Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pineal Res ; 58(3): 321-34, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25707622

RESUMEN

Although melatonin receptors are widely expressed in the mammalian central nervous system and peripheral tissues, there are limited data regarding the functions of melatonin in cerebellar Purkinje cells. Here, we identified a novel functional role of melatonin in modulating P-type Ca(2+) channels and action-potential firing in rat Purkinje neurons. Melatonin at 0.1 µm reversibly decreased peak currents (I(Ba)) by 32.9%. This effect was melatonin receptor 1 (MT(R1)) dependent and was associated with a hyperpolarizing shift in the voltage dependence of inactivation. Pertussis toxin pretreatment, intracellular application of QEHA peptide, and a selective antibody raised against the Gß subunit prevented the inhibitory effects of melatonin. Pretreatment with phosphatidylinositol 3-kinase (PI3K) inhibitors abolished the melatonin-induced decrease in I(Ba). Surprisingly, melatonin responses were not regulated by Akt, a common downstream target of PI3K. Melatonin treatment significantly increased protein kinase C (PKC) activity 2.1-fold. Antagonists of PKC, but not of protein kinase A, abolished the melatonin-induced decrease in I(Ba). Melatonin application increased the membrane abundance of PKCδ, and PKCδ inhibition (either pharmacologically or genetically) abolished the melatonin-induced IBa response. Functionally, melatonin increased spontaneous action-potential firing by 53.0%; knockdown of MT(R1) and blockade of P-type channels abolished this effect. Thus, our results suggest that melatonin inhibits P-type channels through MT(R1) activation, which is coupled sequentially to the ßγ subunits of G(i/o)-protein and to downstream PI3K-dependent PKCδ signaling. This likely contributes to its physiological functions, including spontaneous firing of cerebellar Purkinje neurons.


Asunto(s)
Canales de Calcio Tipo P/efectos de los fármacos , Melatonina/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteína Quinasa C-delta/metabolismo , Células de Purkinje/efectos de los fármacos , Animales , Canales de Calcio Tipo P/metabolismo , Células de Purkinje/metabolismo , Ratas , Ratas Wistar
2.
J Neurochem ; 129(4): 628-36, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24521102

RESUMEN

Urotensin II (U-II) is a cyclic undecapeptide that regulates cardiovascular function at central and peripheral sites. The functional role of U-II nucleus ambiguus, a key site controlling cardiac tone, has not been established, despite the identification of U-II and its receptor at this level. We report here that U-II produces an increase in cytosolic Ca(2+) concentration in retrogradely labeled cardiac vagal neurons of nucleus ambiguus via two pathways: (i) Ca(2+) release from the endoplasmic reticulum via inositol 1,4,5-trisphosphate receptor; and (ii) Ca(2+) influx through P/Q-type Ca(2+) channels. In addition, U-II depolarizes cultured cardiac parasympathetic neurons. Microinjection of increasing concentrations of U-II into nucleus ambiguus elicits dose-dependent bradycardia in conscious rats, indicating the in vivo activation of the cholinergic pathway controlling the heart rate. Both the in vitro and in vivo effects were abolished by the urotensin receptor antagonist, urantide. Our findings suggest that, in addition, to the previously reported increase in sympathetic outflow, U-II activates cardiac vagal neurons of nucleus ambiguus, which may contribute to cardioprotection.


Asunto(s)
Bradicardia/fisiopatología , Tronco Encefálico/fisiopatología , Señalización del Calcio/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiopatología , Neuronas/metabolismo , Sistema Nervioso Parasimpático/fisiopatología , Urotensinas/fisiología , Nervio Vago/fisiopatología , Animales , Animales Recién Nacidos , Fibras Autónomas Preganglionares/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Bradicardia/inducido químicamente , Tronco Encefálico/efectos de los fármacos , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/fisiología , Canales de Calcio Tipo Q/efectos de los fármacos , Canales de Calcio Tipo Q/fisiología , Señalización del Calcio/fisiología , Femenino , Sistema de Conducción Cardíaco/efectos de los fármacos , Receptores de Inositol 1,4,5-Trifosfato/efectos de los fármacos , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Microinyecciones , Modelos Cardiovasculares , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/fisiología , Taquicardia/inducido químicamente , Taquifilaxis , Urotensinas/farmacología , Urotensinas/toxicidad
3.
J Neurochem ; 126(6): 739-48, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23795642

RESUMEN

Nesfatin-1, a peptide whose receptor is yet to be identified, has been involved in the modulation of feeding, stress, and metabolic responses. More recently, increasing evidence supports a modulatory role for nesfatin-1 in autonomic and cardiovascular activity. This study was undertaken to test if the expression of nesfatin-1 in the nucleus ambiguus, a key site for parasympathetic cardiac control, may be correlated with a functional role. As we have previously demonstrated that nesfatin-1 elicits Ca²âº signaling in hypothalamic neurons, we first assessed the effect of this peptide on cytosolic Ca²âº in cardiac pre-ganglionic neurons of nucleus ambiguus. We provide evidence that nesfatin-1 increases cytosolic Ca²âº concentration via a Gi/o-coupled mechanism. The nesfatin-1-induced Ca²âº rise is critically dependent on Ca²âº influx via P/Q-type voltage-activated Ca²âº channels. Repeated administration of nesfatin-1 leads to tachyphylaxis. Furthermore, nesfatin-1 produces a dose-dependent depolarization of cardiac vagal neurons via a Gi/o-coupled mechanism. In vivo studies, using telemetric and tail-cuff monitoring of heart rate and blood pressure, indicate that microinjection of nesfatin-1 into the nucleus ambiguus produces bradycardia not accompanied by a change in blood pressure in conscious rats. Taken together, our results identify for the first time that nesfatin-1 decreases heart rate by activating cardiac vagal neurons of nucleus ambiguus. Our results indicate that nesfatin-1, one of the most potent feeding peptides, increases cytosolic Ca²âº by promoting Ca²âº influx via P/Q channels and depolarizes nucleus ambiguus neurons; both effects are Gi/o-mediated. In vivo studies indicate that microinjection of nesfatin-1 into nucleus ambiguus produces bradycardia in conscious rats. This is the first report that nesfatin-1 increases the parasympathetic cardiac tone.


Asunto(s)
Bradicardia/inducido químicamente , Proteínas de Unión al Calcio/farmacología , Proteínas de Unión al ADN/farmacología , Corazón/efectos de los fármacos , Corazón/inervación , Bulbo Raquídeo/efectos de los fármacos , Proteínas del Tejido Nervioso/farmacología , Neuronas/efectos de los fármacos , Nervio Vago/efectos de los fármacos , Animales , Animales Recién Nacidos , Presión Sanguínea/efectos de los fármacos , Bradicardia/fisiopatología , Calcio/metabolismo , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo Q/efectos de los fármacos , Proteínas de Unión al Calcio/administración & dosificación , Células Cultivadas , Proteínas de Unión al ADN/administración & dosificación , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Bulbo Raquídeo/citología , Potenciales de la Membrana/efectos de los fármacos , Microinyecciones , Proteínas del Tejido Nervioso/administración & dosificación , Nucleobindinas , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/fisiología , Taquifilaxis/fisiología , Telemetría , Nervio Vago/citología
4.
Toxicol Appl Pharmacol ; 248(1): 12-9, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20600211

RESUMEN

The persistent organochlorine pesticide lindane is still abundantly found in the environment and in human and animal tissue samples. Lindane induces a wide range of adverse health effects, which are at least partially mediated via the known inhibition of GABA(A) and glycine receptors. Additionally, lindane has been reported to increase the basal intracellular Ca(2+) concentration ([Ca(2+)](i)). As Ca(2+) triggers many cellular processes, including cell death and vesicular neurotransmitter release (exocytosis), we investigated whether lindane affects exocytosis, Ca(2+) homeostasis, production of reactive oxygen species (ROS) and cytotoxicity in neuroendocrine PC12 cells. Amperometric recordings and [Ca(2+)](i) imaging experiments with fura-2 demonstrated that lindane (≥ 10 µM) rapidly increases basal exocytosis and basal [Ca(2+)](i). Additional imaging and electrophysiological recordings revealed that this increase was largely due to a lindane-induced membrane depolarization and subsequent opening of N- and P/Q-type voltage-gated Ca(2+) channels (VGCC). On the other hand, lindane (≥ 3 µM) induced a concentration-dependent but non-specific inhibition of VGCCs, thereby limiting the lindane-induced increase in basal [Ca(2+)](i) and exocytosis. Importantly, the non-specific inhibition of VGCCs also reduced stimulation-evoked exocytosis and Ca(2+) influx. Though lindane exposure concentration-dependently increased ROS production, cell viability was not affected indicating that the used concentrations were not acute cytotoxic. These combined findings indicate that lindane has two, partly counteracting effects. Lindane causes membrane depolarization, thereby increasing basal [Ca(2+)](i) and exocytosis. In parallel, lindane inhibits VGCCs, thereby limiting the basal effects and reducing stimulation-evoked [Ca(2+)](i) and exocytosis. This study further underlines the need to consider presynaptic, non-receptor-mediated effects in human risk assessment.


Asunto(s)
Calcio/metabolismo , Exocitosis/efectos de los fármacos , Hexaclorociclohexano/toxicidad , Insecticidas/toxicidad , Animales , Canales de Calcio Tipo N/efectos de los fármacos , Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/efectos de los fármacos , Canales de Calcio Tipo Q/metabolismo , Relación Dosis-Respuesta a Droga , Electrofisiología , Hexaclorociclohexano/administración & dosificación , Homeostasis/efectos de los fármacos , Insecticidas/administración & dosificación , Células PC12 , Ratas , Especies Reactivas de Oxígeno/metabolismo
5.
Nat Neurosci ; 9(3): 389-97, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16474392

RESUMEN

Episodic ataxia type-2 (EA2) is caused by mutations in P/Q-type voltage-gated calcium channels that are expressed at high densities in cerebellar Purkinje cells. Because P/Q channels support neurotransmitter release at many synapses, it is believed that ataxia is caused by impaired synaptic transmission. Here we show that in ataxic P/Q channel mutant mice, the precision of Purkinje cell pacemaking is lost such that there is a significant degradation of the synaptic information encoded in their activity. The irregular pacemaking is caused by reduced activation of calcium-activated potassium (K(Ca)) channels and was reversed by pharmacologically increasing their activity with 1-ethyl-2-benzimidazolinone (EBIO). Moreover, chronic in vivo perfusion of EBIO into the cerebellum of ataxic mice significantly improved motor performance. Our data support the hypothesis that the precision of intrinsic pacemaking in Purkinje cells is essential for motor coordination and suggest that K(Ca) channels may constitute a potential therapeutic target in EA2.


Asunto(s)
Potenciales de Acción/genética , Relojes Biológicos/genética , Canales de Calcio Tipo P/genética , Ataxia Cerebelosa/metabolismo , Ataxia Cerebelosa/fisiopatología , Canales de Potasio Calcio-Activados/genética , Células de Purkinje/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Bencimidazoles/farmacología , Relojes Biológicos/efectos de los fármacos , Agonistas de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo P/efectos de los fármacos , Ataxia Cerebelosa/genética , Corteza Cerebelosa/efectos de los fármacos , Corteza Cerebelosa/metabolismo , Corteza Cerebelosa/fisiopatología , Regulación hacia Abajo/genética , Femenino , Masculino , Ratones , Ratones Mutantes Neurológicos , Ratones Transgénicos , Células de Purkinje/efectos de los fármacos , Células de Purkinje/patología , Membranas Sinápticas/genética , Membranas Sinápticas/metabolismo , Membranas Sinápticas/patología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética
6.
Neuron ; 45(6): 953-65, 2005 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-15797555

RESUMEN

While firing rate is well established as a relevant parameter for encoding information exchanged between neurons, the significance of other parameters is more conjectural. Here, we show that regularity of neuronal spike activities affects sensorimotor processing in tottering mutants, which suffer from a mutation in P/Q-type voltage-gated calcium channels. While the modulation amplitude of the simple spike firing rate of their floccular Purkinje cells during optokinetic stimulation is indistinguishable from that of wild-types, the regularity of their firing is markedly disrupted. The gain and phase values of tottering's compensatory eye movements are indistinguishable from those of flocculectomized wild-types or from totterings with the flocculus treated with P/Q-type calcium channel blockers. Moreover, normal eye movements can be evoked in tottering when the flocculus is electrically stimulated with regular spike trains mimicking the firing pattern of normal simple spikes. This study demonstrates the importance of regularity of firing in Purkinje cells for neuronal information processing.


Asunto(s)
Potenciales de Acción/fisiología , Canales de Calcio Tipo P/genética , Corteza Cerebelosa/fisiología , Movimientos Oculares/fisiología , Células de Purkinje/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Artefactos , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo P/efectos de los fármacos , Corteza Cerebelosa/citología , Corteza Cerebelosa/efectos de los fármacos , Estimulación Eléctrica , Retroalimentación/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Nistagmo Optoquinético/fisiología , Núcleo Olivar/fisiología , Estimulación Luminosa , Células de Purkinje/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Núcleos Vestibulares/fisiología , Percepción Visual/fisiología
7.
J Physiol ; 587(Pt 8): 1657-68, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19221121

RESUMEN

The second messenger, 3',5'-cyclic adenosine monophosphate (cAMP), is known to be modulated in taste buds following exposure to gustatory and other stimuli. Which taste cell type(s) (Type I/glial-like cells, Type II/receptor cells, or Type III/presynaptic cells) undergo taste-evoked changes of cAMP and what the functional consequences of such changes are remain unknown. Using Fura-2 imaging of isolated mouse vallate taste cells, we explored how elevating cAMP alters Ca(2+) levels in identified taste cells. Stimulating taste buds with forskolin (Fsk; 1 microm) + isobutylmethylxanthine (IBMX; 100 microm), which elevates cellular cAMP, triggered Ca(2+) transients in 38% of presynaptic cells (n = 128). We used transgenic GAD-GFP mice to show that cAMP-triggered Ca(2+) responses occur only in the subset of presynaptic cells that lack glutamic acid decarboxylase 67 (GAD). We never observed cAMP-stimulated responses in receptor cells, glial-like cells or GAD-expressing presynaptic cells. The response to cAMP was blocked by the protein kinase A inhibitor H89 and by removing extracellular Ca(2+). Thus, the response to elevated cAMP is a PKA-dependent influx of Ca(2+). This Ca(2+) influx was blocked by nifedipine (an inhibitor of L-type voltage-gated Ca(2+) channels) but was unperturbed by omega-agatoxin IVA and omega-conotoxin GVIA (P/Q-type and N-type channel inhibitors, respectively). Single-cell RT-PCR on functionally identified presynaptic cells from GAD-GFP mice confirmed the pharmacological analyses: Ca(v)1.2 (an L-type subunit) is expressed in cells that display cAMP-triggered Ca(2+) influx, while Ca(v)2.1 (a P/Q subunit) is expressed in all presynaptic cells, and underlies depolarization-triggered Ca(2+) influx. Collectively, these data demonstrate cross-talk between cAMP and Ca(2+) signalling in a subclass of taste cells that form synapses with gustatory fibres and may integrate tastant-evoked signals.


Asunto(s)
Calcio/fisiología , AMP Cíclico/fisiología , Receptores Presinapticos/fisiología , Sistemas de Mensajero Secundario/fisiología , Papilas Gustativas/fisiología , 1-Metil-3-Isobutilxantina/farmacología , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/fisiología , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/fisiología , Canales de Calcio Tipo Q/efectos de los fármacos , Canales de Calcio Tipo Q/fisiología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Señalización del Calcio/fisiología , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glutamato Descarboxilasa/biosíntesis , Glutamato Descarboxilasa/genética , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Endogámicos C57BL , Inhibidores de Fosfodiesterasa/farmacología , Receptores Acoplados a Proteínas G/fisiología , Receptores Presinapticos/efectos de los fármacos , Receptores Presinapticos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sistemas de Mensajero Secundario/efectos de los fármacos , Sistemas de Mensajero Secundario/genética , Papilas Gustativas/efectos de los fármacos
8.
J Cell Physiol ; 220(3): 727-47, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19441076

RESUMEN

Blockage of GABA-A receptors in hippocampal neuronal cultures triggers synchronous bursts of spikes initiating neuronal plasticity, partly mediated by changes of gene expression. By using specific pharmacological blockers, we have investigated which sources of Ca2+ entry primarily control changes of gene expression induced by 20 microM gabazine applied for 30 min (GabT). Intracellular Ca2+ transients were monitored with Ca2+ imaging while recording electrical activity with patch clamp microelectrodes. Concomitant transcription profiles were obtained using Affymetrix oligonucleotide microarrays and confirmed with quantitative RT-PCR. Blockage of NMDA receptors with 2-amino-5-phosphonovaleric acid (APV) did not reduce significantly somatic Ca2+ transients, which, on the contrary, were reduced by selective blockage of L, N, and P/Q types voltage gated calcium channels (VGCCs). Therefore, we investigated changes of gene expression in the presence of blockers of NMDA receptors and L, N, and P/Q VGCCs. Our results show that: (i) among genes upregulated by GabT, there are genes selectively dependent on NMDA activation, genes selectively dependent on L-type VGCCs and genes dependent on the activation of both channels; (ii) the majority of genes requires the concomitant activation of NMDA receptors and Ca2+ entry through VGCCs; (iii) blockage of N and P/Q VGCCs has an effect similar but not identical to blockage of L-type VGCCs.


Asunto(s)
Señalización del Calcio , Regulación de la Expresión Génica , Hipocampo/metabolismo , Neuronas/metabolismo , Animales , Animales Recién Nacidos , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/efectos de los fármacos , Canales de Calcio Tipo Q/metabolismo , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Dendritas/metabolismo , Antagonistas del GABA/farmacología , Antagonistas de Receptores de GABA-A , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/citología , Hipocampo/efectos de los fármacos , Potenciales de la Membrana , Microscopía Fluorescente , Neuronas/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transcripción Genética
9.
J Neurochem ; 108(3): 676-84, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19054275

RESUMEN

We investigated if stimulation of T-type Ca(2+) channels with sodium hydrosulfide (NaHS), a donor of hydrogen sulfide (H(2)S), could cause neuronal differentiation of NG108-15 cells. Like dibutyryl cyclic AMP (db-cAMP), treatment with NaHS at 1.5-13.5 mM for 16 h enhanced neurite outgrowth in a concentration-dependent manner. Synergistic neuritogenic effect was obtained in the cells stimulated with NaHS in combination with db-cAMP at subeffective concentrations. Exposure to NaHS or db-cAMP for 2 days resulted in enhancement of expression of high-voltage-activated currents consisting of N-, P/Q-, L- and also other types, but not of T-type currents. Mibefradil, a pan-T-type channel blocker, abolished the neuritogenesis induced by NaHS, but not by db-cAMP. The NaHS-evoked neuritogenesis was also completely blocked by pretreatment with BAPTA/AM, a chelator of intracellular Ca(2+), and by zinc chloride at a concentration known to selectively inhibit Ca(v)3.2 isoform of T-type Ca(2+) channels, but not Ca(v)3.1 or Ca(v)3.3. Further, L-ascorbate, recently proven to selectively inhibit Ca(v)3.2, abolished the neuritogenic effect of NaHS, but not db-cAMP. Our data thus demonstrate that NaHS/H(2)S is a novel inducer of neuronal differentiation in NG108-15 cells, as characterized by neuritogenesis and expression of high-voltage-activated currents, and suggest the involvement of T-type Ca(2+) channels, especially Ca(v)3.2.


Asunto(s)
Canales de Calcio Tipo T/fisiología , Canales de Calcio/biosíntesis , Canales de Calcio/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Neuritas/efectos de los fármacos , Bucladesina/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/fisiología , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/fisiología , Línea Celular Tumoral , Quelantes/farmacología , Cloruros/farmacología , Ditiotreitol/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Humanos , Neuritas/metabolismo , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Reactivos de Sulfhidrilo/farmacología , Compuestos de Zinc/farmacología
10.
J Neurosci ; 27(19): 5236-48, 2007 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-17494710

RESUMEN

P/Q-type (Ca(V)2.1) and N-type (Ca(V)2.2) Ca2+ channels are critical to stimulus-secretion coupling in the nervous system; feedback regulation of these channels by Ca2+ is therefore predicted to profoundly influence neurotransmission. Here we report divergent regulation of Ca2+-dependent inactivation (CDI) of native N- and P/Q-type Ca2+ channels by calmodulin (CaM) in adult chromaffin cells. Robust CDI of N-type channels was observed in response to prolonged step depolarizations, as well as repetitive stimulation with either brief step depolarizations or action potential-like voltage stimuli. Adenoviral expression of Ca2+-insensitive calmodulin mutants eliminated CDI of N-type channels. This is the first demonstration of CaM-dependent CDI of a native N-type channel. CDI of P/Q-type channels was by comparison modest and insensitive to expression of CaM mutants. Cloning of the C terminus of the Ca(V)2.1 alpha1 subunit from chromaffin cells revealed multiple splice variants lacking structural motifs required for CaM-dependent CDI. The physiological relevance of CDI on stimulus-coupled exocytosis was revealed by combining perforated-patch voltage-clamp recordings of pharmacologically isolated Ca2+ currents with membrane capacitance measurements of exocytosis. Increasing stimulus intensity to invoke CDI resulted in a significant decrease in the exocytotic efficiency of N-type channels compared with P/Q-type channels. Our results reveal unexpected diversity in CaM regulation of native Ca(V)2 channels and suggest that the ability of individual Ca2+ channel subtypes to undergo CDI may be tailored by alternative splicing to meet the specific requirements of a particular cellular function.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo P/metabolismo , Señalización del Calcio/fisiología , Calmodulina/metabolismo , Células Cromafines/metabolismo , Exocitosis/fisiología , Animales , Calcio/metabolismo , Calcio/farmacología , Canales de Calcio Tipo N/química , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo P/química , Canales de Calcio Tipo P/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Bovinos , Línea Celular , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Células Cromafines/efectos de los fármacos , Capacidad Eléctrica , Exocitosis/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína/genética
11.
Med Hypotheses ; 68(1): 131-6, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-16899342

RESUMEN

Voltage-dependent calcium channels (VDCCs) are heteromultimeric complexes that mediate calcium influx into cells in response to changes in membrane potential. The alpha1A subunit, encoded by the CACNA1A gene, is the pore-forming subunit specific to the neuronal P/Q-type VDCCs. These are implicated in fast excitatory and inhibitory neurotransmission. Their highest levels of expression are found in the Purkinje cell layer of the cerebellum, and in the hippocampus. Spinocerebellar ataxia type 6 (SCA 6) is an autosomal dominant cerebellar degeneration that shares neuropathological findings with late-onset cortical cerebellar atrophy (CCA). It is caused by an abnormal expansion of a trinucleotide (CAG) repeat in exon 47 of CACNA1A, on chromosome 19p13. This translates into a polyglutamine (polyQ) tract of prolonged length in the carboxyl terminal of the alpha1A subunit. Heterologous expression of mutated alpha1A subunits results in increased channel inactivation in electrophysiological tests. No treatment is known to improve SCA 6 at present, as none of the available drugs is able to reverse alpha1A dysregulation, nor disturbed protein aggregation, transport and localization in this disease. The drugs gabapentin and pregabalin interact with the alpha2delta subunit of the P/Q-type VDCCs. Gabapentin and pregabalin slow the rate of inactivation in recombinant P/Q-type VDCCs, expressed in Xenopus oocytes. These drugs improve ataxia in cases of CCA, olivopontocerebellar atrophy and ataxia-telangiectasia. On the basis of the neuropathological identity of SCA 6 with CCA, and given the capacity of gabapentin and pregabalin to decrease P/Q-type VDCCs inactivation, in this paper the authors put forward the hypothesis that the administration of gabapentin and pregabalin might prove beneficial in SCA 6 as the ataxia caused by this disease would be expected to improve. The authors hope that researchers working with this illness will be inspired and encouraged to undertake the appropriate clinical and experimental work.


Asunto(s)
Aminas/administración & dosificación , Bloqueadores de los Canales de Calcio/administración & dosificación , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo Q/efectos de los fármacos , Ácidos Ciclohexanocarboxílicos/administración & dosificación , Ataxias Espinocerebelosas/tratamiento farmacológico , Ácido gamma-Aminobutírico/análogos & derivados , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Gabapentina , Humanos , Pregabalina , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Ataxias Espinocerebelosas/metabolismo , Ácido gamma-Aminobutírico/administración & dosificación
12.
J Neurosci ; 25(36): 8282-94, 2005 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-16148236

RESUMEN

P-type (CaV2.1) Ca2+ channels are a central conduit of neuronal Ca2+ entry, so their Ca2+ feedback regulation promises widespread neurobiological impact. Heterologous expression of recombinant CaV2.1 channels demonstrates that the Ca2+ sensor calmodulin can trigger Ca2+-dependent facilitation (CDF) of channel opening. This facilitation occurs when local Ca2+ influx through individual channels selectively activates the C-terminal lobe of calmodulin. In neurons, however, such calmodulin-mediated processes have yet to be detected, and CDF of native P-type current has thus far appeared different, arguably triggered by other Ca2+ sensing molecules. Here, in cerebellar Purkinje somata abundant with prototypic P-type channels, we find that the C-terminal lobe of calmodulin does produce CDF, and such facilitation augments Ca2+ entry during stimulation by repetitive action-potential and complex-spike waveforms. Beyond recapitulating key features of recombinant channels, these neurons exhibit an additional modulatory dimension: developmental upregulation of CDF during postnatal week 2. This phenomenon reflects increasing somatic expression of CaV2.1 splice variants that manifest CDF and progressive dendritic targeting of variants lacking CDF. Calmodulin-triggered facilitation is thus fundamental to native CaV2.1 and rapidly enhanced during early development.


Asunto(s)
Canales de Calcio Tipo P/fisiología , Calmodulina/farmacología , Células de Purkinje/fisiología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo P/efectos de los fármacos , Línea Celular , Cerebelo/fisiología , Humanos , Riñón , Nimodipina/farmacología , Plásmidos , Ratas , Ratas Sprague-Dawley , Tetrodotoxina/farmacología , Transfección
13.
Br J Pharmacol ; 147(4): 391-401, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16402042

RESUMEN

1. The profile of opioid and cannabinoid receptors in neurons of the nucleus tractus solitarius (NTS) has been studied using the whole-cell configuration of the patch clamp technique. 2. Experiments with selective agonists and antagonists of opioid, ORL and cannabinoid receptors indicated that mu-opioid, kappa-opioid, ORL-1 and CB1, but not delta-opioid, receptors inhibit VDCCs in NTS. 3. Application of [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO; mu-opioid receptor agonist), Orphanin FQ (ORL-1 receptor agonist) and WIN55,122 (CB1 receptor agonist) caused inhibition of I(Ba) in a concentration-dependent manner, with IC50's of 390 nM, 220 nM and 2.2 microM, respectively. 4. Intracellular dialysis of the G(i)-protein antibody attenuated DAMGO-, Orphanin FQ- and WIN55,122-induced inhibition of I(Ba). 5. Both pretreatment with adenylate cyclase inhibitor and intracellular dialysis of the protein kinase A (PKA) inhibitor attenuated WIN55,122-induced inhibition of I(Ba) but not DAMGO- and Orphanin FQ-induced inhibition. 6. Mainly N- and P/Q-type VDCCs were inhibited by both DAMGO and Orphanin FQ, while L-type VDCCs were inhibited by WIN55,122. 7. These results suggest that mu- and kappa-opioid receptors and ORL-1 receptor inhibit N- and P/Q-type VDCCs via G alpha(i)-protein betagamma subunits, whereas CB1 receptors inhibit L-type VDCCs via G alpha(i)-proteins involving PKA in NTS.


Asunto(s)
Canales de Calcio/metabolismo , Antagonistas de Receptores de Cannabinoides , Antagonistas de Narcóticos , Receptores Opioides/agonistas , Núcleo Solitario/metabolismo , Animales , Animales Recién Nacidos , Calcio/fisiología , Canales de Calcio/clasificación , Canales de Calcio/efectos de los fármacos , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/efectos de los fármacos , Canales de Calcio Tipo Q/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Cinética , Péptidos Opioides/farmacología , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Receptores de Cannabinoides/efectos de los fármacos , Receptores de Cannabinoides/genética , Receptores Opioides/efectos de los fármacos , Receptores Opioides/genética , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/efectos de los fármacos , Núcleo Solitario/citología , Nociceptina
14.
Brain Res ; 1083(1): 164-73, 2006 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-16546143

RESUMEN

Although the etiology of neurodevelopmental mental disorders remains obscure, converging lines of evidence using animal modeling suggest a critical role for activity-dependent neurodevelopmental processes during neonatal life. Here, we report the behavioral effects of a novel technique designed to induce targeted, transient disruption of activity-dependent processes in early development via reduction of calcium-mediated neurotransmitter release. We examined the post-pubertal behavioral effects of neonatal (postnatal day 7) medial prefrontal cortex infusion of either vehicle or N-type and P/Q-type presynaptic voltage-dependent calcium channel blockers (omega-conotoxins MVIIA and MVIIC respectively; 6.8 and 45 pmol infused respectively) in rat pups. In a test of amphetamine-induced behavioral sensitization, neonatal omega-conotoxin MVIIA treatment significantly increased locomotion following repeated amphetamine injections (1.5 mg/kg i.p.) and significantly decreased locomotion following repeated saline injections relative to animals treated neonatally with vehicle. However, there was no effect of conotoxin treatment on the long-term expression of amphetamine sensitization. Neonatal treatment with omega-conotoxins had no effect on the other behaviors assayed, namely, acoustic startle response, prepulse inhibition of startle, novelty- and amphetamine-induced (1.5 mg/kg i.p.) locomotion, and anxiety-like behavior in the elevated plus-maze. These data confirm that transient, region-specific disruption of synaptic transmission during early development can have long-term effects on behaviors relevant to neurodevelopmental mental disorders.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Actividad Motora/fisiología , Corteza Prefrontal/metabolismo , Terminales Presinápticos/metabolismo , Transmisión Sináptica/fisiología , Envejecimiento/fisiología , Anfetamina/farmacología , Animales , Animales Recién Nacidos , Ansiedad/metabolismo , Ansiedad/fisiopatología , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Canales de Calcio/efectos de los fármacos , Canales de Calcio Tipo N/efectos de los fármacos , Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/metabolismo , Estimulantes del Sistema Nervioso Central , Modelos Animales de Enfermedad , Interacciones Farmacológicas/fisiología , Trastornos Mentales/metabolismo , Trastornos Mentales/fisiopatología , Actividad Motora/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/crecimiento & desarrollo , Terminales Presinápticos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología , Transmisión Sináptica/efectos de los fármacos , Tiempo , omega-Conotoxinas/farmacología
15.
Brain Res ; 1115(1): 200-8, 2006 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-16920080

RESUMEN

To identify whether altered expressions of voltage gated Ca(2+) channel (VGCC) are linked to inhibitory transmission abnormalities in the gerbil hippocampus, we investigated the effects of GABA receptor or somatostatin receptor (SST) antagonists/agonists on VGCC immunoreactivity in vivo. VGCC immunoreactivities in the hippocampus were significantly higher in seizure sensitive (SS) gerbils than in seizure resistant (SR) gerbils. P/Q-type VGCC immunoreactivity in the gerbil hippocampus was reduced by enhancement in GABA(A) and GABA(B) receptor-mediated transmission, but not by SST-mediated transmission. N-type VGCC immunoreactivity was reduced only by a SST agonist, whereas L-type (alpha1C) VGCC immunoreactivity was reduced only by a GABA(A) receptor agonist, and L-type (alpha1D) VGCC immunoreactivity was modulated by the GABA(B) receptor acting drugs. These findings provide a comprehensive description of the differential responses of VGCC subunits to alteration in GABAergic or somatostatinergic transmission. These findings also suggest that up-regulated VGCC immunoreactivity may be consequence of the neuronal excitability caused by a reduction in inhibitory neurotransmission in the gerbil hippocampus.


Asunto(s)
Canales de Calcio/metabolismo , Epilepsia/metabolismo , Hipocampo/metabolismo , Somatostatina/metabolismo , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Canales de Calcio/efectos de los fármacos , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo N/efectos de los fármacos , Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Epilepsia/fisiopatología , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/fisiopatología , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Agonistas de Receptores de GABA-A , Antagonistas de Receptores de GABA-A , Agonistas de Receptores GABA-B , Antagonistas de Receptores de GABA-B , Gerbillinae , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Inmunohistoquímica , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de GABA-A/metabolismo , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/metabolismo , Transmisión Sináptica/efectos de los fármacos
16.
J Neurosci ; 24(46): 10402-9, 2004 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-15548655

RESUMEN

Presynaptic calcium influx at most excitatory central synapses is carried by both Cav2.1 and Cav2.2 channels. The kinetics and modulation of Cav2.1 and Cav2.2 channels differ and may affect presynaptic calcium influx. We compared release dynamics at CA3/CA1 synapses in rat hippocampus after selective blockade of either channel subtype and subsequent quantal content restoration. Selective blockade of Cav2.1 channels enhanced paired-pulse facilitation, whereas blockade of Cav2.2 channels decreased it. This effect was observed at short (50 msec) but not longer (500 msec) intervals and was maintained during prolonged bursts of presynaptic activity. It did not reflect differences in the distance of the channels from the calcium sensor. The suppression of this effect by preincubation with the G(o/i)-protein inhibitor pertussis toxin suggests instead that high-frequency stimulation relieves inhibition of Cav2.2 by G(o/i), thereby increasing the number of available channels.


Asunto(s)
Canales de Calcio Tipo N/fisiología , Canales de Calcio Tipo P/fisiología , Canales de Calcio Tipo Q/fisiología , Calcio/metabolismo , Hipocampo/metabolismo , Sinapsis/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/efectos de los fármacos , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo Q/efectos de los fármacos , Estimulación Eléctrica , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Hipocampo/ultraestructura , Técnicas In Vitro , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica
17.
J Neurosci ; 20(19): 7174-82, 2000 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11007873

RESUMEN

The recognition of neuronal Ca channel diversity has led to considerable efforts to identify useful classification criteria. Here, we revisit the pharmacological definition of P- and Q-type Ca channels, which is based on their respective high and low sensitivity to the spider omega-agatoxin-IVA (omega-Aga-IVA), using whole-cell recordings of the Ca channel currents carried by 5 mM Ba(2+) in isolated rat subthalamic and sympathetic neurons. In subthalamic neurons, omega-Aga-IVA (1 microM) targeted multiple Ca channels. One population was blocked with high potency. These channels carried 50.4 +/- 3.4% (n = 5) of the control current and showed the same inactivation kinetics and voltage-dependent high affinity for omega-Aga-IVA as do prototypic P-type Ca channels. Other Ca channels were targeted with weaker potency. This heterogeneous population contributed to 14.0 +/- 1.7% (n = 5) of the control current. It included N-type Ca channels as well as high-threshold Ca channels that displayed the pharmacological signature of Q-type Ca channels but resembled P-type Ca channels in their gating properties. N-type Ca current block by omega-Aga-IVA (1 microM) was further investigated in sympathetic neurons, which mainly express this Ca channel type. Block was incomplete ( approximately 30% of the control current). Its relief at positive potentials was consistent with omega-Aga-IVA acting as a channel-gating modifier. These effects did not reflect a complete loss of selectivity, because omega-Aga-IVA (1 microM) had no effect on subthalamic Na and K currents or their T- and L-type Ca currents. Our data confirm that omega-Aga-IVA is a selective P-type Ca channel blocker. However, its diminished selectivity in the micromolar range limits its usefulness for functional studies of Q-type Ca channels.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , omega-Agatoxina IVA/farmacología , Animales , Bario/metabolismo , Bario/farmacología , Canales de Calcio Tipo N/efectos de los fármacos , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/metabolismo , Separación Celular , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Ganglios Simpáticos/citología , Ganglios Simpáticos/efectos de los fármacos , Ganglios Simpáticos/metabolismo , Técnicas In Vitro , Activación del Canal Iónico/efectos de los fármacos , Neuronas/citología , Técnicas de Placa-Clamp , Potasio/metabolismo , Células de Purkinje/citología , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo , Ratas , Sodio/metabolismo , Núcleo Subtalámico/citología , Núcleo Subtalámico/efectos de los fármacos , Núcleo Subtalámico/metabolismo
18.
J Neurosci ; 20(24): 9046-52, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11124981

RESUMEN

Ca(2+) channel inactivation is a key element in controlling the level of Ca(2+) entry through voltage-gated Ca(2+) channels. Interaction between the pore-forming alpha(1) subunit and the auxiliary beta subunit is known to be a strong modulator of voltage-dependent inactivation. Here, we demonstrate that an N-terminal membrane anchoring site (MAS) of the beta(2a) subunit strongly reduces alpha(1A) (Ca(V)2.1) Ca(2+) channel inactivation. This effect can be mimicked by the addition of a transmembrane segment to the N terminus of the beta(2a) subunit. Inhibition of inactivation by beta(2a) also requires a link between MAS and another important molecular determinant, the beta interaction domain (BID). Our data suggest that mobility of the Ca(2+) channel I-II loop is necessary for channel inactivation. Interaction of this loop with other identified intracellular channel domains may constitute the basis of voltage-dependent inactivation. We thus propose a conceptually novel mechanism for slowing of inactivation by the beta(2a) subunit, in which the immobilization of the channel inactivation gate occurs by means of MAS and BID.


Asunto(s)
Canales de Calcio/metabolismo , Activación del Canal Iónico/fisiología , Subunidades de Proteína , Animales , Bario/farmacología , Sitios de Unión/genética , Canales de Calcio/efectos de los fármacos , Canales de Calcio/genética , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/genética , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/efectos de los fármacos , Canales de Calcio Tipo Q/genética , Canales de Calcio Tipo Q/metabolismo , Línea Celular , Quelantes/farmacología , Electrofisiología , Técnica del Anticuerpo Fluorescente , Glutatión Transferasa/genética , Humanos , Activación del Canal Iónico/efectos de los fármacos , Mutagénesis Sitio-Dirigida , Oocitos/citología , Oocitos/metabolismo , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Relación Estructura-Actividad , Transfección , Xenopus
19.
J Neurosci ; 20(22): 8323-30, 2000 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11069939

RESUMEN

Patch-clamp measurements of Ca(2+) currents and membrane capacitance were performed on slices of mouse adrenal glands, using the perforated-patch configuration of the patch-clamp technique. These recording conditions are much closer to the in vivo situation than those used so far in most electrophysiological studies in adrenal chromaffin cells (isolated cells maintained in culture and whole-cell configuration). We observed profound discrepancies in the quantities of Ca(2+) channel subtypes (P-, Q-, N-, and L-type Ca(2+) channels) described for isolated mouse chromaffin cells maintained in culture. Differences with respect to previous studies may be attributable not only to culture conditions, but also to the patch-clamp configuration used. Our experiments revealed the presence of a Ca(2+) channel subtype never before described in chromaffin cells, a toxin and dihydropyridine-resistant Ca(2+) channel with fast inactivation kinetics, similar to the R-type Ca(2+) channel described in neurons. This channel contributes 22% to the total Ca(2+) current and controls 55% of the rapid secretory response evoked by short depolarizing pulses. Our results indicate that R-type Ca(2+) channels are in close proximity with the exocytotic machinery to rapidly regulate the secretory process.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Canales de Calcio Tipo R/metabolismo , Células Cromafines/metabolismo , Glándulas Suprarrenales/citología , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/efectos de los fármacos , Canales de Calcio Tipo Q/metabolismo , Canales de Calcio Tipo R/efectos de los fármacos , Membrana Celular/fisiología , Células Cromafines/citología , Conductividad Eléctrica , Estimulación Eléctrica , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/fisiología , Exocitosis/fisiología , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Técnicas de Placa-Clamp
20.
J Neurosci ; 22(5): RC213, 2002 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11880534

RESUMEN

The discovery of mis-sense mutations in the alpha1A subunit of the P/Q-type calcium channel in patients with familial hemiplegic migraine indicates the potential involvement of dysfunctional ion channels in migraine. The periaqueductal gray (PAG) region of the brainstem modulates craniovascular nociception and, through its role in the descending pain modulation system, may contribute to migraine pathophysiology. In this study we sought to investigate the possible link between the genetic mutations found in migraineurs and the PAG as a modulator of craniovascular nociception. We microinjected the P/Q-type calcium-channel blocker omega-agatoxin IVA into the rat ventrolateral PAG (vlPAG). We examined its effect on the nociceptive transmission of second-order neurons recorded in the trigeminal nucleus caudalis and activated by stimulation of the parietal dura mater. After injection of agatoxin into the vlPAG (n = 20) responses to dural stimulation were facilitated by 143% (p < 0.0001) for Adelta-fiber activity and 180% for C-fiber activity (p < 0.05). Similarly, spontaneous background activity increased by 163% (p < 0.0001). These results demonstrate that P/Q-type calcium channels in the PAG play a role in modulating trigeminal nociception and suggest a role for dysfunctional P/Q-type calcium channels in migraine pathophysiology.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo P/efectos de los fármacos , Canales de Calcio Tipo Q/efectos de los fármacos , Trastornos Migrañosos/etiología , Sustancia Gris Periacueductal/efectos de los fármacos , Neuralgia del Trigémino/inducido químicamente , Animales , Presión Sanguínea/efectos de los fármacos , Canales de Calcio Tipo P/genética , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/genética , Canales de Calcio Tipo Q/metabolismo , Antagonistas del GABA/farmacología , Masculino , Microinyecciones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Dimensión del Dolor/efectos de los fármacos , Sustancia Gris Periacueductal/metabolismo , Ratas , Ratas Sprague-Dawley , Núcleo Caudal del Trigémino/efectos de los fármacos , Núcleo Caudal del Trigémino/fisiopatología , Neuralgia del Trigémino/fisiopatología , omega-Agatoxina IVA/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA