Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Histochem Cell Biol ; 146(2): 167-82, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27060907

RESUMEN

Thymus development is a complex process in which cell-to-cell interactions between thymocytes and thymic epithelial cells (TECs) are essential to allow a proper maturation of both thymic cell components. Although signals that control thymocyte development are well known, mechanisms governing TEC maturation are poorly understood, especially those that regulate the maturation of immature TEC populations during early fetal thymus development. In this study, we show that EphB2-deficient, EphB2LacZ and EphB3-deficient fetal thymuses present a lower number of cells and delayed maturation of DN cell subsets compared to WT values. Moreover, deficits in the production of chemokines, known to be involved in the lymphoid seeding into the thymus, contribute in decreased proportions of intrathymic T cell progenitors (PIRA/B(+)) in the mutant thymuses from early stages of development. These features correlate with increased proportions of MTS20(+) cells but fewer MTS20(-) cells from E13.5 onward in the deficient thymuses, suggesting a delayed development of the first epithelial cells. In addition, in vitro the lack of thymocytes or the blockade of Eph/ephrin-B-mediated cell-to-cell interactions between either thymocytes-TECs or TECs-TECs in E13.5 fetal thymic lobes coursed with increased proportions of MTS20(+) TECs. This confirms, for the first time, that the presence of CD45(+) cells, corresponding at these stages to DN1 and DN2 cells, and Eph/ephrin-B-mediated heterotypic or homotypic cell interactions between thymocytes and TECs, or between TECs and themselves, contribute to the early maturation of MTS20(+) TECs.


Asunto(s)
Efrina-B2/metabolismo , Efrina-B3/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Timo/citología , Timo/metabolismo , Animales , Efrina-B2/deficiencia , Efrina-B3/deficiencia , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Noqueados , Técnicas de Cultivo de Tejidos
2.
Cell Death Dis ; 5: e1207, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24810043

RESUMEN

Eph receptor tyrosine kinases and their membrane-bound ligands, ephrins, have a variety of roles in the developing and adult central nervous system that require direct cell-cell interactions; including regulating axon path finding, cell proliferation, migration and synaptic plasticity. Recently, we identified a novel pro-survival role for ephrins in the adult subventricular zone, where ephrinB3 blocks Eph-mediated cell death during adult neurogenesis. Here, we examined whether EphB3 mediates cell death in the adult forebrain following traumatic brain injury and whether ephrinB3 infusion could limit this effect. We show that EphB3 co-labels with microtubule-associated protein 2-positive neurons in the adult cortex and is closely associated with ephrinB3 ligand, which is reduced following controlled cortical impact (CCI) injury. In the complete absence of EphB3 (EphB3(-/-)), we observed reduced terminal deoxynucleotidyl transferase-dUTP nick end labeling (TUNEL), and functional improvements in motor deficits after CCI injury as compared with wild-type and ephrinB3(-/-) mice. We also demonstrated that EphB3 exhibits dependence receptor characteristics as it is cleaved by caspases and induces cell death, which is not observed in the presence of ephrinB3. Following trauma, infusion of pre-clustered ephrinB3-Fc molecules (eB3-Fc) into the contralateral ventricle reduced cortical infarct volume and TUNEL staining in the cortex, dentate gyrus and CA3 hippocampus of wild-type and ephrinB3(-/-) mice, but not EphB3(-/-) mice. Similarly, application of eB3-Fc improved motor functions after CCI injury. We conclude that EphB3 mediates cell death in the adult cortex through a novel dependence receptor-mediated cell death mechanism in the injured adult cortex and is attenuated following ephrinB3 stimulation.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Efrina-B3/administración & dosificación , Efrina-B3/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Receptor EphB3/metabolismo , Animales , Apoptosis/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Encéfalo/fisiopatología , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Línea Celular Tumoral , Citoprotección , Modelos Animales de Enfermedad , Efrina-B3/deficiencia , Efrina-B3/genética , Células HEK293 , Humanos , Infusiones Intraventriculares , Masculino , Ratones , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Degeneración Nerviosa , Neuronas/enzimología , Neuronas/patología , Receptor EphB3/deficiencia , Receptor EphB3/genética , Recuperación de la Función , Prueba de Desempeño de Rotación con Aceleración Constante , Factores de Tiempo , Transfección
3.
Immunol Cell Biol ; 91(2): 130-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23146940

RESUMEN

The signals that determine the survival/death of the thymic epithelial cells (TECs) component during embryonic development of the thymus are largely unknown. In this study, we combine different in vivo and in vitro experimental approaches to define the role played by the tyrosine kinase receptors EphB2 and EphB3 and their ligands, ephrinsB, in the survival of embryonic and newborn (NB) TECs. Our results conclude that EphB2 and EphB3 are involved in the control of TEC survival and that the absence of these molecules causes increased apoptotic TEC proportions that result in decreased numbers of thymic cells and a smaller-sized gland. Furthermore, in vitro studies using either EphB2-Fc or ephrinB1-Fc fusion proteins demonstrate that the blockade of Eph/ephrinB signalling increases TEC apoptosis, whereas its activation rescues TECs from cell death. In these assays, both heterotypic thymocyte-TEC and homotypic TEC-TEC interactions are important for Eph/ephrinB-mediated TEC survival.


Asunto(s)
Efrina-B2/metabolismo , Efrina-B3/metabolismo , Células Epiteliales/citología , Receptor EphB2/metabolismo , Receptor EphB3/metabolismo , Transducción de Señal , Timocitos/citología , Animales , Animales Recién Nacidos , Apoptosis , Comunicación Celular , Supervivencia Celular , Efrina-B2/deficiencia , Efrina-B3/deficiencia , Células Epiteliales/metabolismo , Feto/metabolismo , Ratones , Timocitos/metabolismo
4.
Proc Natl Acad Sci U S A ; 109(13): 5063-8, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22411787

RESUMEN

Recovery of neurological function after traumatic injury of the adult mammalian central nervous system is limited by lack of axonal growth. Myelin-derived inhibitors contribute to axonal growth restriction, with ephrinB3 being a developmentally important axonal guidance cue whose expression in mature oligodendrocytes suggests a role in regeneration. Here we explored the in vivo regeneration role of ephrinB3 using mice lacking a functional ephrinB3 gene. We confirm that ephrinB3 accounts for a substantial portion of detergent-resistant myelin-derived inhibition in vitro. To assess in vivo regeneration, we crushed the optic nerve and examined retinal ganglion fibers extending past the crush site. Significantly increased axonal regeneration is detected in ephrinB3(-/-) mice. Studies of spinal cord injury in ephrinB3(-/-) mice must take into account altered spinal cord development and an abnormal hopping gait before injury. In a near-total thoracic transection model, ephrinB3(-/-) mice show greater spasticity than wild-type mice for 2 mo, with slightly greater hindlimb function at later time points, but no evidence for axonal regeneration. After a dorsal hemisection injury, increased corticospinal and raphespinal growth in the caudal spinal cord are detected by 6 wk. This increased axonal growth is accompanied by improved locomotor performance measured in the open field and by kinematic analysis. Thus, ephrinB3 contributes to myelin-derived axonal growth inhibition and limits recovery from adult CNS trauma.


Asunto(s)
Envejecimiento/patología , Axones/patología , Efrina-B3/metabolismo , Vaina de Mielina/metabolismo , Regeneración Nerviosa/fisiología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Envejecimiento/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Detergentes/farmacología , Efrina-B3/deficiencia , Eliminación de Gen , Ratones , Actividad Motora/efectos de los fármacos , Vaina de Mielina/efectos de los fármacos , Compresión Nerviosa , Nervio Óptico/efectos de los fármacos , Nervio Óptico/patología , Nervio Óptico/fisiopatología , Tractos Piramidales/efectos de los fármacos , Tractos Piramidales/patología , Tractos Piramidales/fisiopatología , Núcleos del Rafe/efectos de los fármacos , Núcleos del Rafe/patología , Núcleos del Rafe/fisiopatología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/cirugía
5.
Acta Neuropathol ; 122(4): 429-42, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21779764

RESUMEN

Cerebral ischemia stimulates endogenous neurogenesis. However, the functional relevance of this phenomenon remains unclear because of poor survival and low neuronal differentiation rates of newborn cells. Therefore, further studies on mechanisms regulating neurogenesis under ischemic conditions are required, among which ephrin-ligands and ephrin-receptors (Eph) are an interesting target. Although Eph/ephrin proteins like ephrin-B3 are known to negatively regulate neurogenesis under physiological conditions, their role in cerebral ischemia is largely unknown. We therefore studied neurogenesis, brain injury and functional outcome in ephrin-B3(-/-) (knockout) and ephrin-B3(+/+) (wild-type) mice submitted to cerebral ischemia. Induction of stroke resulted in enhanced cell proliferation and neuronal differentiation around the lesion site of ephrin-B3(-/-) compared to ephrin-B3(+/+) mice. However, prominent post-ischemic neurogenesis in ephrin-B3(-/-) mice was accompanied by significantly increased ischemic injury and motor coordination deficits that persisted up to 4 weeks. Ischemic injury in ephrin-B3(-/-) mice was associated with a caspase-3-dependent activation of the signal transducer and activator of transcription 1 (STAT1). Whereas inhibition of caspase-3 had no effect on brain injury in ephrin-B3(+/+) animals, infarct size in ephrin-B3(-/-) mice was strongly reduced, suggesting that aggravated brain injury in these animals might involve a caspase-3-dependent activation of STAT1. In conclusion, post-ischemic neurogenesis in ephrin-B3(-/-) mice is strongly enhanced, but fails to contribute to functional recovery because of caspase-3-mediated aggravation of ischemic injury in these animals. Our results suggest that ephrin-B3 might be an interesting target for overcoming some of the limitations of further cell-based therapies in stroke.


Asunto(s)
Isquemia Encefálica/genética , Isquemia Encefálica/patología , Efrina-B3/deficiencia , Efrina-B3/genética , Neurogénesis/genética , Regulación hacia Arriba/genética , Animales , Isquemia Encefálica/metabolismo , Modelos Animales de Enfermedad , Efrina-B3/biosíntesis , Masculino , Ratones , Ratones Noqueados , Factores de Tiempo
6.
Nature ; 472(7343): 356-60, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21460838

RESUMEN

Coordinated migration of neurons in the developing and adult brain is essential for its proper function. The secreted glycoprotein Reelin (also known as RELN) guides migration of neurons by binding to two lipoprotein receptors, the very-low-density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2, also known as LRP8). Loss of Reelin function in humans results in the severe developmental disorder lissencephaly and it has also been associated with other neurological disorders such as epilepsy, schizophrenia and Alzheimer's disease. The molecular mechanisms by which Reelin activates its receptors and controls cellular functions are largely unknown. Here we show that the neuronal guidance cues ephrin B proteins are essential for Reelin signalling during the development of laminated structures in the brain. We show that ephrin Bs genetically interact with Reelin. Notably, compound mouse mutants (Reln(+/-); Efnb3(-/-) or Reln(+/-); Efnb2(-/-)) and triple ephrin B1, B2, B3 knockouts show neuronal migration defects that recapitulate the ones observed in the neocortex, hippocampus and cerebellum of the reeler mouse. Mechanistically, we show that Reelin binds to the extracellular domain of ephrin Bs, which associate at the membrane with VLDLR and ApoER2 in neurons. Clustering of ephrin Bs leads to the recruitment and phosphorylation of Dab1 which is necessary for Reelin signalling. Conversely, loss of function of ephrin Bs severely impairs Reelin-induced Dab1 phosphorylation. Importantly, activation of ephrin Bs can rescue the reeler neuronal migration defects in the absence of Reelin protein. Together, our results identify ephrin Bs as essential components of the Reelin receptor/signalling pathway to control neuronal migration during the development of the nervous system.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Corteza Cerebral/citología , Efrinas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Serina Endopeptidasas/metabolismo , Transducción de Señal , Animales , Moléculas de Adhesión Celular Neuronal/genética , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Efrina-B1/deficiencia , Efrina-B1/genética , Efrina-B1/metabolismo , Efrina-B2/deficiencia , Efrina-B2/genética , Efrina-B2/metabolismo , Efrina-B3/deficiencia , Efrina-B3/genética , Efrina-B3/metabolismo , Efrinas/deficiencia , Efrinas/genética , Proteínas de la Matriz Extracelular/genética , Femenino , Proteínas Relacionadas con Receptor de LDL/metabolismo , Ligandos , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Fenotipo , Fosforilación , Unión Proteica , Receptores de LDL/metabolismo , Proteína Reelina , Serina Endopeptidasas/genética
7.
J Neurosci ; 30(47): 16015-24, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21106840

RESUMEN

There is growing evidence that astrocytes play critical roles in neuron-glial interactions at the synapse. Astrocytes are believed to regulate presynaptic and postsynaptic structures and functions, in part, by the release of gliotransmitters such as glutamate, ATP, and d-serine; however, little is known of how neurons and astrocytes communicate to regulate these processes. Here, we investigated a family of transmembrane proteins called ephrinBs and Eph receptors that are expressed in the synapse and are known to regulate synaptic transmission and plasticity. In addition to their presence on CA1 hippocampal neurons, we determined that ephrins and Eph receptors are also expressed on hippocampal astrocytes. Stimulation of hippocampal astrocytes with soluble ephrinB3, known to be expressed on CA1 postsynaptic dendrites, enhanced d-serine synthesis and release in culture. Conversely, ephrinB3 had no effect on d-serine release from astrocytes deficient in EphB3 and EphA4, which are the primary receptors for ephrinB3. Eph receptors mediate this response through interactions with PICK1 (protein interacting with C-kinase) and by dephosphorylating protein kinase C α to activate the conversion of l-serine to d-serine by serine racemase. These findings are supported in vivo, where reduced d-serine levels and synaptic transmissions are observed in the absence of EphB3 and EphA4. These data support a role for ephrins and Eph receptors in regulating astrocyte gliotransmitters, which may have important implications on synaptic transmission and plasticity.


Asunto(s)
Astrocitos/metabolismo , Efrina-B3/fisiología , Serina/biosíntesis , Serina/metabolismo , Animales , Células Cultivadas , Efrina-B3/deficiencia , Hipocampo/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Plasticidad Neuronal/genética , Biosíntesis de Proteínas/genética , Receptor EphA4/biosíntesis , Receptor EphA4/deficiencia , Receptor EphA4/fisiología , Serina/análogos & derivados , Estereoisomerismo , Transmisión Sináptica/genética
8.
Stem Cells ; 28(7): 1231-42, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20496368

RESUMEN

Ephrins and Eph receptor(s) have recently been implicated in regulating neurogenesis in the adult subventricular zone (SVZ) and rostral migratory stream. Here, we examined the role of ephrinB3-EphB3 signaling in mediating the SVZ response to traumatic brain injury (TBI). Analysis of EphB3 expression showed colocalization with glial fibrillary acidic protein-positive neural stem progenitor cells (NSPCs) and doublecortin-positive neuroblasts, whereas ephrinB3 was expressed outside the neurogenic region. TBI resulted in a significant reduction in EphB3 expression, which coincided with enhanced NSPC survival and proliferation at 3 and 7 days postinjury. Analysis of mice lacking either ephrinB3 (ephrinB3(-/-)) or EphB3 (EphB3(-/-)) showed a significant increase in bromodeoxyuridine (BrdU) incorporation and Ki67 immunoreactivity in the SVZ. Interestingly, cell death was dissimilar between knockout mice, where cell death was reduced in EphB3(-/-) but increased in ephrinB3(-/-) mice. Lateral ventricle infusion of soluble preclustered ephrinB3-Fc reversed the proliferative and cell death defects in ephrinB3(-/-) but not EphB3(-/-) mice and prevented TBI-induced proliferation in wild-type NSPCs. Coincidently, tumor suppressor p53 expression was increased following EphB3 stimulation and is reduced in the absence of either EphB3 or ephrinB3. Furthermore, pharmacological inhibition and siRNA knockdown of p53-attenuated ephrinB3-Fc-mediated growth suppression while having no effect on cell death in cultured NSPCs. These data demonstrate that EphB3 signaling suppresses NSPC proliferation in a p53-dependent manner, induces cell death in the absence of ligand stimulation and is transiently reduced in the SVZ to initiate the expansion and survival of endogenous adult NSPCs following TBI.


Asunto(s)
Lesiones Encefálicas/metabolismo , Ventrículos Cerebrales/metabolismo , Homeostasis , Neuronas/metabolismo , Receptor EphB3/metabolismo , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Envejecimiento , Animales , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Proliferación Celular , Células Cultivadas , Ventrículos Cerebrales/citología , Efrina-B3/deficiencia , Efrina-B3/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/citología , ARN Interferente Pequeño/genética , Receptor EphB3/deficiencia , Células Madre/citología , Proteína p53 Supresora de Tumor/genética
9.
Proc Natl Acad Sci U S A ; 107(19): 8830-5, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20410461

RESUMEN

Nervous system function requires tight control over the number of synapses individual neurons receive, but the underlying cellular and molecular mechanisms that regulate synapse number remain obscure. Here we present evidence that a trans-synaptic interaction between EphB2 in the presynaptic compartment and ephrin-B3 in the postsynaptic compartment regulates synapse density and the formation of dendritic spines. Observations in cultured cortical neurons demonstrate that synapse density scales with ephrin-B3 expression level and is controlled by ephrin-B3-dependent competitive cell-cell interactions. RNA interference and biochemical experiments support the model that ephrin-B3 regulates synapse density by directly binding to Erk1/2 to inhibit postsynaptic Ras/mitogen-activated protein kinase signaling. Together these findings define a mechanism that contributes to synapse maturation and controls the number of excitatory synaptic inputs received by individual neurons.


Asunto(s)
Efrina-B2/metabolismo , Efrina-B3/metabolismo , Potenciales Postsinápticos Excitadores , Sistema de Señalización de MAP Quinasas , Sinapsis/enzimología , Animales , Comunicación Celular , Línea Celular , Espinas Dendríticas/metabolismo , Efrina-B3/deficiencia , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ligandos , Ratones , Terminales Presinápticos/metabolismo , Unión Proteica , Ratas
10.
Mech Dev ; 126(3-4): 230-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19032981

RESUMEN

Ephs and ephrins are cell surface receptors that bind to each other and initiate distinct, bidirectional signalling pathways in processes known as forward (Eph) and reverse (ephrin) signalling. Previous work had shown that the loss of ephrinB1 protein alone or compound loss of EphB2 and EphB3 leads to cleft palate. Because of the bidirectional signalling capability of these molecules, it was not clear whether forward or reverse signalling caused the cleft palate in the ephrinB1 protein null or EphB2 and EphB3 compound null mice. We demonstrate that forward signalling is essential for palatogenesis. Foetuses with a cytoplasmically truncated EphB2 protein, which could initiate reverse but not forward signalling, and were protein null for EphB3 had a cleft palate. This happened because their palatal shelves, which could elevate in vivo and adhere and fuse in culture, were too small to contact one another. Small shelf size was due to reduced proliferation in the palatal mesenchyme. The reduced proliferation was not the result of abnormal vascular development within the palate. In conclusion, strong evidence is provided for specific and co-operative roles of EphB2 and EphB3 in palate development.


Asunto(s)
Efrina-B3/metabolismo , Hueso Paladar/embriología , Hueso Paladar/metabolismo , Receptor EphB2/metabolismo , Transducción de Señal , Adhesividad , Animales , Apoptosis , Recuento de Células , Proliferación Celular , Fisura del Paladar/embriología , Fisura del Paladar/patología , Efrina-B3/deficiencia , Matriz Extracelular/metabolismo , Femenino , Feto/anomalías , Feto/metabolismo , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Hueso Paladar/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor EphB2/deficiencia , beta-Galactosidasa/metabolismo
11.
Dev Biol ; 292(1): 34-45, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16466709

RESUMEN

The transmembrane ephrinB ligands and their Eph receptor tyrosine kinases are known to regulate excitatory synaptic functions in the hippocampus. In the CA3-CA1 synapse, ephrinB ligands are localized to the post-synaptic membrane, while their cognate Eph receptors are presumed to be pre-synaptic. Interaction of ephrinB molecules with Eph receptors leads to changes in long-term potentiation (LTP), which has been reported to be mediated by reverse signaling into the post-synaptic membrane. Here, we demonstrate that the cytoplasmic domain of ephrinB3 and hence reverse signaling is not required for ephrinB dependent learning and memory tasks or for LTP of these synapses. Consistent with previous reports, we find that ephrinB3(KO) null mutant mice exhibit a striking reduction in CA3-CA1 LTP that is associated with defective learning and memory tasks. We find the null mutants also show changes in both pre- and post-synaptic proteins including increased levels of synapsin and synaptobrevin and reduced levels of NMDA receptor subunits. These abnormalities are not observed in ephrinB3(lacZ) reverse signaling mutants that specifically delete the ephrinB3 intracellular region, supporting a cytoplasmic domain-independent forward signaling role for ephrinB3 in these processes. We also find that both ephrinB3(KO) and ephrinB3(lacZ) mice show an increased number of excitatory synapses, demonstrating a cytoplasmic-dependent reverse signaling role of ephrinB3 in regulating synapse number. Together, these data suggest that ephrinB3 may act like a receptor to transduce reverse signals to regulate the number of synapses formed in the hippocampus, and that it likely acts to stimulate forward signaling to modulate a number of other proteins involved in synaptic activity and learning/memory.


Asunto(s)
Efrina-B3/fisiología , Hipocampo/embriología , Hipocampo/fisiología , Transducción de Señal/fisiología , Sinapsis/fisiología , Animales , Axones/fisiología , Células Cultivadas , Citoplasma/genética , Dendritas/fisiología , Giro Dentado/fisiología , Efrina-B3/biosíntesis , Efrina-B3/deficiencia , Efrina-B3/genética , Hipocampo/citología , Potenciación a Largo Plazo/genética , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos , Ratones Noqueados , Microscopía de Interferencia , Fosforilación , Estructura Terciaria de Proteína/genética , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/fisiología , Transducción de Señal/genética
12.
J Neurosci ; 24(10): 2366-74, 2004 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-15014111

RESUMEN

Axons travel frequently in bundles to reach their target. After arriving at the target, axon terminals defasciculate, migrate to topographically defined positions, and form synapses with appropriate target neurons. Here we present evidence that the B-type receptors of the erythropoietin-producing hepatocellular (Eph) family and a ligand, ephrin-B3, influence hippocampal axon defasciculation. The EphB receptors are expressed in the hippocampus, and the ligand, ephrin-B3, is transcribed in the lateral septum, the major subcortical target of hippocampal neurons. Ephrin-B3 promotes adhesion of hippocampal neurons to the ligand-expressing substrates in vitro, and the loss of the receptor EphB2 abrogates the effects of ephrin-B3. In mice deficient in EphB2 and EphB3, many hippocampal axons remain in bundles. This phenotype was also observed in mice that were specifically deleted for the cytoplasmic domain of EphB2. These observations indicate that the EphB receptors and their ligand regulate hippocampal axon defasciculation at the septal target, possibly through a receptor-mediated forward signaling mechanism.


Asunto(s)
Axones/patología , Hipocampo/patología , Malformaciones del Sistema Nervioso/patología , Receptores de la Familia Eph/genética , Animales , Adhesión Celular/fisiología , Efrina-B1/genética , Efrina-B2/deficiencia , Efrina-B2/genética , Efrina-B2/metabolismo , Efrina-B3/deficiencia , Efrina-B3/genética , Efrina-B3/metabolismo , Colorantes Fluorescentes , Hipocampo/anomalías , Hipocampo/metabolismo , Ligandos , Ratones , Ratones Mutantes Neurológicos , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo , Fenotipo , ARN Mensajero/biosíntesis , Receptores de la Familia Eph/biosíntesis , Tabique del Cerebro/anomalías , Tabique del Cerebro/metabolismo , Tabique del Cerebro/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...