Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
BMC Ophthalmol ; 20(1): 182, 2020 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375703

RESUMEN

BACKGROUND: So far, only indirect evidence exists for the pharmacoresistant R-type voltage-gated Ca2+ channel (VGCC) to be involved in transretinal signaling by triggering GABA-release onto ON-bipolar neurons. This release of inhibitory neurotransmitters was deduced from the sensitivity of the b-wave to stimulation by Ni2+, Zn2+ and Cu2+. To further confirm the interpretation of these findings, we compared the effects of Cu2+ application and chelation (using kainic acid, KA) on the neural retina from wildtype and Cav2.3-deficient mice. Furthermore, the immediately effect of KA on the ERG b-wave modulation was assessed. METHODS: Transretinal signaling was recorded as an ERG from the superfused murine retina isolated from wildtype and Cav2.3-deficient mice. RESULTS: In mice, the stimulating effect of 100 nM CuCl2 is absent in the retinae from Cav2.3-deficient mice, but prominent in Cav2.3-competent mice. Application of up to 3 mM tricine does not affect the murine b-wave in both genotypes, most likely because of chelating amino acids present in the murine nutrient solution. Application of 27 µM KA significantly increased the b-wave amplitude in wild type and Cav2.3 (-|-) mice. This effect can most likely be explained by the stimulation of endogenous KA-receptors described in horizontal, OFF-bipolar, amacrine or ganglion cells, which could not be fully blocked in the present study. CONCLUSION: Cu2+-dependent modulation of transretinal signaling only occurs in the murine retina from Cav2.3 competent mice, supporting the ideas derived from previous work in the bovine retina that R-type Ca2+ channels are involved in shaping transretinal responses during light perception.


Asunto(s)
Cobre/metabolismo , Electrorretinografía/métodos , Retina/metabolismo , Animales , Canales de Calcio Tipo R/deficiencia , Proteínas de Transporte de Catión/deficiencia , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Estimulación Luminosa , Retina/citología , Transducción de Señal
2.
Cell Physiol Biochem ; 44(3): 935-947, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29176325

RESUMEN

BACKGROUND/AIMS: Lamotrigine (LTG) is a popular modern antiepileptic drug (AED), however, its mechanism of action has yet to be fully understood, as it is known to modulate many members of several ion channel families. In heterologous systems, LTG inhibits Cav2.3 (R-type) calcium currents, which contribute to kainic-acid- (KA) induced epilepsy in vivo. To gain insight into the role of R-type currents in LTG drug action in vivo, we compared the effects of LTG to topiramate and lacosamide in Cav2.3-deficient mice and controls on KA-induced seizures. METHODS: Behavioral seizure rating and quantitative electrocorticography were performed after injection of 20 mg/kg [and 30 mg/kg] KA. One hour before KA injection, mice were pretreated with either 30 mg/kg LTG, 50 mg/kg topiramate (TPM) or 30 mg/kg lacosamide (LSM). RESULTS: Ablation of Cav2.3 reduced total seizure scores by 28.6% (p=0.0012) and pretreatment with LTG reduced seizure activity of control mice by 23.2% (p=0.02). In Cav2.3-deficient mice LTG pretreatment increased seizure activity by 22.1% (p=0.018) and increased the percentage of degenerated CA1 pyramidal neurons (p=0.02). All three tested AEDs reduced seizure activity in control mice, however only the non-calcium channel modulating AED, LSM had an anticonvulsive effect in Cav2.3-deficient mice. Furthermore LTG altered electrocorticographic parameters differently in the two genotypes, decreasing relative power of ictal spikes in control mice compared to Cav2.3-defcient mice. CONCLUSION: These findings give first in vivo evidence for an essential role for Cav2.3 in LTG pharmacology and shed light on a paradoxical effect of LTG in their absence. Furthermore, LTG appears to promote ictal activity in Cav2.3-deficient mice resulting in increased neurotoxicity in the CA1 region. This paradoxical mechanism, possibly reflecting rebound hyperexcitation of pyramidal CA1 neurons after increased inhibition, may be key in understanding LTG-induced seizure aggravation, observed in clinical practice.


Asunto(s)
Anticonvulsivantes/farmacología , Conducta Animal/efectos de los fármacos , Canales de Calcio Tipo R/genética , Epilepsia/patología , Fármacos Neuroprotectores/farmacología , Triazinas/farmacología , Acetamidas/farmacología , Acetamidas/uso terapéutico , Animales , Anticonvulsivantes/uso terapéutico , Canales de Calcio Tipo R/deficiencia , Electrocorticografía , Epilepsia/inducido químicamente , Epilepsia/prevención & control , Fructosa/análogos & derivados , Fructosa/farmacología , Fructosa/uso terapéutico , Genotipo , Inmunohistoquímica , Ácido Kaínico/toxicidad , Lacosamida , Lamotrigina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Neuroprotectores/uso terapéutico , Células Piramidales/efectos de los fármacos , Células Piramidales/patología , Topiramato , Triazinas/uso terapéutico
3.
Biochim Biophys Acta ; 1853(5): 953-64, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25603538

RESUMEN

Peptide-hormone secretion is partially triggered by Ca2+ influx through voltage-gated Ca2+ channels (VGCCs) and gene inactivation of Zn2+-sensitive Cav2.3-type VGCCs is associated with disturbed glucose homeostasis in mice. Zn2+ has been implicated in pancreatic islet cell crosstalk and recent findings indicate that sudden cessation of Zn2+ supply during hypoglycemia triggers glucagon secretion in rodents. Here we show that diethyldithiocarbamate (DEDTC), a chelating agent for Zn2+ and other group IIB metal ions, differentially affects blood glucose and serum peptide hormone level in wild-type mice and mice lacking the Cav2.3-subunit. Fasting glucose and glucagon level were significantly higher in Cav2.3-deficient compared to wild-type mice, while DEDTC Zn2+-chelation produced a significant and correlated increase of blood glucose and serum glucagon concentration in wild-type but not Cav2.3-deficient mice. Glucose tolerance tests revealed severe glucose intolerance in Zn2+-depleted Cav2.3-deficient but not vehicle-treated Cav2.3-deficient or Zn2+-depleted wildtype mice. Collectively, these findings indicate that Cav2.3 channels are critically involved in the Zn2+-mediated suppression of glucagon secretion during hyperglycemia. Especially under conditions of Zn2+ deficiency, ablation or dysfunction of Cav2.3 channels may lead to severe disturbances in glucose homeostasis.


Asunto(s)
Canales de Calcio Tipo R/metabolismo , Proteínas de Transporte de Catión/metabolismo , Quelantes/farmacología , Ditiocarba/farmacología , Glucagón/metabolismo , Zinc/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Canales de Calcio Tipo R/deficiencia , Proteínas de Transporte de Catión/deficiencia , Ayuno/sangre , Femenino , Eliminación de Gen , Glucagón/sangre , Prueba de Tolerancia a la Glucosa , Células HEK293 , Humanos , Hiperinsulinismo/metabolismo , Insulina/metabolismo , Secreción de Insulina , Activación del Canal Iónico/efectos de los fármacos , Iones , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Modelos Biológicos
4.
J Neurosci ; 32(39): 13555-67, 2012 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23015445

RESUMEN

R-type calcium channels (RTCCs) are well known for their role in synaptic plasticity, but little is known about their subcellular distribution across various neuronal compartments. Using subtype-specific antibodies, we characterized the regional and subcellular localization of Ca(v)2.3 in mice and rats at both light and electron microscopic levels. Ca(v)2.3 immunogold particles were found to be predominantly presynaptic in the interpeduncular nucleus, but postsynaptic in other brain regions. Serial section analysis of electron microscopic images from the hippocampal CA1 revealed a higher density of immunogold particles in the dendritic shaft plasma membrane compared with the pyramidal cell somata. However, the labeling densities were not significantly different among the apical, oblique, or basal dendrites. Immunogold particles were also observed over the plasma membrane of dendritic spines, including both synaptic and extrasynaptic sites. Individual spine heads contained <20 immunogold particles, with an average density of ∼260 immunoparticles per µm(3) spine head volume, in accordance with the density of RTCCs estimated using calcium imaging (Sabatini and Svoboda, 2000). The Ca(v)2.3 density was variable among similar-sized spine heads and did not correlate with the density in the parent dendrite, implying that spines are individual calcium compartments operating autonomously from their parent dendrites.


Asunto(s)
Canales de Calcio Tipo R/metabolismo , Canales de Calcio Tipo R/ultraestructura , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/ultraestructura , Neuronas/metabolismo , Neuronas/ultraestructura , Análisis de Varianza , Animales , Animales Recién Nacidos , Encéfalo/citología , Canales de Calcio Tipo R/química , Canales de Calcio Tipo R/deficiencia , Proteínas de Transporte de Catión/química , Proteínas de Transporte de Catión/deficiencia , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Dendritas/metabolismo , Dendritas/ultraestructura , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Epítopos/metabolismo , Femenino , Cobayas , Imagenología Tridimensional , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Inmunoelectrónica , Péptidos/metabolismo , Densidad Postsináptica/metabolismo , Densidad Postsináptica/ultraestructura , Ratas , Estadística como Asunto , Estadísticas no Paramétricas , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/ultraestructura
5.
Cell Biochem Funct ; 31(5): 434-49, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23086800

RESUMEN

Voltage-gated Ca(2+) channels regulate cardiac automaticity, rhythmicity and excitation-contraction coupling. Whereas L-type (Cav 1·2, Cav 1·3) and T-type (Cav 3·1, Cav 3·2) channels are widely accepted for their functional relevance in the heart, the role of Cav 2·3 Ca(2+) channels expressing R-type currents remains to be elucidated. We have investigated heart rate dynamics in control and Cav 2·3-deficient mice using implantable electrocardiogram radiotelemetry and pharmacological injection experiments. Autonomic block revealed that the intrinsic heart rate does not differ between both genotypes. Systemic administration of isoproterenol resulted in a significant reduction in interbeat interval in both genotypes. It remained unaffected after administering propranolol in Cav 2·3(-|-) mice. Heart rate from isolated hearts as well as atrioventricular conduction for both genotypes differed significantly. Additionally, we identified and analysed the developmental expression of two splice variants, i.e. Cav 2·3c and Cav 2·3e. Using patch clamp technology, R-type currents could be detected in isolated prenatal cardiomyocytes and be related to R-type Ca(2+) channels. Our results indicate that on the systemic level, the pharmacologically inducible heart rate range and heart rate reserve are impaired in Cav 2·3 (-|-) mice. In addition, experiments on Langendorff perfused hearts elucidate differences in basic properties between both genotypes. Thus, Cav 2·3 does not only contribute to the cardiac autonomous nervous system but also to intrinsic rhythm propagation.


Asunto(s)
Canales de Calcio Tipo R/genética , Proteínas de Transporte de Catión/genética , Frecuencia Cardíaca/efectos de los fármacos , Corazón/efectos de los fármacos , Isoproterenol/farmacología , Miocitos Cardíacos/efectos de los fármacos , Propranolol/farmacología , Empalme Alternativo , Animales , Antiarrítmicos/farmacología , Calcio/metabolismo , Canales de Calcio Tipo R/deficiencia , Cardiotónicos/farmacología , Proteínas de Transporte de Catión/deficiencia , Células Cultivadas , Corazón/fisiología , Frecuencia Cardíaca/fisiología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Noqueados , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Telemetría
6.
Sci Rep ; 11(1): 13972, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34234221

RESUMEN

High voltage-activated Cav2.3 R-type Ca2+ channels and low voltage-activated Cav3.2 T-type Ca2+ channels were reported to be involved in numerous physiological and pathophysiological processes. Many of these findings are based on studies in Cav2.3 and Cav3.2 deficient mice. Recently, it has been proposed that inbreeding of Cav2.3 and Cav3.2 deficient mice exhibits significant deviation from Mendelian inheritance and might be an indication for potential prenatal lethality in these lines. In our study, we analyzed 926 offspring from Cav3.2 breedings and 1142 offspring from Cav2.3 breedings. Our results demonstrate that breeding of Cav2.3 deficient mice shows typical Mendelian inheritance and that there is no indication of prenatal lethality. In contrast, Cav3.2 breeding exhibits a complex inheritance pattern. It might be speculated that the differences in inheritance, particularly for Cav2.3 breeding, are related to other factors, such as genetic specificities of the mutant lines, compensatory mechanisms and altered sperm activity.


Asunto(s)
Canales de Calcio Tipo R/deficiencia , Canales de Calcio Tipo T/genética , Proteínas de Transporte de Catión/deficiencia , Genotipo , Endogamia , Patrón de Herencia , Herencia Multifactorial , Mutación , Animales , Femenino , Endogamia/métodos , Masculino , Ratones , Fenotipo
7.
PLoS One ; 10(9): e0139332, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26418566

RESUMEN

SK2- and KV4.2-containing K+ channels modulate evoked synaptic potentials in CA1 pyramidal neurons. Each is coupled to a distinct Ca2+ source that provides Ca2+-dependent feedback regulation to limit AMPA receptor (AMPAR)- and NMDA receptor (NMDAR)-mediated postsynaptic depolarization. SK2-containing channels are activated by Ca2+ entry through NMDARs, whereas KV4.2-containing channel availability is increased by Ca2+ entry through SNX-482 (SNX) sensitive CaV2.3 R-type Ca2+ channels. Recent studies have challenged the functional coupling between NMDARs and SK2-containing channels, suggesting that synaptic SK2-containing channels are instead activated by Ca2+ entry through R-type Ca2+ channels. Furthermore, SNX has been implicated to have off target affects, which would challenge the proposed coupling between R-type Ca2+ channels and KV4.2-containing K+ channels. To reconcile these conflicting results, we evaluated the effect of SK channel blocker apamin and R-type Ca2+ channel blocker SNX on evoked excitatory postsynaptic potentials (EPSPs) in CA1 pyramidal neurons from CaV2.3 null mice. The results show that in the absence of CaV2.3 channels, apamin application still boosted EPSPs. The boosting effect of CaV2.3 channel blockers on EPSPs observed in neurons from wild type mice was not observed in neurons from CaV2.3 null mice. These data are consistent with a model in which SK2-containing channels are functionally coupled to NMDARs and KV4.2-containing channels to CaV2.3 channels to provide negative feedback regulation of EPSPs in the spines of CA1 pyramidal neurons.


Asunto(s)
Apamina/farmacología , Canales de Calcio Tipo R/fisiología , Proteínas de Transporte de Catión/fisiología , Células Piramidales/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos , Animales , Región CA1 Hipocampal/citología , Calcio/metabolismo , Canales de Calcio Tipo R/deficiencia , Canales de Calcio Tipo R/genética , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/genética , Potenciales Evocados/fisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Células Piramidales/metabolismo , Células Piramidales/fisiología , Venenos de Araña/farmacología , Potenciales Sinápticos/genética , Potenciales Sinápticos/fisiología
8.
Neurosci Res ; 43(1): 1-7, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12074836

RESUMEN

Recently several mutant mouse lines lacking neuronal voltage-dependent Ca(2+) channels (VDCCs) have been established by the use of gene targeting in embryonic stem cells. Pain-related behaviors in Ca(v)2.2 (alpha(1B)) and Ca(v)2.3 (alpha(1E)) knockout mice were studied to gain further insight into the mechanism of pain transmission, where VDCCs are thought to play important roles. We review here the data from these recent studies. Ca(v)2.3-/- mice showed normal responses to acute painful stimuli, and reduced responses to the somatic inflammatory pain stimuli. Ca(v)2.3+/- mice exhibited reduced symptoms of visceral inflammatory pain. Ca(v)2.3-/- mice showed abnormal behavior related to the descending antinociceptive mechanism activated by the intraperitoneal injection of acetic acid. Ca(v)2.2-/- mice showed variable acute nociceptive responses depending on the mutant lines. However, all the lines of Ca(v)2.2-/- mice exhibited reduced responses in the phase 2 of the formalin test, suggesting a suppression of inflammatory pain. Furthermore Ca(v)2.2-/- mice showed markedly reduced neuropathic pain symptoms after spinal nerve ligation. Impaired antinociception, similar to that seen in the Ca(v)2.3-/- mice, was also observed in the Ca(v)2.2-/- mice. Therefore, it is suggested that these mutant mice could provide novel models to delineate the nociceptive and antinociceptive mechanisms.


Asunto(s)
Canales de Calcio Tipo N/deficiencia , Canales de Calcio Tipo R/deficiencia , Sistema Nervioso Central/metabolismo , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Nociceptores/metabolismo , Dolor/metabolismo , Transmisión Sináptica/fisiología , Ácido Acético/farmacología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo R/genética , Sistema Nervioso Central/fisiopatología , Modelos Animales de Enfermedad , Femenino , Calor/efectos adversos , Inflamación/genética , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Ratones , Ratones Noqueados , Modelos Neurológicos , Vías Nerviosas/fisiopatología , Nociceptores/efectos de los fármacos , Dolor/genética , Dolor/fisiopatología , Dimensión del Dolor , Fenotipo , Estimulación Física
9.
Neuroscience ; 205: 125-39, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22240250

RESUMEN

Hippocampal theta oscillations are key elements in numerous behavioral and cognitive processes. Based on the dualistic theory of theta oscillations, one can differentiate between atropine-sensitive and atropine-insensitive theta subtypes. Urethane-induced atropine-sensitive theta oscillations are driven by muscarinic signal transduction pathways through G protein q/11 alpha subunit (Gα(q/11)), phospholipase ß( ») (PLCß( »), inositol trisphosphate (InsP3), diacylglycerole (DAG), and protein kinase C (PKC). Recent findings illustrate that Ca(v)2.3 Ca²âº channels are important targets of muscarinic signaling in the hippocampus mediating plateau potential generation, epileptiform burst activity, and complex rhythm generation in the septohippocampal network. To investigate the physiological implications of Ca(v)2.3 Ca²âº channels in hippocampal theta oscillations we performed radiotelemetric intrahippocampal (cornu ammonis (CA1)) recordings in urethane (800 mg/kg, i.p.) and atropine (50 mg/kg, i.p.) treated Ca(v)2.3⁺/⁺ and Ca(v)2.3⁻/⁻ mice followed by wavelet analysis of EEG data. Our results demonstrate that Ca(v)2.3 ablation, unlike PLCß1 deletion, does not result in complete abolishment of urethane-induced theta oscillations and that both mean and total theta duration is not significantly inhibited by subsequent atropine treatment, indicating that Ca(v)2.3 Ca²âº channels are important mediators of atropine-sensitive theta. Although theta frequency remained unchanged between both genotypes, the temporal characteristics of theta distribution, that is, theta architecture were significantly affected by the loss of Ca(v)2.3 Ca²âº channels. Our data suggest, for the first time, that Ca(v)2.3 voltage-gated Ca²âº channels (VGCC) are an important factor in septohippocampal synchronization associated with theta oscillation.


Asunto(s)
Atropina/farmacología , Relojes Biológicos/fisiología , Canales de Calcio Tipo R/fisiología , Proteínas de Transporte de Catión/fisiología , Hipocampo/metabolismo , Ritmo Teta/fisiología , Animales , Relojes Biológicos/efectos de los fármacos , Canales de Calcio Tipo R/deficiencia , Canales de Calcio Tipo R/genética , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Ratones , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Ritmo Teta/efectos de los fármacos
10.
Neuron ; 70(1): 95-108, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21482359

RESUMEN

Neurons of the reticular thalamus (RT) display oscillatory burst discharges that are believed to be critical for thalamocortical network oscillations related to absence epilepsy. Ca²+-dependent mechanisms underlie such oscillatory discharges. However, involvement of high-voltage activated (HVA) Ca²+ channels in this process has been discounted. We examined this issue closely using mice deficient for the HVA Ca(v)2.3 channels. In brain slices of Ca(v)2.3⁻/⁻, a hyperpolarizing current injection initiated a low-threshold burst of spikes in RT neurons; however, subsequent oscillatory burst discharges were severely suppressed, with a significantly reduced slow afterhyperpolarization (AHP). Consequently, the lack of Ca(v)2.3 resulted in a marked decrease in the sensitivity of the animal to γ-butyrolactone-induced absence epilepsy. Local blockade of Ca(v)2.3 channels in the RT mimicked the results of Ca(v)2.3⁻/⁻ mice. These results provide strong evidence that Ca(v)2.3 channels are critical for oscillatory burst discharges in RT neurons and for the expression of absence epilepsy.


Asunto(s)
Potenciales de Acción/fisiología , Canales de Calcio Tipo R/fisiología , Proteínas de Transporte de Catión/fisiología , Electroencefalografía , Epilepsia Tipo Ausencia/fisiopatología , Formación Reticular/fisiología , Núcleos Talámicos/fisiología , 4-Butirolactona/toxicidad , Potenciales de Acción/genética , Animales , Canales de Calcio Tipo R/deficiencia , Canales de Calcio Tipo R/genética , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Electroencefalografía/métodos , Epilepsia Tipo Ausencia/inducido químicamente , Epilepsia Tipo Ausencia/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
11.
PLoS One ; 6(6): e20939, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21695265

RESUMEN

The majority of glutamatergic synapses formed onto principal neurons of the mammalian central nervous system are associated with dendritic spines. Spines are tiny protuberances that house the proteins that mediate the response of the postsynaptic cell to the presynaptic release of glutamate. Postsynaptic signals are regulated by an ion channel signaling cascade that is active in individual dendritic spines and involves voltage-gated calcium (Ca) channels, small conductance (SK)-type Ca-activated potassium channels, and NMDA-type glutamate receptors. Pharmacological studies using the toxin SNX-482 indicated that the voltage-gated Ca channels that signal within spines to open SK channels belong to the class Ca(V)2.3, which is encoded by the Alpha-1E pore-forming subunit. In order to specifically test this conclusion, we examined the effects of SNX-482 on synaptic signals in acute hippocampal slices from knock-out mice lacking the Alpha-1E gene. We find that in these mice, application of SNX-482 has no effect on glutamate-uncaging evoked synaptic potentials and Ca influx, indicating that that SNX-482 indeed acts via the Alpha-1E-encoded Ca(V)2.3 channel.


Asunto(s)
Canales de Calcio Tipo R/genética , Canales de Calcio Tipo R/metabolismo , Calcio/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Espinas Dendríticas/efectos de los fármacos , Péptidos/farmacología , Venenos de Araña/farmacología , Potenciales Sinápticos/efectos de los fármacos , Animales , Canales de Calcio Tipo R/deficiencia , Proteínas de Transporte de Catión/deficiencia , Espinas Dendríticas/metabolismo , Técnicas de Inactivación de Genes , Ratones , Transducción de Señal/efectos de los fármacos
12.
J Neurophysiol ; 97(5): 3660-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17376845

RESUMEN

Voltage-gated calcium channels are key components in the etiology and pathogenesis of epilepsies. Former studies mainly focused on P/Q-type Ca(v)2.1 and T-type Ca(v)3.2 Ca(2+) channels involved in absence epileptogenesis, but recent findings also point to an intriguing role of the Ca(v)2.3 E/R-type Ca(2+) channel in ictogenesis and seizure propagation. Based on the observation that Ca(v)2.3 is thought to induce plateau potentials in CA1 pyramidal cells, which can trigger epileptiform activity, our recent investigation revealed reduced PTZ-seizure susceptibility and altered seizure architecture in Ca(v)2.3(-/-) mice compared with controls. In the present study we tested hippocampal seizure susceptibility in Ca(v)2.3-deficient mice using surface and deep intrahippocampal telemetric EEG recordings as well as phenotypic seizure video analysis. Administration of kainic acid (30 mg/kg ip) revealed clear alteration in behavioral seizure architecture and dramatic resistance to limbic seizures in Ca(v)2.3(-/-) mice compared with controls, whereas no difference in hippocampal EEG seizure activity between both genotypes could be detected at this suprathreshold dosage. The same tendency was observed for NMDA seizure susceptibility (150 mg/kg ip) approaching the level of significance. In addition, histochemical analysis within the hippocampus revealed that excitotoxic effects after kainic acid administration are absent in Ca(v)2.3(-/-) mice, whereas Ca(v)2.3(+/+) animals exhibited clear and typical signs of excitotoxic cell death. These findings clearly indicate that the Ca(v)2.3 voltage-gated calcium channel plays a crucial role in both hippocampal ictogenesis and seizure generalization and is of central importance in neuronal degeneration after excitotoxic events.


Asunto(s)
Canales de Calcio Tipo R/deficiencia , Proteínas de Transporte de Catión/deficiencia , Hipocampo/fisiopatología , Neuronas/fisiología , Convulsiones/etiología , Convulsiones/patología , Animales , Electroencefalografía/métodos , Hipocampo/efectos de los fármacos , Ácido Kaínico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , N-Metilaspartato , Neuronas/efectos de los fármacos , Convulsiones/fisiopatología
13.
Proc Natl Acad Sci U S A ; 97(11): 6132-7, 2000 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-10801976

RESUMEN

alpha(1) subunit of the voltage-dependent Ca(2+) channel is essential for channel function and determines the functional specificity of various channel types. alpha(1E) subunit was originally identified as a neuron-specific one, but the physiological function of the Ca(2+) channel containing this subunit (alpha(1E) Ca(2+) channel) was not clear compared with other types of Ca(2+) channels because of the limited availability of specific blockers. To clarify the physiological roles of the alpha(1E) Ca(2+) channel, we have generated alpha(1E) mutant (alpha(1E)-/-) mice by gene targeting. The lacZ gene was inserted in-frame and used as a marker for alpha(1E) subunit expression. alpha(1E)-/- mice showed reduced spontaneous locomotor activities and signs of timidness, but other general behaviors were apparently normal. As involvement of alpha(1E) in pain transmission was suggested by localization analyses with 5-bromo-4-chloro-3-indolyl beta-d-galactopyranoside staining, we conducted several pain-related behavioral tests using the mutant mice. Although alpha(1E)+/- and alpha(1E)-/- mice exhibited normal pain behaviors against acute mechanical, thermal, and chemical stimuli, they both showed reduced responses to somatic inflammatory pain. alpha(1E)+/- mice showed reduced response to visceral inflammatory pain, whereas alpha(1E)-/- mice showed apparently normal response compared with that of wild-type mice. Furthermore, alpha(1E)-/- mice that had been presensitized with a visceral noxious conditioning stimulus showed increased responses to a somatic inflammatory pain, in marked contrast with the wild-type mice in which long-lasting effects of descending antinociceptive pathway were predominant. These results suggest that the alpha(1E) Ca(2 +) channel controls pain behaviors by both spinal and supraspinal mechanisms.


Asunto(s)
Canales de Calcio Tipo R/fisiología , Calcio/fisiología , Insensibilidad Congénita al Dolor/etiología , Dolor/fisiopatología , Ácido Acético/toxicidad , Animales , Ansiedad/genética , Canales de Calcio Tipo R/deficiencia , Canales de Calcio Tipo R/genética , Conducta Exploratoria , Miedo , Formaldehído/toxicidad , Expresión Génica , Inflamación/inducido químicamente , Inflamación/fisiopatología , Transporte Iónico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nociceptores/fisiopatología , Insensibilidad Congénita al Dolor/genética , Insensibilidad Congénita al Dolor/fisiopatología , Dimensión del Dolor , Peritonitis/inducido químicamente , Peritonitis/fisiopatología , Proteínas Recombinantes de Fusión/fisiología , Reflejo de Sobresalto/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
J Physiol ; 542(Pt 3): 699-710, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12154172

RESUMEN

Different subtypes of voltage-dependent Ca(2+) currents in native neurones are essential in coupling action potential firing to Ca(2+) influx. For most of these currents, the underlying Ca(2+) channel subunits have been identified on the basis of pharmacological and biophysical similarities. In contrast, the molecular basis of R-type Ca(2+) currents remains controversial. We have therefore examined the contribution of the Ca(V)2.3 (alpha(1E)) subunits to R-type currents in different types of central neurones using wild-type mice and mice in which the Ca(V)2.3 subunit gene was deleted. In hippocampal CA1 pyramidal cells and dentate granule neurones, as well as neocortical neurones of wild-type mice, Ca(2+) current components resistant to the combined application of omega-conotoxin GVIA and MVIIC, omega-agatoxin IVa and nifedipine (I(Ca,R)) were detected that were composed of a large R-type and a smaller T-type component. In Ca(V)2.3-deficient mice, I(Ca,R) was considerably reduced in CA1 neurones (79 %) and cortical neurones (87 %), with less reduction occurring in dentate granule neurones (47 %). Analysis of tail currents revealed that the reduction of I(Ca,R) is due to a selective reduction of the rapidly deactivating R-type current component in CA1 and cortical neurones. In all cell types, I(Ca,R) was highly sensitive to Ni(2+) (100 microM: 71-86 % block). A selective antagonist of cloned Ca(V)2.3 channels, the spider toxin SNX-482, partially inhibited I(Ca,R) at concentrations up to 300 nM in dentate granule cells and cortical neurones (50 and 57 % block, EC(50) 30 and 47 nM, respectively). I(Ca,R) in CA1 neurones was significantly less sensitive to SNX-482 (27 % block, 300 nM SNX-482). Taken together, our results show clearly that Ca(V)2.3 subunits underlie a significant fraction of I(Ca,R) in different types of central neurones. They also indicate that Ca(V)2.3 subunits may give rise to Ca(2+) currents with differing pharmacological properties in native neurones.


Asunto(s)
Canales de Calcio Tipo R/fisiología , Canales de Calcio Tipo T/fisiología , Hipocampo/metabolismo , Neocórtex/metabolismo , Neuronas/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo R/deficiencia , Canales de Calcio Tipo R/genética , Canales de Calcio Tipo T/efectos de los fármacos , Giro Dentado/metabolismo , Resistencia a Medicamentos , Conductividad Eléctrica , Electrofisiología , Proteínas de Transporte de Membrana , Ratones , Ratones Noqueados/genética , Células Piramidales/metabolismo , Valores de Referencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA