Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Neurosci ; 34(17): 6084-97, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24760868

RESUMEN

Tau is a microtubule-associated protein well known for its stabilization of microtubules in axons. Recently, it has emerged that tau participates in synaptic function as part of the molecular pathway leading to amyloid-beta (Aß)-driven synaptotoxicity in the context of Alzheimer's disease. Here, we report the implication of tau in the profound functional synaptic modification associated with synaptic plasticity. By exposing murine cultured cortical neurons to a pharmacological synaptic activation, we induced translocation of endogenous tau from the dendritic to the postsynaptic compartment. We observed similar tau translocation to the postsynaptic fraction in acute hippocampal slices subjected to long-term potentiation. When we performed live confocal microscopy on cortical neurons transfected with human-tau-GFP, we visualized an activity-dependent accumulation of tau in the postsynaptic density. Coprecipitation using phalloidin revealed that tau interacts with the most predominant cytoskeletal component present, filamentous actin. Finally, when we exposed cortical cultures to 100 nm human synthetic Aß oligomers (Aßo's) for 15 min, we induced mislocalization of tau into the spines under resting conditions and abrogated subsequent activity-dependent synaptic tau translocation. These changes in synaptic tau dynamics may rely on a difference between physiological and pathological phosphorylation of tau. Together, these results suggest that intense synaptic activity drives tau to the postsynaptic density of excitatory synapses and that Aßo-driven tau translocation to the spine deserves further investigation as a key event toward synaptotoxicity in neurodegenerative diseases.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Corteza Cerebral/efectos de los fármacos , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Sinapsis/efectos de los fármacos , Proteínas tau/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/metabolismo , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Ratones , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Sinapsis/metabolismo
2.
Front Neuroendocrinol ; 31(3): 241-58, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20546773

RESUMEN

As the final common pathway for the central control of gonadotropin secretion, GnRH neurons are subjected to numerous regulatory homeostatic and external factors to achieve levels of fertility appropriate to the organism. The GnRH system thus provides an excellent model in which to investigate the complex relationships between neurosecretion, morphological plasticity and the expression of a physiological function. Throughout the reproductive cycle beginning from postnatal sexual development and the onset of puberty to reproductive senescence, and even within the ovarian cycle itself, all levels of the GnRH system undergo morphological plasticity. This structural plasticity within the GnRH system appears crucial to the timely control of reproductive competence within the individual, and as such must have coordinated actions of multiple signals secreted from glial cells, endothelial cells, and GnRH neurons. Thus, the GnRH system must be viewed as a complete neuro-glial-vascular unit that works in concert to maintain the reproductive axis.


Asunto(s)
Comunicación Celular/fisiología , Células Endoteliales/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Neuroglía/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Animales , Células Endoteliales/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/fisiología , Humanos , Modelos Biológicos , Neuroglía/metabolismo , Neuronas/metabolismo , Ovario/metabolismo , Ovario/fisiología , Pubertad/metabolismo , Pubertad/fisiología , Receptores LHRH/metabolismo , Receptores LHRH/fisiología
3.
Neuroendocrinology ; 93(2): 74-89, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21335953

RESUMEN

Nitric oxide (NO) is a peculiar chemical transmitter that freely diffuses through aqueous and lipid environments and plays a role in major aspects of brain function. Within the hypothalamus, NO exerts critical effects upon the gonadotropin-releasing hormone (GnRH) network to maintain fertility. Here, we review recent evidence that NO regulates major aspects of the GnRH neuron physiology. Far more active than once thought, NO powerfully controls GnRH neuronal activity, GnRH release and structural plasticity at the neurohemal junction. In the preoptic region, neuronal nitric oxide synthase (nNOS) activity is tightly regulated by estrogens and is found to be maximal at the proestrus stage. Natural fluctuations of estrogens control both the differential coupling of this Ca²+-activated enzyme to glutamate N-methyl-D-aspartic acid receptor channels and phosphorylation-mediated nNOS activation. Furthermore, NO endogenously produced by neurons expressing nNOS acutely and directly suppresses spontaneous firing in GnRH neurons, which suggests that neuronal NO may serve as a synchronizing switch within the preoptic region. At the median eminence, NO is spontaneously released from an endothelial source and follows a pulsatile and cyclic pattern of secretion. Importantly, GnRH release appears to be causally related to endothelial NO release. NO is also highly involved in mediating the dialogue set in motion between vascular endothelial cells and tanycytes that control the direct access of GnRH neurons to the pituitary portal blood during the estrous cycle. Altogether, these data raise the intriguing possibility that the neuroendocrine brain uses NO to coordinate both GnRH neuronal activity and GnRH release at key stages of reproductive physiology.


Asunto(s)
Encéfalo/fisiología , Células Endoteliales/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Sistemas Neurosecretores/fisiología , Óxido Nítrico/fisiología , Reproducción/fisiología , Transducción de Señal/fisiología , Animales , Hormona Liberadora de Gonadotropina/fisiología , Modelos Biológicos , Óxido Nítrico/biosíntesis
4.
J Neurosci ; 24(46): 10353-63, 2004 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-15548649

RESUMEN

Glial and endothelial cells interact throughout the brain to define specific functional domains. Whether endothelial cells convey signals to glia in the mature brain is unknown but is amenable to examination in circumventricular organs. Here we report that purified endothelial cells of one of these organs, the median eminence of the hypothalamus, induce acute actin cytoskeleton remodeling in isolated ependymoglial cells and show that this plasticity is mediated by nitric oxide (NO), a diffusible factor. We found that both soluble guanylyl cyclase and cyclooxygenase products are involved in this endothelial-mediated control of ependymoglia cytoarchitecture. We also demonstrate by electron microscopy that activation of endogenous NO release in the median eminence induces rapid structural changes, allowing a direct access of neurosecretory axons containing gonadotropin-releasing hormone (GnRH) (the neuropeptide controlling reproductive function) to the portal vasculature. Local in vivo inhibition of NO synthesis disrupts reproductive cyclicity, a process that requires a pulsatile, coordinated delivery of GnRH into the hypothalamic-adenohypophyseal portal system. Our results identify a previously unknown function for endothelial cells in inducing neuroglial plasticity and raise the intriguing possibility that endothelial cells throughout the brain may use a similar signaling mechanism to regulate glial-neuronal interactions.


Asunto(s)
Astrocitos/fisiología , Encéfalo/citología , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Epéndimo/fisiología , Plasticidad Neuronal/fisiología , Sistemas Neurosecretores/citología , Actinas/ultraestructura , Animales , Astrocitos/ultraestructura , Encéfalo/ultraestructura , Células Cultivadas , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Citoesqueleto/ultraestructura , Endotelio Vascular/citología , Endotelio Vascular/ultraestructura , Epéndimo/ultraestructura , Ciclo Estral/fisiología , Femenino , Hormona Liberadora de Gonadotropina/fisiología , Guanilato Ciclasa/metabolismo , Eminencia Media/irrigación sanguínea , Eminencia Media/fisiología , Eminencia Media/ultraestructura , Proteínas de la Membrana , Sistemas Neurosecretores/ultraestructura , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/fisiología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal
5.
Endocrinology ; 145(4): 1794-801, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14670985

RESUMEN

In vitro studies using immortalized GT1 cells suggest that hypothalamic astrocytes employ TGFbeta(1) to directly regulate the secretion of GnRH, the neurohormone that controls sexual maturation and adult reproductive function. However, whether such astrocyte-GnRH neuron signaling occurs in vivo is not clear. In the present study, we used in situ hybridization and immunohistochemistry to determine whether astrocytes and GnRH neurons express the molecular components necessary to set in motion communication processes involving TGFbeta(1) signaling. Double-labeling experiments showed that astrocytes in the male rat preoptic region (POA) expressed TGFbeta(1) mRNA and that GnRH perikarya were often found in close association with TGFbeta(1) mRNA-expressing cells. In addition, GnRH neuronal cell bodies in the POA expressed both type II TGFbeta receptors (TGFbeta-RII), which selectively bind TGFbeta, and Smad2/3, one of the primary transducers of TGFbeta signaling, suggesting that they are fully capable of responding directly to TGFbeta(1) stimulation. Consistent with this hypothesis, incubation of POA explants with TGFbeta(1) caused a significant, dose-dependent decrease in GnRH mRNA expression in individual neurons. This effect was observed within 1 h after TGFbeta(1)-treatment and was inhibited by addition of the soluble form of TGFbeta-RII to the incubation medium. In contrast, whereas both TGFbeta(1) and TGFbeta-RII mRNAs were abundantly expressed in both glial cells and capillaries in the median eminence, the projection field of GnRH neurons, TGFbeta-RII immunoreactivity was mainly restricted to the processes of tanycytes and did not colocalize with GnRH-immunoreactive fibers. This observation supports previous in vivo studies showing that TGFbeta(1) is unable to directly modulate decapeptide release from GnRH nerve terminals. Thus, astrocyte-derived TGFbeta(1) may directly influence GnRH expression and/or secretion in vivo by acting on the perikarya, but not the terminals, of GnRH neurons.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Eminencia Media/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Astrocitos/metabolismo , Expresión Génica , Hormona Liberadora de Gonadotropina/genética , Hipotálamo/citología , Hipotálamo Medio/metabolismo , Masculino , Neuronas/metabolismo , Fenotipo , Área Preóptica/metabolismo , Proteínas Serina-Treonina Quinasas , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1
6.
Endocrinology ; 151(4): 1760-72, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20133455

RESUMEN

In the ever-changing physiological context of the neuroendocrine brain, the mechanisms by which cellular events involving neurons, astroglia, and vascular cells are coordinated to bring forth the appropriate neuronal signaling is not yet known but is amenable to examination. In the median eminence of the hypothalamus, endothelial cells are key players in the plasticity of tanycytes (specialized astroglia) and neuroendocrine synapse efficacy. Here we report that estradiol acts on both purified endothelial cells and isolated tanycytes to trigger endothelial-to-glial communication that leads to a sudden and massive retraction of tanycyte processes. The blockade of endothelial nitric oxide synthase by in vitro adenoviral-mediated gene transfer of a dominant-negative form of endothelial nitric oxide synthase abrogates the estradiol-induced tanycyte plasticity mediated by endothelial cells. In parallel, increases in prostaglandin-E(2) (PGE(2)) due to changes in cyclooxygenase (COX)-1 and COX-2 expression induced by the exposure of tanycytes to estradiol promote acute tanycyte plasticity. We also demonstrate by electron microscopy that the administration of PGE(2) to median eminence explants induces rapid neuroglial plasticity at the neurovascular junction of neurons that release GnRH (the neuropeptide controlling reproduction). Conversely, preventing local PGE(2) synthesis in the median eminence of adult female rats with the COX inhibitor indomethacin impairs the ovarian cycle, a process that requires a pulsatile, coordinated delivery of GnRH into the hypothalamo-hypophyseal portal system. Taken together, our findings show that estradiol controls the dialog between endothelial cells and astroglia to regulate neuroglial plasticity in the neuroendocrine brain.


Asunto(s)
Forma de la Célula/fisiología , Células Endoteliales/fisiología , Epéndimo/fisiología , Estradiol/fisiología , Eminencia Media/fisiología , Neuroglía/fisiología , Análisis de Varianza , Animales , Western Blotting , Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , Técnicas de Cultivo de Célula , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Dinoprostona/farmacología , Células Endoteliales/efectos de los fármacos , Epéndimo/efectos de los fármacos , Estradiol/farmacología , Sistema Hipotálamo-Hipofisario/fisiología , Neuroglía/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/fisiología , Prostaglandina-Endoperóxido Sintasas/fisiología , Ratas , Ratas Sprague-Dawley
7.
J Biol Chem ; 281(46): 35030-8, 2006 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-16984913

RESUMEN

The Annexin2 tetramer (A2t), which consists of two Annexin2 molecules bound to a S100A10 dimer, is implicated in membrane-trafficking events. Here, we showed using a yeast triple-hybrid experiment and in vitro binding assay that Annexin2 is required for strong binding of S100A10 to the C-terminal domain of the protein Ahnak. We also revealed that this effect involves only the Annexin2 N-terminal tail, which is implicated in S100A10/Annexin2 tetramerization. The minimal A2t binding motif (A2tBP1) in Ahnak was mapped to a 20-amino acid peptide, and this peptide is highly specific for A2t. We also identified a second A2t binding motif (A2tBP2) present in the N-terminal domain of Ahnak, which binds to A2t, albeit with less affinity. When overexpressed as an EGFP fusion protein in MDCK cells, A2tBPs cofractionate in a calcium-dependent manner and co-immunoprecipitate with S100A10 and Annexin2. In living cells, A2tBPs target EGFP to the cytoplasm as does Annexin2. In response to oxidative and mechanical stress, EGFP-A2tBPs relocalize within minutes to the plasma membrane; a behavior shared with Annexin2-GFP. These results suggest that the A2t complex exists within the cytoplasm of resting living cells and that its localization at the plasma membrane relies on cellular signaling. Together, our data demonstrate that A2tBP1 is a specific A2t complex binding domain and may be a powerful tool to help elucidate A2t structure and cellular functions.


Asunto(s)
Anexina A2/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas S100/metabolismo , Secuencias de Aminoácidos , Animales , Anexina A2/química , Línea Celular , Perros , Células Epiteliales , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Unión Proteica , Transporte de Proteínas , Proteínas S100/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA