Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 109(41): 16624-9, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23012472

RESUMEN

Nitric oxide (NO) generated by neuronal NO synthase (nNOS) initiates penile erection, but has not been thought to participate in the sustained erection required for normal sexual performance. We now show that cAMP-dependent phosphorylation of nNOS mediates erectile physiology, including sustained erection. nNOS is phosphorylated by cAMP-dependent protein kinase (PKA) at serine(S)1412. Electrical stimulation of the penile innervation increases S1412 phosphorylation that is blocked by PKA inhibitors but not by PI3-kinase/Akt inhibitors. Stimulation of cAMP formation by forskolin also activates nNOS phosphorylation. Sustained penile erection elicited by either intracavernous forskolin injection, or augmented by forskolin during cavernous nerve electrical stimulation, is prevented by the NOS inhibitor L-NAME or in nNOS-deleted mice. Thus, nNOS mediates both initiation and maintenance of penile erection, implying unique approaches for treating erectile dysfunction.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Erección Peniana/fisiología , Animales , Western Blotting , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/genética , Erección Peniana/efectos de los fármacos , Pene/inervación , Pene/metabolismo , Pene/fisiología , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Serina/metabolismo
2.
J Neurosci ; 31(6): 1991-9, 2011 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21307237

RESUMEN

Glucose transporter 3 (GLUT3) is the main facilitative glucose transporter in neurons. Glucose provides neurons with a critical energy source for neuronal activity. However, the mechanism by which neuronal activity controls glucose influx via GLUT3 is unknown. We investigated the influence of synaptic stimulation on GLUT3 surface expression and glucose import in primary cultured cortical and hippocampal neurons. Synaptic activity increased surface expression of GLUT3 leading to an elevation of intracellular glucose. The effect was blocked by NMDA receptor (NMDAR) and neuronal nitric oxide synthase (nNOS) inhibition. The Akt inhibitor I (Akt-I) blocked NMDAR-induced GLUT3 surface expression while a nNOS-phosphomimetic mutant (S1412D) enhanced GLUT3 expression at cell surface. These results suggest that NMDAR/Akt-dependent nNOS phosphorylation is coupled to GLUT3 trafficking. We demonstrated that activation of cGMP-dependent protein kinase (cGK) increased the surface expression of GLUT3, which was repressed by Rp-8-pCPT-cGMPS, a potent cell-permeable inhibitor of cGKs. These studies characterize the molecular basis for activity-dependent increases in surface GLUT3 after stimulation of the NMDARs. NMDAR-induced increase in surface GLUT3 represents a novel pathway for control of energy supply during neuronal activity that is critical for maintaining glucose homeostasis during neuronal transmission.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Transportador de Glucosa de Tipo 3/metabolismo , Neuronas/metabolismo , 4-Aminopiridina/farmacología , Análisis de Varianza , Animales , Bicuculina/farmacología , Biotinilación/métodos , Corteza Cerebral/citología , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Homólogo 4 de la Proteína Discs Large , Perros , Interacciones Farmacológicas , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Receptores de GABA-A/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Transportador de Glucosa de Tipo 3/genética , Proteínas Fluorescentes Verdes/genética , Hexosas/metabolismo , Hipocampo/citología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Bloqueadores de los Canales de Potasio/farmacología , Transporte de Proteínas/genética , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinaptosomas/metabolismo , Tionucleótidos/farmacología , Factores de Tiempo , Transfección , Valina/análogos & derivados , Valina/farmacología
3.
Mol Cell Neurosci ; 43(4): 341-52, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20083202

RESUMEN

AMPA receptor binding protein (ABP) is a multi-PDZ domain scaffold that binds and stabilizes AMPA receptor (AMPAR) GluR2/3 subunits at synapses. A palmitoylated N-terminal splice variant (pABP-L) concentrates in spine heads, whereas a non-palmitoylated form (ABP-L) is intracellular. We show that postsynaptic Sindbis viral expression of pABP-L increased AMPAR mediated mEPSC amplitude and frequency and elevated surface levels of GluR1 and GluR2, suggesting an increase in AMPA receptors at individual synapses. Spines were enlarged and more numerous and nerve terminals contacting these cells displayed enlarged synaptophysin puncta. A non-palmitoylated pABP-L mutant (C11A) did not change spine density or size. Exogenous pABP-L and endogenous GRIP, a related scaffold, colocalized with NPRAP (delta-catenin), to which ABP and GRIP bind, and with cadherins, which bind NPRAP. Thus postsynaptic pABP-L induces pre and postsynaptic changes that are dependent on palmitoylation and likely achieved through ABP association with a multi-molecular cell surface signaling complex.


Asunto(s)
Proteínas Portadoras/metabolismo , Lipoilación/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Animales , Cadherinas/metabolismo , Forma de la Célula , Células Cultivadas , Espinas Dendríticas/metabolismo , Electrofisiología , Potenciales Postsinápticos Excitadores/fisiología , Hipocampo/citología , Hipocampo/metabolismo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular , Potenciales Postsinápticos Miniatura/fisiología , Neuronas/citología , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Sinaptofisina/metabolismo
4.
J Neurosci ; 27(13): 3445-55, 2007 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-17392461

RESUMEN

Postsynaptic nitric oxide (NO) production affects synaptic plasticity and neuronal cell death. Ca2+ fluxes through the NMDA receptor (NMDAR) stimulate the production of NO by neuronal nitric oxide synthase (nNOS). However, the mechanisms by which nNOS activity is regulated are poorly understood. We evaluated the effect of neuronal stimulation with glutamate on the phosphorylation of nNOS. We show that, in cortical neurons, a low glutamate concentration (30 microM) induces rapid and transient NMDAR-dependent phosphorylation of S1412 by Akt, followed by sustained phosphorylation of S847 by CaMKII (calcium-calmodulin-dependent kinase II). We demonstrate that phosphorylation of S1412 by Akt is necessary for activation of nNOS by the NMDAR. nNOS mutagenesis confirms that these phosphorylations respectively activate and inhibit nNOS and, thus, transiently activate NO production. A constitutively active (S1412D), but not a constitutively repressed (S847D) nNOS mutant elevated surface glutamate receptor 2 levels, demonstrating that these phosphorylations can control AMPA receptor trafficking via NO. Notably, an excitotoxic stimulus (150 microM glutamate) induced S1412, but not S847 phosphorylation, leading to deregulated nNOS activation. S1412D did not kill neurons; however, it enhanced the excitotoxicity of a concomitant glutamate stimulus. We propose a swinging domain model for the regulation of nNOS: S1412 phosphorylation facilitates electron flow within the reductase module of nNOS, increasing nNOS sensitivity to Ca2+-calmodulin. These findings suggest a critical role for a kinetically complex and novel series of regulatory nNOS phosphorylations induced by the NMDA receptor for the in vivo control of nNOS.


Asunto(s)
Muerte Celular/fisiología , Neuronas/enzimología , Óxido Nítrico Sintasa/metabolismo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Ácido Glutámico/metabolismo , Hipocampo/embriología , Hipocampo/metabolismo , Fosforilación , Transporte de Proteínas/fisiología , Ratas
5.
Neurobiol Aging ; 24(8): 1123-33, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14643384

RESUMEN

Stimulation of NMDA receptors activates neuronal nitric oxide synthase (nNOS) and the production of nitric oxide (NO). Dephosphorylation of nNOS increases nNOS enzymatic activity. We have examined the regulation of nNOS phosphorylation in rat cortical neurons following NMDA receptor activation. We show that nNOS is constitutively phosphorylated and that NMDA receptor activation decreases the level of nNOS phosphorylation by a mechanism that is blocked specifically by NMDA receptor antagonists and inhibitors of the Ca2+-regulated phosphatases calcineurin and PP1/PP2A. Using quantitative digital microscopy, we show that NMDA receptor activation induces the accumulation of nitrotyrosine, a measure of nNOS activity, and TdT-mediated fluorescein-dUTP nick end labeling (TUNEL) positivity, a measure of cell death. A calcineurin inhibitor blocked the increase in both TUNEL and nitrotyrosine positivity. Notably, TUNEL was increased in those neurons that were most strongly positive for nitrotyrosine. We conclude that NMDA receptor activation induces death of neurons by a cell autonomous pathway involving nNOS dephosphorylation by a calcineurin-dependent mechanism.


Asunto(s)
Apoptosis/fisiología , Degeneración Nerviosa/enzimología , Neuronas/enzimología , Óxido Nítrico Sintasa/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Tirosina/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Calcineurina/metabolismo , Inhibidores de la Calcineurina , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Etiquetado Corte-Fin in Situ , Degeneración Nerviosa/fisiopatología , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Tirosina/metabolismo
6.
J Biol Chem ; 279(14): 14307-14, 2004 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-14722119

RESUMEN

At glutamatergic synapses, the scaffolding protein PSD95 links the neuronal isoform of nitric-oxide synthase (nNOS) to the N-methyl-d-aspartate (NMDA) receptor. Phosphorylation of nNOS at serine 847 (Ser(847)) by the calcium-calmodulin protein kinase II (CaMKII) inhibits nNOS activity, possibly by blocking the binding of Ca(2+)-CaM. Here we show that the NMDA mediates a novel bidirectional regulation of Ser(847) phosphorylation. nNOS phosphorylated at Ser(847) colocalizes with the NMDA receptor at spines of cultured hippocampal neurons. Treatment of neurons with 5 microm glutamate stimulated CaMKII phosphorylation of nNOS at Ser(847), whereas excitotoxic concentrations of glutamate, 100 and 500 microm, induced Ser(847)-PO(4) dephosphorylation by protein phosphatase 1. Strong NMDA receptor stimulation was likely to activate nNOS under these conditions because protein nitration to form nitrotyrosine, a marker of nNOS activity, correlated in individual neurons with Ser(847)-PO(4) dephosphorylation. Of particular note, stimulation with low glutamate that increased phosphorylation of nNOS at Ser(847) could be reversed by subsequent high glutamate treatment which induced dephosphorylation. The reversibility of NMDA receptor-induced phosphorylation at Ser(847) by different doses of glutamate suggests two mechanisms with opposite effects: 1). a time-dependent negative feedback induced by physiological concentrations of glutamate that limits nNOS activation and precludes the overproduction of NO; and 2). a pathological stimulation by high concentrations of glutamate that leads to unregulated nNOS activation and production of toxic levels of NO. These mechanisms may share pathways, respectively, with NMDA receptor-induced forms of synaptic plasticity and excitotoxicity.


Asunto(s)
Neuronas/enzimología , Óxido Nítrico Sintasa/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Ácido Glutámico/metabolismo , Hipocampo/citología , Plasticidad Neuronal/fisiología , Neuronas/citología , Óxido Nítrico Sintasa de Tipo I , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Proteína Fosfatasa 1 , Ratas , Serina/metabolismo , Sinapsis/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA