metabolismo
3.
Cereb Cortex ; 23(1): 162-77, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22275477

RESUMEN

A novel function for the neural cell adhesion molecule (NCAM) was identified in ephrinA/EphA-mediated repulsion as an important regulatory mechanism for development of GABAergic inhibitory synaptic connections in mouse prefrontal cortex. Deletion of NCAM, EphA3, or ephrinA2/3/5 in null mutant mice increased the numbers and size of perisomatic synapses between GABAergic basket interneurons and pyramidal cells in the developing cingulate cortex (layers II/III). A functional consequence of NCAM loss was increased amplitudes and faster kinetics of miniature inhibitory postsynaptic currents in NCAM null cingulate cortex. NCAM and EphA3 formed a molecular complex and colocalized with the inhibitory presynaptic marker vesicular GABA transporter (VGAT) in perisomatic puncta and neuropil in the cingulate cortex. EphrinA5 treatment promoted axon remodeling of enhanced green fluorescent protein-labeled basket interneurons in cortical slice cultures and induced growth cone collapse in wild-type but not NCAM null mutant neurons. NCAM modified with polysialic acid (PSA) was required to promote ephrinA5-induced axon remodeling of basket interneurons in cortical slices, likely by providing a permissive environment for ephrinA5/EphA3 signaling. These results reveal a new mechanism in which NCAM and ephrinAs/EphA3 coordinate to constrain GABAergic interneuronal arborization and perisomatic innervation, potentially contributing to excitatory/inhibitory balance in prefrontal cortical circuitry.


Asunto(s)
Efrinas/metabolismo , Neuronas GABAérgicas/fisiología , Interneuronas/fisiología , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Plasticidad Neuronal/fisiología , Corteza Prefrontal/fisiología , Sinapsis/fisiología , Animales , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Transgénicos , Ácido gamma-Aminobutírico/metabolismo
4.
J Neurosci ; 31(4): 1545-58, 2011 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-21273439

RESUMEN

NrCAM is a neural cell adhesion molecule of the L1 family that has been linked to autism spectrum disorders, a disease spectrum in which abnormal thalamocortical connectivity may contribute to visual processing defects. Here we show that NrCAM interaction with neuropilin-2 (Npn-2) is critical for semaphorin 3F (Sema3F)-induced guidance of thalamocortical axon subpopulations at the ventral telencephalon (VTe), an intermediate target for thalamic axon sorting. Genetic deletion of NrCAM or Npn-2 caused contingents of embryonic thalamic axons to misproject caudally in the VTe. The resultant thalamocortical map of NrCAM-null mutants showed striking mistargeting of motor and somatosensory thalamic axon contingents to the primary visual cortex, but retinogeniculate targeting and segregation were normal. NrCAM formed a molecular complex with Npn-2 in brain and neural cells, and was required for Sema3F-induced growth cone collapse in thalamic neuron cultures, consistent with a vital function for NrCAM in Sema3F-induced axon repulsion. NrCAM-null mice displayed reduced responses to visual evoked potentials recorded from layer IV in the binocular zone of primary visual cortex (V1), particularly when evoked from the ipsilateral eye, indicating abnormal visual acuity and ocularity. These results demonstrate that NrCAM is required for normal maturation of cortical visual acuity, and suggest that the aberrant projection of thalamic motor and somatosensory axons to the visual cortex in NrCAM-null mutant mice impairs cortical functions.


Asunto(s)
Axones/fisiología , Moléculas de Adhesión Celular/fisiología , Corteza Motora/ultraestructura , Corteza Somatosensorial/ultraestructura , Tálamo/ultraestructura , Agudeza Visual , Corteza Visual/ultraestructura , Animales , Moléculas de Adhesión Celular/genética , Potenciales Evocados Visuales , Femenino , Conos de Crecimiento/fisiología , Masculino , Proteínas de la Membrana/fisiología , Ratones , Ratones Noqueados , Corteza Motora/embriología , Corteza Motora/crecimiento & desarrollo , Proteínas del Tejido Nervioso/fisiología , Neuropilina-2/genética , Neuropilina-2/fisiología , Corteza Somatosensorial/embriología , Corteza Somatosensorial/crecimiento & desarrollo , Tálamo/embriología , Tálamo/crecimiento & desarrollo , Corteza Visual/embriología , Corteza Visual/crecimiento & desarrollo
5.
Cereb Cortex ; 21(2): 401-12, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20576928

RESUMEN

Neural cell adhesion molecule close homolog of L1 (CHL1) is a regulator of topographic targeting of thalamic axons to the somatosensory cortex (S1) but little is known about its cooperation with other L1 class molecules. To investigate this, CHL1(-/-)/L1(-/y) double mutant mice were generated and analyzed for thalamocortical axon topography. Double mutants exhibited a striking posterior shift of axons from motor thalamic nuclei to the visual cortex (V1), which was not observed in single mutants. In wild-type (WT) embryos, L1 and CHL1 were coexpressed in the dorsal thalamus (DT) and on fibers along the thalamocortical projection in the ventral telencephalon and cortex. L1 and CHL1 colocalized on growth cones and neurites of cortical and thalamic neurons in culture. Growth cone collapse assays with WT and mutant neurons demonstrated a requirement for L1 and CHL1 in repellent responses to EphrinA5, a guidance factor for thalamic axons. L1 coimmunoprecipitated with the principal EphrinA5 receptors expressed in the DT (EphA3, EphA4, and EphA7), whereas CHL1 associated selectively with EphA7. These results implicate a novel mechanism in which L1 and CHL1 interact with individual EphA receptors and cooperate to guide subpopulations of thalamic axons to distinct neocortical areas essential for thalamocortical connectivity.


Asunto(s)
Axones/fisiología , Moléculas de Adhesión Celular/metabolismo , Corteza Cerebral/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Vías Nerviosas/fisiología , Tálamo/metabolismo , Aminoácidos/metabolismo , Animales , Axones/efectos de los fármacos , Axones/ultraestructura , Moléculas de Adhesión Celular/deficiencia , Células Cultivadas , Corteza Cerebral/citología , Embrión de Mamíferos , Efrina-A5/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Conos de Crecimiento/fisiología , Humanos , Inmunoprecipitación/métodos , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Molécula L1 de Adhesión de Célula Nerviosa/deficiencia , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Receptores de la Familia Eph/genética , Receptores de la Familia Eph/metabolismo , Tálamo/citología , Transfección/métodos
6.
J Neurosci ; 29(50): 15630-41, 2009 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20016077

RESUMEN

ALCAM [activated leukocyte cell adhesion molecule (BEN/SC-1/DM-GRASP)] is a transmembrane recognition molecule of the Ig superfamily (IgSF) containing five Ig domains (two V-type, three C2-type). Although broadly expressed in the nervous and immune systems, few of its developmental functions have been elucidated. Because ALCAM has been suggested to interact with the IgSF adhesion molecule L1, a determinant of retinocollicular mapping, we hypothesized that ALCAM might direct topographic targeting to the superior colliculus (SC) by serving as a substrate within the SC for L1 on incoming retinal ganglion cell (RGC) axons. ALCAM was expressed in the SC during RGC axon targeting and on RGC axons as they formed the optic nerve; however, it was downregulated distally on RGC axons as they entered the SC. Axon tracing with DiI revealed pronounced mistargeting of RGC axons from the temporal retina half of ALCAM null mice to abnormally lateral sites in the contralateral SC, in which these axons formed multiple ectopic termination zones. ALCAM null mutant axons were specifically compromised in medial orientation of interstitial branches, which is known to require the ankyrin binding function of L1. As a substrate, ALCAM-Fc protein promoted L1-dependent attachment of acutely dissociated retinal cells and an L1-expressing, ALCAM-negative cell line, consistent with an ALCAM-L1 heterophilic molecular interaction. Together, these results suggest a model in which ALCAM in the SC interacts with L1 on RGC axons to promote medial extension of RGC axon branches important for mediolateral axon targeting in the formation of retinocollicular maps.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado/fisiología , Mapeo Encefálico , Retina/fisiología , Colículos Superiores/fisiología , Molécula de Adhesión Celular del Leucocito Activado/genética , Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Animales , Animales Recién Nacidos , Mapeo Encefálico/métodos , Línea Celular Tumoral , Lateralidad Funcional/fisiología , Humanos , Ratones , Ratones Noqueados , Ratones Mutantes , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/fisiología , Células Ganglionares de la Retina/fisiología , Vías Visuales/fisiología
7.
Behav Brain Res ; 205(1): 123-31, 2009 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-19540269

RESUMEN

NRCAM (Neuronal Cell Adhesion Molecule) has an important role in axonal guidance and the organization of neural circuitry during brain development. Association analyses in human populations have identified NRCAM as a candidate gene for autism susceptibility. In the present study, we evaluated Nrcam-null mice for sociability, social novelty preference, and reversal learning as a model for the social deficits, repetitive behavior, and cognitive rigidity characteristic of autism. Prepulse inhibition of acoustic startle responses was also measured, to reflect sensorimotor-gating deficits in autism spectrum disorders. Assays for anxiety-like behavior in an elevated plus maze and open field, motor coordination, and olfactory ability in a buried food test were conducted to provide control measures for the interpretation of results. Overall, the loss of Nrcam led to behavioral alterations in sociability, acquisition of a spatial task, and reversal learning, dependent on sex. In comparison to male wild type mice, male Nrcam-null mutants had significantly decreased sociability in a three-chambered choice task. Low sociability in the male null mutants was not associated with changes in anxiety-like behavior, activity, or motor coordination. Male, but not female, Nrcam-null mice had small decreases in prepulse inhibition. Nrcam deficiency in female mice led to impaired acquisition of spatial learning in the Morris water maze task. Reversal learning deficits were observed in both male and female Nrcam-null mice. These results provide evidence that NRCAM mediates domains of function relevant to symptoms observed in autism.


Asunto(s)
Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/metabolismo , Discapacidades para el Aprendizaje/fisiopatología , Trastorno de la Conducta Social/fisiopatología , Animales , Ansiedad/fisiopatología , Trastorno Autístico , Moléculas de Adhesión Celular/genética , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos Mentales , Ratones , Ratones Noqueados , Pruebas Neuropsicológicas , Aprendizaje Inverso/fisiología , Caracteres Sexuales , Conducta Social , Percepción Espacial/fisiología
8.
J Neurosci ; 28(1): 177-88, 2008 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-18171935

RESUMEN

Dynamic modulation of adhesion provided by anchorage of axonal receptors with the cytoskeleton contributes to attractant or repellent responses that guide axons to topographic targets in the brain. The neural cell adhesion molecule L1 engages the spectrin-actin cytoskeleton through reversible linkage of its cytoplasmic domain to ankyrin. To investigate a role for L1 association with the cytoskeleton in topographic guidance of retinal axons to the superior colliculus, a novel mouse strain was generated by genetic knock-in that expresses an L1 point mutation (Tyr1229His) abolishing ankyrin binding. Axon tracing revealed a striking mistargeting of mutant ganglion cell axons from the ventral retina, which express high levels of ephrinB receptors, to abnormally lateral sites in the contralateral superior colliculus, where they formed multiple ectopic arborizations. These axons were compromised in extending interstitial branches in the medial direction, a normal response to the high medial to low lateral SC gradient of ephrinB1. Furthermore, ventral but not dorsal L1(Y1229H) retinal cells were impaired for ephrinB1-stimulated adhesion through beta1 integrins in culture. The retinocollicular phenotype of the L1(Tyr1229His) mutant provides the first evidence that L1 regulates topographic mapping of retinal axons through adhesion mediated by linkage to the actin cytoskeleton and functional interaction with the ephrinB/EphB targeting system.


Asunto(s)
Mapeo Encefálico , Regulación del Desarrollo de la Expresión Génica/fisiología , Molécula L1 de Adhesión de Célula Nerviosa/fisiología , Retina/metabolismo , Colículos Superiores/metabolismo , Aminoácidos/metabolismo , Animales , Animales Recién Nacidos , Ancirinas/fisiología , Axones/metabolismo , Histidina/genética , Inmunoprecipitación/métodos , Ratones , Ratones Transgénicos , Mutación , Molécula L1 de Adhesión de Célula Nerviosa/genética , Retina/crecimiento & desarrollo , Colículos Superiores/citología , Colículos Superiores/crecimiento & desarrollo , Tirosina/genética , Vías Visuales/crecimiento & desarrollo , Vías Visuales/fisiología
9.
J Neurosci ; 27(50): 13667-79, 2007 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-18077678

RESUMEN

We report a cooperation between the neural adhesion molecule close homolog of L1 (CHL1) and the semaphorin 3A (Sema3A) receptor, neuropilin 1 (Npn1), important for establishment of area-specific thalamocortical projections. CHL1 deletion in mice selectively disrupted the projection of somatosensory thalamic axons from the ventrobasal (VB) nuclei, causing them to shift caudally and target the visual cortex. At the ventral telencephalon, an intermediate target with graded Sema3A expression, VB axons were caudally shifted in CHL1- embryos and in Npn1(Sema-/-) mutants, in which axons are nonresponsive to Sema3A. CHL1 colocalized with Npn1 on thalamic axons, and associated with Npn1 through a sequence in the CHL1 Ig1 domain that was required for Sema3A-induced growth cone collapse. These results identify a novel function for CHL1 in thalamic axon responsiveness to ventral telencephalic cues, and demonstrate a role for CHL1 and Npn1 in establishment of proper targeting of specific thalamocortical projections.


Asunto(s)
Axones/fisiología , Moléculas de Adhesión Celular/fisiología , Vías Nerviosas/fisiología , Neuropilina-1/fisiología , Telencéfalo/fisiología , Tálamo/fisiología , Animales , Moléculas de Adhesión Celular/genética , Cruzamientos Genéticos , Conos de Crecimiento/fisiología , Ratones , Ratones Mutantes , Ratones Transgénicos , Vías Nerviosas/citología , Vías Nerviosas/embriología , Neuropilina-1/genética , Estructura Terciaria de Proteína/fisiología , Telencéfalo/citología , Telencéfalo/embriología , Tálamo/citología , Tálamo/embriología
10.
Brain Res Dev Brain Res ; 160(1): 1-8, 2005 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-16154207

RESUMEN

Cyclic GMP-dependent protein kinase type I (cGKI) is a key signaling intermediate important for synaptic potentiation in the hippocampus and cerebellum, but its expression and function in cortical development have not been elucidated. The expression of cGKI in the developing mouse neocortex was evaluated by immunofluorescence labeling, and effect of cGKI deletion on cortical development was studied in adult cGKI knockout mice. cGKI was expressed at highest levels at embryonic stages in young neurons and radial glial fibers, corresponding to the major period of radial migration and laminar development of pyramidal neurons (embryonic day E13.5-E14.5), declining upon maturation (E17.5-postnatal day P28). The cerebral cortex of homozygous null mutant mice lacking cGKI exhibited heterotopic collections of neurons in the upper cortical layers and abnormal invaginations of layer I, in accord with a neuronal migration or positioning defect. Some cGKI mutant mice displayed defects in midline development resulting in partial fusion of cerebral hemispheres with adjacent neuronal heterotopias. Apical dendrites of cortical pyramidal neurons were misoriented in the cerebral cortex of cGKI null mutants, as shown in reporter mice expressing yellow fluorescent protein in layer V pyramidal neurons and by Golgi impregnation. These results demonstrate a role for cGKI signaling in cortical development related to neuronal migration/positioning that is important for dendritic orientation and connectivity.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/genética , Neocórtex/anomalías , Neocórtex/enzimología , Malformaciones del Sistema Nervioso/enzimología , Neuronas/enzimología , Animales , Proteínas Bacterianas , Diferenciación Celular/genética , Movimiento Celular/genética , Forma de la Célula/genética , Coristoma/enzimología , Coristoma/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Dendritas/enzimología , Dendritas/patología , Inmunohistoquímica , Proteínas Luminiscentes , Ratones , Ratones Noqueados , Neocórtex/patología , Malformaciones del Sistema Nervioso/genética , Neuroglía/enzimología , Neuroglía/patología , Neuronas/patología , Células Piramidales/enzimología , Células Piramidales/patología , Tinción con Nitrato de Plata , Células Madre/enzimología , Células Madre/patología
11.
J Neurosci ; 25(18): 4659-71, 2005 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-15872114

RESUMEN

The extracellular region of the transmembrane neural cell adhesion molecule (NCAM-EC) is shed as a soluble fragment at elevated levels in the schizophrenic brain. A novel transgenic mouse line was generated to identify consequences on cortical development and function of expressing soluble NCAM-EC from the neuron-specific enolase promoter in the developing and mature neocortex and hippocampus. NCAM-EC transgenic mice exhibited a striking reduction in synaptic puncta of GABAergic interneurons in the cingulate, frontal association cortex, and amygdala but not hippocampus, as shown by decreased immunolabeling of glutamic acid decarboxylase-65 (GAD65), GAD67, and GABA transporter 1. Interneuron cell density was unaltered in the transgenic mice. Affected subpopulations of interneurons included basket interneurons evident in NCAM-EC transgenic mice intercrossed with a reporter line expressing green fluorescent protein and by parvalbumin staining. In addition, there appeared to be a reduction in excitatory synapses, as revealed by synaptophysin staining and apical dendritic spine density of cortical pyramidal cells. Behavioral analyses demonstrated higher basal locomotor activity of NCAM-EC mice and enhanced responses to amphetamine and (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate compared with wild-type controls. Transgenic mice were deficient in prepulse inhibition, which was restored by clozapine but not by haloperidol. Additionally, NCAM-EC mice were impaired in contextual and cued fear conditioning. These results suggested that elevated shedding of NCAM perturbs synaptic connectivity of GABAergic interneurons and produces abnormal behaviors that may be relevant to schizophrenia and other neuropsychiatric disorders.


Asunto(s)
Conducta Animal/fisiología , Interneuronas/metabolismo , Interneuronas/patología , Ratones Transgénicos/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Anfetamina/farmacología , Animales , Western Blotting/métodos , Recuento de Células/métodos , Estimulantes del Sistema Nervioso Central/farmacología , Condicionamiento Psicológico/fisiología , Maleato de Dizocilpina/farmacología , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Miedo , Técnica del Anticuerpo Fluorescente/métodos , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Glutamato Descarboxilasa/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Locomoción/efectos de los fármacos , Locomoción/genética , Ratones , Ratones Endogámicos C57BL , Moléculas de Adhesión de Célula Nerviosa/genética , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Coloración y Etiquetado/métodos , Sinaptofisina/metabolismo
12.
Neuron ; 44(3): 423-37, 2004 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-15504324

RESUMEN

We show that the neural cell recognition molecule Close Homolog of L1 (CHL1) is required for neuronal positioning and dendritic growth of pyramidal neurons in the posterior region of the developing mouse neocortex. CHL1 was expressed in pyramidal neurons in a high-caudal to low-rostral gradient within the developing cortex. Deep layer pyramidal neurons of CHL1-minus mice were shifted to lower laminar positions in the visual and somatosensory cortex and developed misoriented, often inverted apical dendrites. Impaired migration of CHL1-minus cortical neurons was suggested by strikingly slower rates of radial migration in cortical slices, failure to potentiate integrin-dependent haptotactic cell migration in vitro, and accumulation of migratory cells in the intermediate and ventricular/subventricular zones in vivo. The restriction of CHL1 expression and effects of its deletion in posterior neocortical areas suggests that CHL1 may regulate area-specific neuronal connectivity and, by extension, function in the visual and somatosensory cortex.


Asunto(s)
Corteza Cerebral/citología , Dendritas/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas/fisiología , Células Piramidales/fisiología , Animales , Animales Recién Nacidos , Western Blotting/métodos , Bromodesoxiuridina/metabolismo , Carbocianinas/metabolismo , Moléculas de Adhesión Celular , Recuento de Células/métodos , Movimiento Celular/fisiología , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiología , Embrión de Mamíferos , Técnica del Anticuerpo Fluorescente/métodos , Técnicas In Vitro , Integrina beta1/metabolismo , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal/métodos , Proteínas/genética , Factores de Tiempo
13.
J Neurosci ; 23(2): 530-8, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12533613

RESUMEN

The retinocollicular projection is a preferred axon guidance pathway for investigating molecular mechanisms of synaptic targeting in the mammalian CNS. Here we identify a previously unrecognized role of the L1 cell adhesion molecule in topographic mapping of retinal ganglion cell (RGC) axons to their targets in the mouse superior colliculus (SC). L1 was transiently expressed on RGC axons during axon growth and targeting. DiI labeling of retinal axons revealed that temporal axons of L1-minus mice bypassed correct target locations in the anterior SC, forming termination zones at incorrect posterior sites, which were often skewed along the mediolateral axis. During development of the retinotopic map L1-minus temporal axons extended across the anteroposterior axis of the SC like wild-type axons but failed to arborize at normal anterior target sites. L1-minus RGC axons exhibited normal crossing at the optic chiasm and fasciculation of the optic nerve. Results suggest that retinal axons require the function of L1 in addition to repellent EphA guidance receptors to achieve proper topographic mapping.


Asunto(s)
Axones/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/biosíntesis , Retina/citología , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Axones/ultraestructura , Efrina-A2/biosíntesis , Colorantes Fluorescentes , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Molécula L1 de Adhesión de Célula Nerviosa/deficiencia , Molécula L1 de Adhesión de Célula Nerviosa/genética , Nervio Óptico/citología , Nervio Óptico/crecimiento & desarrollo , Nervio Óptico/metabolismo , Fenotipo , Receptores de la Familia Eph/biosíntesis , Retina/embriología , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/ultraestructura , Colículos Superiores/citología , Vías Visuales/citología , Vías Visuales/crecimiento & desarrollo , Vías Visuales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...