Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Brain Res ; 1764: 147438, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-33753067

RESUMO

Central sensitization (CS) is characteristic of difficult to treat painful conditions, such as fibromyalgia and neuropathies and have sexual dimorphism involved. The calcium influx in nociceptive neurons is a key trigger for CS and the role of Cav2.1 and Cav2.2 voltage gated calcium channels (VGCC) in this role were evidenced with the use of ω-agatoxin IVA and ω-agatoxin MVIIA blockers, respectively. However, the participation of the α1 subunit of the voltage-gated channel Cav2.3, which conducts R-type currents, in CS is unknown. Furthermore, the role of sexual differences in painful conditions is still poorly understood. Thus, we investigated the role of Cav2.3 in capsaicin-induced secondary hyperalgesia in mice, which serve as a CS model predictive of the efficacy of novel analgesic drugs. Capsaicin injection in C57BL/6 mice caused secondary hyperalgesia from one to five hours after injection, and the effects were similar in male and female mice. In female but not male mice, intrathecal treatment with the Cav2.3 inhibitor SNX-482 partially and briefly reversed secondary hyperalgesia at a dose (300 pmol/site) that did not cause adverse effects. Moreover, Cav2.3 expression in the dorsal root ganglia (DRG) and spinal cord was reduced by intrathecal treatment with an antisense oligonucleotide (ASO) targeting Cav2.3 in female and male mice. However, ASO treatment was able to provide a robust and durable prevention of secondary hyperalgesia caused by capsaicin in female mice, but not in male mice. Thus, our results demonstrate that Cav2.3 inhibition, especially in female mice, has a relevant impact on a model of CS. Our results provide a proof of concept for Cav2.3 as a molecular target. In addition, the result associated to the role of differences in painful conditions linked to sex opens a range of possibilities to be explored and needs more attention. Thus, the relevance of testing Cav2.3 inhibition or knockdown in clinically relevant pain models is needed.


Assuntos
Canais de Cálcio Tipo R/genética , Proteínas de Transporte de Cátions/genética , Sensibilização do Sistema Nervoso Central/genética , Hiperalgesia/genética , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo R/efeitos dos fármacos , Capsaicina , Proteínas de Transporte de Cátions/efeitos dos fármacos , Sensibilização do Sistema Nervoso Central/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Gânglios Espinais/metabolismo , Técnicas de Silenciamento de Genes , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligonucleotídeos Antissenso/farmacologia , Caracteres Sexuais , Venenos de Aranha/farmacologia , Medula Espinal/metabolismo
2.
J Neurosci ; 39(40): 7826-7839, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31420457

RESUMO

Dendritic excitability regulates how neurons integrate synaptic inputs and thereby influences neuronal output. As active dendritic events are associated with significant calcium influx they are likely to be modulated by calcium-dependent processes, such as calcium-activated potassium channels. Here we investigate the impact of small conductance calcium-activated potassium channels (SK channels) on dendritic excitability in male and female rat cortical pyramidal neurons in vitro and in vivo Using local applications of the SK channel antagonist apamin in vitro, we show that blocking somatic SK channels enhances action potential output, whereas blocking dendritic SK channels paradoxically reduces the generation of dendritic calcium spikes and associated somatic burst firing. Opposite effects were observed using the SK channel enhancer NS309. The effect of apamin on dendritic SK channels was occluded when R-type calcium channels were blocked, indicating that the inhibitory impact of apamin on dendritic calcium spikes involved R-type calcium channels. Comparable effects were observed in vivo Intracellular application of apamin via the somatic whole-cell recording pipette reduced the medium afterhyperpolarization and increased action potential output during UP states. In contrast, extracellular application of apamin to the cortical surface to block dendritic SK channels shifted the distribution of action potentials within UP states from an initial burst to a more distributed firing pattern, while having no impact on overall action potential firing frequency or UP and DOWN states. These data indicate that somatic and dendritic SK channels have opposite effects on neuronal excitability, with dendritic SK channels counter-intuitively promoting rather than suppressing neuronal output.SIGNIFICANCE STATEMENT Neurons typically receive input from other neurons onto processes called dendrites, and use electrical events such as action potentials for signaling. As electrical events in neurons are usually associated with calcium influx they can be regulated by calcium-dependent processes. One such process is through the activation of calcium-dependent potassium channels, which usually act to reduce action potential signaling. Although this is the case for calcium-dependent potassium channels found at the cell body, we show here that calcium-dependent potassium channels in dendrites of cortical pyramidal neurons counter-intuitively promote rather than suppress action potential output.


Assuntos
Dendritos/fisiologia , Células Piramidais/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Animais , Apamina/farmacologia , Canais de Cálcio Tipo R/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Dendritos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Feminino , Indóis/farmacologia , Masculino , Oximas/farmacologia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar
3.
ACS Chem Neurosci ; 10(6): 2939-2955, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-30946560

RESUMO

No universally efficacious therapy exists for chronic pain, a disease affecting one-fifth of the global population. An overreliance on the prescription of opioids for chronic pain despite their poor ability to improve function has led to a national opioid crisis. In 2018, the NIH launched a Helping to End Addiction Long-term plan to spur discovery and validation of novel targets and mechanisms to develop alternative nonaddictive treatment options. Phytochemicals with medicinal properties have long been used for various treatments worldwide. The natural product physalin F, isolated from the Physalis acutifolia (family: Solanaceae) herb, demonstrated antinociceptive effects in models of inflammatory pain, consistent with earlier reports of its anti-inflammatory and immunomodulatory activities. However, the target of action of physalin F remained unknown. Here, using whole-cell and slice electrophysiology, competition binding assays, and experimental models of neuropathic pain, we uncovered a molecular target for physalin F's antinociceptive actions. We found that physalin F (i) blocks CaV2.3 (R-type) and CaV2.2 (N-type) voltage-gated calcium channels in dorsal root ganglion (DRG) neurons, (ii) does not affect CaV3 (T-type) voltage-gated calcium channels or voltage-gated sodium or potassium channels, (iii) does not bind G-protein coupled opioid receptors, (iv) inhibits the frequency of spontaneous excitatory postsynaptic currents (EPSCs) in spinal cord slices, and (v) reverses tactile hypersensitivity in models of paclitaxel-induced peripheral neuropathy and spinal nerve ligation. Identifying CaV2.2 as a molecular target of physalin F may spur its use as a tool for mechanistic studies and position it as a structural template for future synthetic compounds.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo N/efeitos dos fármacos , Canais de Cálcio Tipo R/efeitos dos fármacos , Proteínas de Transporte de Cátions/efeitos dos fármacos , Neuralgia/metabolismo , Secoesteroides/farmacologia , Analgésicos/farmacologia , Animais , Proteínas de Transporte de Cátions/antagonistas & inibidores , Gânglios Espinais/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley
4.
J Neurochem ; 147(3): 310-322, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29972687

RESUMO

Kainic acid (KA) is a potent agonist at non-N-methyl-D-aspartate (non-NMDA) ionotropic glutamate receptors and commonly used to induce seizures and excitotoxicity in animal models of human temporal lobe epilepsy. Among other factors, Cav 2.3 voltage-gated calcium channels have been implicated in the pathogenesis of KA-induced seizures. At physiologically relevant concentrations, endogenous trace metal ions (Cu2+ , Zn2+ ) occupy an allosteric binding site on the domain I gating module of these channels and interfere with voltage-dependent gating. Using whole-cell patch-clamp recordings in human embryonic kidney (HEK-293) cells stably transfected with human Cav 2.3d and ß3 -subunits, we identified a novel, glutamate receptor-independent mechanism by which KA can potently sensitize these channels. Our findings demonstrate that KA releases these channels from the tonic inhibition exerted by low nanomolar concentrations of Cu2+ and produces a hyperpolarizing shift in channel voltage-dependence by about 10 mV, thereby reconciling the effects of Cu2+ chelation with tricine. When tricine was used as a surrogate to study the receptor-independent action of KA in electroretinographic recordings from the isolated bovine retina, it selectively suppressed a late b-wave component, which we have previously shown to be enhanced by genetic or pharmacological ablation of Cav 2.3 channels. Although the pathophysiological relevance remains to be firmly established, we speculate that reversal of Cu2+ -induced allosteric suppression, presumably via formation of stable kainate-Cu2+ complexes, could contribute to the receptor-mediated excitatory effects of KA. In addition, we discuss experimental implications for the use of KA in vitro, with particular emphasis on the seemingly high incidence of trace metal contamination in common physiological solutions.


Assuntos
Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/metabolismo , Proteínas de Transporte de Cátions/efeitos dos fármacos , Proteínas de Transporte de Cátions/metabolismo , Cobre/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Ácido Caínico/farmacologia , Animais , Bovinos , Quelantes/farmacologia , Eletrorretinografia , Glicina/análogos & derivados , Glicina/farmacologia , Células HEK293 , Humanos , Técnicas de Patch-Clamp , Receptores de Glutamato/metabolismo , Retina/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Zinco/farmacologia
5.
Assay Drug Dev Technol ; 7(3): 266-80, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19530894

RESUMO

T-type voltage-gated Ca(2+) channels have been implicated in contributing to a broad variety of human disorders, including pain, epilepsy, sleep disturbances, cardiac arrhythmias, and certain types of cancer. However, potent and selective T-type Ca(2+) channel modulators are not yet available for clinical use. This may in part be due to their unique biophysical properties that have delayed the development of high-throughput screening (HTS) assays for identifying blockers. One notable challenge is that at the normal resting membrane potential (V(m)) of cell lines commonly utilized for drug screening purposes, T-type Ca(2+) channels are largely inactivated and thus cannot be supported by typical formats of functional HTS assays to both evoke and quantify the Ca(2+) channel signal. Here we describe a simple method that can successfully support a fluorescence-based functional assay for compounds that modulate T-type Ca(2+)channels. The assay functions by exploiting the pore-forming properties of gramicidin to control the cellular V(m) in advance of T-type Ca(2+) channel activation. Using selected ionic conditions in the presence of gramicidin, T-type Ca(2+) channels are converted from the unavailable, inactivated state to the available, resting state, where they can be subsequently activated by application of extracellular K(+). The fidelity of the assay has been pharmacologically characterized with sample T-type Ca(2+) channel blockers whose potency has been determined by conventional manual patch-clamp techniques. This method has the potential for applications in high-throughput fluorometric imaging plate reader (FLIPR(R), Molecular Devices, Sunnyvale, CA) formats with cell lines expressing either recombinant or endogenous T-type Ca(2+) channels.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Algoritmos , Bloqueadores dos Canais de Cálcio/síntese química , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/metabolismo , Canais de Cálcio Tipo T/metabolismo , Proteínas de Transporte de Cátions/efeitos dos fármacos , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , DNA Complementar/biossíntese , DNA Complementar/genética , Avaliação Pré-Clínica de Medicamentos , Eletrofisiologia , Gramicidina/farmacologia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Soluções Farmacêuticas , Espectrometria de Fluorescência
6.
J Comp Neurol ; 513(2): 188-96, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19127523

RESUMO

Molecular studies have revealed the presence of R-type voltage-gated Ca(2+) channels at pre- and postsynaptic regions; however, no evidence for the participation of these channels in transmitter release has been presented for the spinal cord. Here we characterize the effects of SNX-482, a selective R channel blocker, on the monosynaptic excitatory postsynaptic potentials (EPSPs) evoked in motoneurons by stimulation of dorsolateral funiculus (DLF) terminals in a slice preparation from the adult turtle spinal cord. SNX-482 inhibited neurotransmission in a dose-dependent manner, with an IC(50) of approximately 9 +/- 1 nM. The EPSP time course and membrane time constant of the motoneurons were not altered, suggesting a presynaptic mechanism. The toxin inhibited the residual component of the EPSPs recorded in the presence of N- and P/Q-type Ca(2+) channel blockers, strongly suggesting a role for the R channels in neurotransmission at the spinal cord DLF terminals. Consistently with this, RT-PCR analysis of turtle spinal cord segments revealed the expression of the Ca(V)2.3 pore-forming (alpha(1E)) subunit of R channels, whereas the use of anti-alpha(1E)-specific antibodies resulted in its localization in the DLF fibers as demonstrated by immunohistochemistry coupled with laser confocal microscopy.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo R/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Neurônios Motores/metabolismo , Inibição Neural/fisiologia , Venenos de Aranha/farmacologia , Medula Espinal/citologia , Transmissão Sináptica/fisiologia , Análise de Variância , Animais , Western Blotting , Bloqueadores dos Canais de Cálcio/administração & dosagem , Canais de Cálcio Tipo R/genética , Canais de Cálcio Tipo R/metabolismo , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Imuno-Histoquímica , Microscopia Confocal , Neurônios Motores/citologia , Neurônios Motores/efeitos dos fármacos , Inibição Neural/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Venenos de Aranha/administração & dosagem , Medula Espinal/anatomia & histologia , Transmissão Sináptica/efeitos dos fármacos , Fatores de Tempo , Tartarugas
7.
Auton Neurosci ; 146(1-2): 56-61, 2009 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-19162562

RESUMO

Release of acetylcholine (ACh) from preganglionic nerve terminals requires calcium entry through voltage-gated calcium channels. The calcium channel subtype required for ACh release varies depending on the particular ganglionic synapse. I have investigated the functional role of calcium channels in transmitter release from parasympathetic and sympathetic preganglionic terminals in pelvic and celiac ganglia of female mice. Single electrode voltage clamp was used to measure EPSC amplitude in the absence and presence of selective calcium channel antagonists. In pelvic ganglia omega- conotoxin GVIA, a selective N-type calcium channel antagonist, reduced the amplitude of EPSCs evoked by pelvic nerve stimulation by 46+/-5% (n=8, P=0.015). In contrast, in the celiac ganglion, omega- conotoxin GVIA had no effect on the amplitude of EPSCs evoked by splanchnic nerve stimulation (P=0.09, n=7). EPSCs in both pelvic and celiac ganglia were resistant to the P-type calcium channel antagonist agatoxin (50 nM, n=5 for both ganglia) and the R-type calcium channel antagonist SNX482 (100 nM, n=4 for both ganglia). These results indicate that in female mice, release of ACh in sympathetic pathways to prevertebral ganglia does not require calcium entry from N-type calcium channels. However, release of ACh from sacral parasympathetic preganglionic neurons requires calcium entry from both N-type and toxin-resistant calcium channels.


Assuntos
Fibras Autônomas Pré-Ganglionares/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo N/fisiologia , Sinalização do Cálcio/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Gânglios Simpáticos/efeitos dos fármacos , Plexo Hipogástrico/efeitos dos fármacos , ômega-Conotoxina GVIA/farmacologia , Acetilcolina/metabolismo , Animais , Fibras Autônomas Pré-Ganglionares/metabolismo , Fibras Autônomas Pré-Ganglionares/fisiologia , Canais de Cálcio Tipo P/efeitos dos fármacos , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Gânglios Simpáticos/fisiologia , Plexo Hipogástrico/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Venenos de Aranha/farmacologia , Nervos Esplâncnicos/fisiologia , ômega-Agatoxina IVA/farmacologia
8.
Cell Signal ; 20(12): 2286-95, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18793718

RESUMO

Somatostatin (SST) inhibits Ca(2+) entry into pancreatic B-cells via voltage-operated Ca(2+) channels (VOCCs) of L-type, leading to the suppression of insulin secretion. Activation of R-type channels increases insulin secretion. However, the role of R-type Ca(2+) channels (Ca(V)2.3) in mediating the effects of SST on insulin secretion has not been so far investigated. Here, we identify the SST-receptor subtypes (SSTR) expressed on insulin-producing INS-1 cells by RT-PCR and by functional assays. The role of R-type channels in regulating [Ca(2+)](i) in response to SST-treatment was detected by cell fluorescence imaging and patch-clamp technique. INS-1 expressed SSTR2 and SSTR3 and agonists (ag.) selective for these receptors reduced 10 nM exendin-4/20 mM glucose-stimulated insulin secretion. Surprisingly, SST and SST2-ag. transiently increased [Ca(2+)](i). Subsequently, these agonists led to a decrease in [Ca(2+)](i) below the basal levels. In contrast, SST3-ag. failed to induce a transient peak of [Ca(2+)](i). Instead, a persistent minor suppression of [Ca(2+)](i) was detected from 25 min. R-type channel blocker SNX-482 altered [Ca(2+)](i) in SST- and SST2-ag.-treated cells. Notably, the inhibition of insulin secretion by SST and SST2-ag., but not SST3-ag. was attenuated by SNX-482. Taken together, SST and SSTR2 regulate [Ca(2+)](i) and insulin secretion in INS-1 cells via R-type channels. In contrast, the R-type calcium channel does not mediate the effects of SST3-ag. on insulin secretion. We conclude that R-type channels play a major role in the inhibition of insulin secretion by somatostatin in INS-1 cells.


Assuntos
Canais de Cálcio Tipo R/fisiologia , Insulina/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Animais , Sequência de Bases , Cálcio/metabolismo , Canais de Cálcio Tipo R/efeitos dos fármacos , Fenômenos Eletrofisiológicos , RNA Mensageiro/metabolismo , Ratos , Receptores de Somatostatina/agonistas , Espectrometria de Fluorescência , Venenos de Aranha/farmacologia , Fatores de Tempo
9.
Acta Neurochir Suppl ; 104: 99-102, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18456998

RESUMO

Voltage-dependent potassium (Kv) and calcium (VDCC) channels play an important role in the regulation of membrane potential and intracellular calcium concentration in cerebral artery myocytes. Recent evidence suggests VDCC activity is increased and Kv channel activity is decreased in cerebral arteries following subarachnoid hemorrhage (SAH), promoting enhanced constriction. We have examined the impact of the blood component oxyhemoglobin on Kv and VDCC function in small (100-200 microm) diameter cerebral arteries. Acute (10 min) exposure of oxyhemoglobin caused cerebral artery constriction and Kv current suppression that was abolished by tyrosine kinase inhibitors and a Kv channel blocker. Although short-term oxyhemoglobin application did not directly alter VDCC activity, five-day exposure to oxyhemoglobin was associated with enhanced expression of voltage-dependent calcium channels. This work suggests that acute and chronic effects of oxyhemoglobin act synergistically to promote membrane depolarization and increased VDCC activity in cerebral arteries. These actions of oxyhemoglobin may contribute to the development of cerebral vasospasm following aneurysmal subarachnoid hemorrhage.


Assuntos
Artérias Cerebrais/fisiologia , Canais Iônicos/fisiologia , Oxiemoglobinas/farmacologia , Animais , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/fisiologia , Artérias Cerebrais/efeitos dos fármacos , Canais Iônicos/efeitos dos fármacos , Modelos Animais , Técnicas de Cultura de Órgãos , Coelhos , Vasoconstrição/efeitos dos fármacos
10.
Brain Res ; 1210: 1-10, 2008 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-18420182

RESUMO

Protein kinase C (PKC) is implicated in the potentiation of Ca v 2.3 currents by acetyl-beta-methylcholine (MCh), a muscarinic M1 receptor agonist or phorbol-12-myristate, 13-acetate (PMA). The PKC isozymes responsible for the action of MCh and PMA were investigated using translocation as a measure of activation and with isozyme-selective antagonists and siRNA. Ca v channels were expressed with alpha1 2.3, beta1b and alpha2delta subunits and muscarinic M1 receptors in the Xenopus oocytes and the expressed currents (I Ba) were studied using Ba2+ as the charge carrier. Translocation of PKC isozymes to the membrane studied by Western blot revealed that all eleven known PKC isozymes are present in the Xenopus oocytes. Exposure of the oocytes to MCh led to the translocation of PKC alpha whereas PMA activated PKC betaII and epsilon isozymes. The action of MCh was inhibited by Go 6976, an inhibitor of cPKC isozymes or PKC alpha siRNA. PMA-induced potentiation of Ca v 2.3 currents was inhibited by CG533 53, a PKC betaII antagonist, betaIIV5.3, a peptide translocation inhibitor of PKC betaII or PKC betaII siRNA. Similarly, epsilonV1.2, a peptide translocation inhibitor of PKC epsilon or PKC epsilon siRNA inhibited PMA action. The inhibitors of PKC increased the basal I Ba slightly. It is possible that some PKC isozymes have negative control over the I Ba. Our results implicate PKC alpha in the potentiation of Ca v 2.3 currents by MCh and PKC betaII and epsilon in the potentiation of Ca v 2.3 currents by PMA.


Assuntos
Canais de Cálcio/efeitos dos fármacos , Cloreto de Metacolina/farmacologia , Proteína Quinase C/efeitos dos fármacos , Receptor Muscarínico M1/agonistas , Acetato de Tetradecanoilforbol/farmacologia , Animais , Canais de Cálcio/metabolismo , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/metabolismo , Carcinógenos/farmacologia , Proteínas de Transporte de Cátions/efeitos dos fármacos , Proteínas de Transporte de Cátions/metabolismo , Membrana Celular/enzimologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Inibidores Enzimáticos/farmacologia , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Isoenzimas/efeitos dos fármacos , Isoenzimas/genética , Isoenzimas/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Agonistas Muscarínicos/farmacologia , Oócitos , Técnicas de Patch-Clamp , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Transporte Proteico/genética , RNA Interferente Pequeno/genética , Receptor Muscarínico M1/metabolismo , Xenopus laevis
11.
Neuroreport ; 19(3): 299-303, 2008 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-18303570

RESUMO

In Purkinje cells from cerebellar slice cultures, low-threshold Ca spike (LTS) gives rise to complex bursts in the soma that resemble the complex spike induced by climbing fibers stimulation. We show that LTS is reduced by T-type and R-type Ca channel blockers (SNX-482, nickel, or mibefradil). We propose that LTS is generated by openings of T-type Ca channels (alpha-1G and/or alpha-1I subunits) and R-type Ca channels (alpha-1E subunit isoforms with a weak sensitivity to SNX-482 and to nickel). Using mibefradil we show that climbing fiber stimulation activates LTS, which contributes to the shape of the response. This Ca entry may be involved in Ca-dependent synaptic plasticity of the parallel fiber input induced by climbing fiber activation.


Assuntos
Sinalização do Cálcio/fisiologia , Células de Purkinje/fisiologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo R/efeitos dos fármacos , Eletrofisiologia , Ácido Glutâmico/metabolismo , Mibefradil/farmacologia , Fibras Nervosas/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Níquel/farmacologia , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Estimulação Luminosa , Ratos , Venenos de Aranha/farmacologia
12.
J Dent Res ; 87(2): 137-41, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18218839

RESUMO

Eugenol, a natural congener of capsaicin, is a routine analgesic agent in dentistry. We have recently demonstrated the inhibition of Ca(V)2.2 calcium channel and sodium channel currents to be molecular mechanisms underlying the analgesic effect of eugenol. We hypothesized that Ca(V)2.3 channels are also modulated by eugenol and investigated its mode of action using the whole-cell patch-clamp technique in a heterologous expression system. Eugenol inhibited calcium currents in the E52 cell line, stably expressing the human Ca(V)2.3 calcium channels, where TRPV1 is not endogenously expressed. The extent of current inhibition was not significantly different between naïve E52 cells and TRPV1-expressing E52 cells, suggesting no involvement of TRPV1. In contrast, TRPV1 activation is prerequisite for the inhibition of Ca(V)2.3 calcium channels by capsaicin. The results indicate that eugenol has mechanisms distinct from those of capsaicin for modulating Ca(V)2.3 channels. We suggest that inhibition of Ca(V)2.3 channels by eugenol might contribute to its analgesic effect.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo R/efeitos dos fármacos , Proteínas de Transporte de Cátions/efeitos dos fármacos , Materiais Dentários/farmacologia , Eugenol/farmacologia , Analgésicos/farmacologia , Capsaicina/farmacologia , Linhagem Celular , Humanos , Técnicas de Patch-Clamp , Plasmídeos , Fármacos do Sistema Sensorial/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Cátion TRPV/genética , Transfecção
13.
Neurogastroenterol Motil ; 19(10): 845-55, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17883436

RESUMO

Alpha2-adrenoceptors inhibit Ca2+ influx through voltage-gated Ca2+ channels throughout the nervous system and Ca2+ channel function is modulated following activation of some G-protein coupled receptors. We studied the specific Ca2+ channel inhibited following alpha2-adrenoceptor activation in guinea-pig small intestinal myenteric neurons. Ca2+ currents (I(Ca2+)) were studied using whole-cell patch-clamp techniques. Changes in intracellular Ca2+ (delta[Ca2+]i) in nerve cell bodies and varicosities were studied using digital imaging where Ca2+ influx was evoked by KCl (60 mmol L(-1)) depolarization. The alpha2-adrenoceptor agonist, UK 14 304 (0.01-1 micromol L(-1)) inhibited I(Ca2+) and delta[Ca2+]i; maximum inhibition of I(Ca2+) was 40%. UK 14 304 did not affect I(Ca2+) in the presence of SNX-482 or NiCl2 (R-type Ca2+ channel antagonists). UK 14 304 inhibited I(Ca2+) in the presence of nifedipine, omega-agatoxin IVA or omega-conotoxin, inhibitors of L-, P/Q- and N-type Ca2+ channels. UK 14 304 induced inhibition of I(Ca2+) was blocked by pertussis toxin pretreatment (1 microg mL(-1) for 2 h). Alpha2-adrenoceptors couple to inhibition of R-type Ca2+ channels via a pertussis toxin-sensitive pathway in myenteric neurons. R-type channels may be a target for the inhibitory actions of noradrenaline released from sympathetic nerves on to myenteric neurons.


Assuntos
Canais de Cálcio Tipo R/metabolismo , Intestino Delgado/fisiologia , Plexo Mientérico/fisiologia , Neurônios/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo R/efeitos dos fármacos , Células Cultivadas , Cobaias , Intestino Delgado/inervação , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Plexo Mientérico/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Receptores Adrenérgicos alfa 2/efeitos dos fármacos
14.
J Pharmacol Exp Ther ; 319(3): 1009-20, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16982704

RESUMO

The mutation in the alpha(1A) subunit gene of the P/Q-type (Ca(v)2.1) Ca(2+) channel present in tottering (tg) mice causes ataxia and motor seizures that resemble absence epilepsy in humans. P/Q-type Ca(2+)channels are primarily involved in acetylcholine (ACh) release at mammalian neuromuscular junctions. Unmasking of L-type (Ca(v)1.1-1.2) Ca(2+) channels occurs in cerebellar Purkinje cells of tg mice. However, whether L-type Ca(2+) channels are also up-regulated at neuromuscular junctions of tg mice is unknown. We characterized thoroughly the pharmacological sensitivity of the Ca(2+) channels, which control ACh release at adult tg neuromuscular junctions. Block of N- and R-type (Ca(v)2.2-2.3), but not L-type Ca(2+) channels, significantly reduced quantal content of end-plate potentials in tg preparations. Neither resting nor KCl-evoked miniature end-plate potential frequency differed significantly between tg and wild type (WT). Immunolabeling of Ca(2+) channel subunits alpha(1A), alpha(1B), alpha(1C), and alpha(1E) revealed an apparent increase of alpha(1B), and alpha(1E) staining, at tg but not WT neuromuscular junctions. This presumably compensates for the deficit of P/Q-type Ca(2+)channels, which localized presynaptically at WT neuromuscular junctions. No alpha(1C) subunits juxtaposed with pre- or postsynaptic markers at either WT or tg neuromuscular junctions. Thus, in adult tg mice, immunocytochemical and electrophysiological data indicate that N- and R-type channels both assume control of ACh release at motor nerve terminals. Recruitment of alternate subtypes of Ca(2+) channels to control transmitter release seems to represent a commonly occurring method of neuronal plasticity. However, it is unclear which conditions underlie recruitment of Ca(v)2 as opposed to Ca(v)1-type Ca(2+) channels.


Assuntos
Acetilcolina/metabolismo , Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo R/fisiologia , Junção Neuromuscular/fisiologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo N/efeitos dos fármacos , Canais de Cálcio Tipo R/efeitos dos fármacos , Eletrofisiologia , Imuno-Histoquímica , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes Neurológicos , Microscopia Confocal , Placa Motora/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , Junção Neuromuscular/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Cloreto de Potássio/farmacologia , ômega-Agatoxina IVA/farmacologia , ômega-Conotoxinas/farmacologia
15.
J Neurosci ; 26(23): 6249-58, 2006 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-16763032

RESUMO

The "toxin-resistant" R-type Ca2+ channels are expressed widely in the CNS and distributed mainly in apical dendrites and spines. They play important roles in regulating signal transduction and intrinsic properties of neurons, but the modulation of these channels in the mammalian CNS has not been studied. In this study we used whole-cell patch-clamp recordings and found that muscarinic activation enhances R-type, but does not affect T-type, Ca2+ currents in hippocampal CA1 pyramidal neurons after N, P/Q, and L-type Ca2+ currents selectively were blocked. M1/M3 cholinergic receptors mediated the muscarinic stimulation of R-type Ca2+ channels. The signaling pathway underlying the R-type enhancement was independent of intracellular [Ca2+] changes and required the activation of a Ca(2+)-independent PKC pathway. Furthermore, we found that the enhancement of R-type Ca2+ currents resulted in the de novo appearance of Ca2+ spikes and in remarkable changes in the firing pattern of R-type Ca2+ spikes, which could fire repetitively in the theta frequency. Therefore, muscarinic enhancement of R-type Ca2+ channels could play an important role in modifying the dendritic response to synaptic inputs and in the intrinsic resonance properties of neurons.


Assuntos
Canais de Cálcio Tipo R/fisiologia , Hipocampo/metabolismo , Muscarina/metabolismo , Células Piramidais/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo T/efeitos dos fármacos , Canais de Cálcio Tipo T/fisiologia , Carbacol/farmacologia , Colinérgicos/farmacologia , Condutividade Elétrica , Eletrofisiologia , Hipocampo/citologia , Técnicas In Vitro , Técnicas de Patch-Clamp , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Muscarínico M1/fisiologia , Receptor Muscarínico M3/fisiologia , Transdução de Sinais/fisiologia , Ritmo Teta
16.
J Neurophysiol ; 96(3): 1267-77, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16738209

RESUMO

Endocannabinoids released by postsynaptic cells inhibit neurotransmitter release in many central synapses by activating presynaptic cannabinoid CB1 receptors. In particular, in the cerebellum, endocannabinoids inhibit synaptic transmission at granule cell to Purkinje cell synapses by modulating presynaptic calcium influx via N-, P/Q-, and R-type calcium channels. Using whole cell patch-clamp techniques, we show that in addition to this presynaptic action, both synthetic and endogenous cannabinoids inhibit P-type calcium currents in isolated rat Purkinje neurons independent of CB1 receptor activation. The IC50 for the anandamide (AEA)-induced inhibition of P-current peak amplitude was 1.04 +/- 0.04 microM. In addition, we demonstrate that all the tested cannabinoids in a physiologically relevant range of concentrations strongly accelerate inactivation of P currents. The effects of AEA cannot be attributed to the metabolism of AEA because a nonhydrolyzing analogue of AEA, methanandamide inhibited P-type currents with a similar efficacy. All effects of cannabinoids on P-type Ca2+ currents were insensitive to antagonists of CB1 cannabinoid or vanilloid TRPV1 receptors. In cerebellar slices, WIN 55,212-2 significantly affected spontaneous firing of Purkinje neurons in the presence of CB1 receptor antagonist, in a manner similar to that of a specific P-type channel antagonist, indicating a possible functional implication of the direct effects of cannabinoids on P current. Taken together these findings demonstrate a functionally important direct action of cannabinoids on P-type calcium currents.


Assuntos
Canais de Cálcio Tipo P/fisiologia , Canabinoides/farmacologia , Células de Purkinje/fisiologia , Animais , Ácidos Araquidônicos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo P/efeitos dos fármacos , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/fisiologia , Cerebelo/fisiologia , Endocanabinoides , Técnicas In Vitro , Cinética , Técnicas de Patch-Clamp , Alcamidas Poli-Insaturadas , Células de Purkinje/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Cátion TRPV/fisiologia
17.
J Neuroendocrinol ; 17(9): 583-90, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16101897

RESUMO

Oxytocin release from neurophypophysial terminals is particularly sensitive to inhibition by the micro-opioid receptor agonist, DAMGO. Because the R-type component of the neurophypophysial terminal Ca2+ current (ICa) mediates exclusively oxytocin release, we hypothesised that micro-opioids could preferentially inhibit oxytocin release by blocking this channel subtype. Whole-terminal recordings showed that DAMGO and the R-type selective blocker SNX-482 inhibit a similar ICa component. Measurements of [Ca2+]i levels and oxytocin release confirmed that the effects of DAMGO and SNX-482 are not additive. Finally, isolation of the R-type component and its associated rise in [Ca2+]i and oxytocin release allowed us to demonstrate the selective inhibition by DAMGO of this channel subtype. Thus, micro-opioid agonists modulate specifically oxytocin release in neurophypophysial terminals by selectively targeting R-type Ca2+ channels. Modulation of Ca2+ channel subtypes could be a general mechanism for drugs of abuse to regulate the release of specific neurotransmitters at central nervous system synapses.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo R/efeitos dos fármacos , Ocitocina/metabolismo , Neuro-Hipófise/metabolismo , Receptores Opioides mu/agonistas , Analgésicos Opioides/farmacologia , Animais , Arginina Vasopressina/metabolismo , Cálcio/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Terminações Nervosas/efeitos dos fármacos , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Venenos de Aranha/farmacologia
18.
Epilepsia ; 46(4): 481-9, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15816941

RESUMO

PURPOSE: Cholinergic-dependent plateau potentials (PPs) are intrinsically generated conductances that can elicit ictal-type seizure activity. The aim of this study was to investigate the actions of topiramate (TPM) on the generation of PPs. METHODS: We used whole-cell patch-clamp recordings from CA1 pyramidal neurons in rat hippocampal slices to examine the effects of TPM on the PPs. RESULTS: In current-clamp mode, action potentials evoked PPs after cholinergic receptor stimulation. Therapeutically relevant concentrations of TPM (50 microM) depressed the PPs evoked by action potentials. Surprisingly, in voltage-clamp mode, we discovered that the cyclic nucleotide-gated (CNG) current that underlies PP generation (denoted as I(tail)) was not depressed. However, significantly longer depolarizing voltage steps were required to elicit I(tail). This suggested that the calcium entry trigger for evoking PPs was depressed by TPM and not I(tail) itself. TPM had no effect on calcium spikes in control conditions; however, TPM did reduce calcium spikes after cholinergic-receptor stimulation. We recently found that R-type calcium spikes are enhanced by cholinergic-receptor stimulation. Therefore we isolated R-type calcium spikes with a cocktail containing tetrodotoxin, omega-conotoxin MVIIC, omega-conotoxin-GVIA, omega-agatoxin IVA, and nifedipine. R-type calcium spikes were significantly depressed by TPM. We also examined the effects of TPM on recombinant Ca(V)2.3 calcium channels expressed in tsA-201 cells. TPM depressed currents mediated by Ca(V)2.3 subunits by a hyperpolarizing shift in steady-state inactivation. CONCLUSIONS: We have found that TPM reduces ictal-like activity in CA1 hippocampal neurons through a novel inhibitory action of R-type calcium channels.


Assuntos
Anticonvulsivantes/farmacologia , Canais de Cálcio Tipo R/efeitos dos fármacos , Frutose/análogos & derivados , Frutose/farmacologia , Hipocampo/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo R/fisiologia , Carbacol/farmacologia , Células Cultivadas , Hipocampo/citologia , Hipocampo/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Células Piramidais/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Muscarínicos/efeitos dos fármacos , Receptores Muscarínicos/fisiologia , Convulsões/fisiopatologia , Convulsões/prevenção & controle , Topiramato , Transfecção
19.
Circ Res ; 96(4): 419-26, 2005 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-15692089

RESUMO

Cerebral aneurysm rupture and subarachnoid hemorrhage (SAH) inflict disability and death on thousands of individuals each year. In addition to vasospasm in large diameter arteries, enhanced constriction of resistance arteries within the cerebral vasculature may contribute to decreased cerebral blood flow and the development of delayed neurological deficits after SAH. In this study, we provide novel evidence that SAH leads to enhanced Ca2+ entry in myocytes of small diameter cerebral arteries through the emergence of R-type voltage-dependent Ca2+ channels (VDCCs) encoded by the gene CaV 2.3. Using in vitro diameter measurements and patch clamp electrophysiology, we have found that L-type VDCC antagonists abolish cerebral artery constriction and block VDCC currents in cerebral artery myocytes from healthy animals. However, 5 days after the intracisternal injection of blood into rabbits to mimic SAH, cerebral artery constriction and VDCC currents were enhanced and partially resistant to L-type VDCC blockers. Further, SNX-482, a blocker of R-type Ca2+ channels, reduced constriction and membrane currents in cerebral arteries from SAH animals, but was without effect on cerebral arteries of healthy animals. Consistent with our biophysical and functional data, cerebral arteries from healthy animals were found to express only L-type VDCCs (CaV 1.2), whereas after SAH, cerebral arteries were found to express both CaV 1.2 and CaV 2.3. We propose that R-type VDCCs may contribute to enhanced cerebral artery constriction after SAH and may represent a novel therapeutic target in the treatment of neurological deficits after SAH.


Assuntos
Canais de Cálcio Tipo R/fisiologia , Artérias Cerebrais/fisiopatologia , Hemorragia Subaracnóidea/fisiopatologia , Vasoconstrição/fisiologia , Vasoespasmo Intracraniano/fisiopatologia , Animais , Sangue , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio Tipo R/efeitos dos fármacos , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/metabolismo , Artérias Cerebrais/patologia , Cisterna Magna , Di-Hidropiridinas/farmacologia , Diltiazem/farmacologia , Modelos Animais de Doenças , Resistência a Medicamentos , Injeções , Transporte de Íons/efeitos dos fármacos , Masculino , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Nifedipino/farmacologia , Técnicas de Patch-Clamp , Coelhos , Venenos de Aranha/farmacologia , Hemorragia Subaracnóidea/etiologia , Vasoconstrição/efeitos dos fármacos , Vasoespasmo Intracraniano/etiologia , ômega-Agatoxina IVA/farmacologia , ômega-Conotoxina GVIA/farmacologia
20.
Brain Res ; 1036(1-2): 50-9, 2005 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-15725401

RESUMO

By using whole-cell recordings in acute and organotypic hypothalamic slices, we found that following K+ channel blockade, sustained plateau potentials can be elicited by current injection in suprachiasmatic neurons. In an attempt to determine the ionic basis of these potentials, ion-substitution experiments were carried out. It appeared that to generate plateau potentials, calcium influx was required. Plateau potentials were also present when extracellular calcium was replaced by barium, but were independent upon an increase in the intracellular free calcium concentration. Substitution of extracellular sodium by the impermeant cation N-methyl-D-glucamine indicated that sodium influx could also contribute to plateau potentials. To gain some information on the pharmacological profile of the Ca++ channels responsible for plateau potentials, selective blocker of various types of Ca++ channel were tested. Plateau potentials were unaffected by isradipine, an L-type Ca++ channel blocker. However, they were slightly reduced by omega-conotoxin GVIA and omega-agatoxin TK, blockers of N-type and P/Q-type Ca++ channels, respectively. These data suggest that R-type Ca++ channels probably play a major role in the genesis of plateau potentials. We speculate that neurotransmitters/neuromodulators capable of reducing or suppressing potassium conductance(s) may elicit a Ca++-dependent plateau potential in suprachiasmatic neurons, thus promoting sustained firing activity and neuropeptide release.


Assuntos
Canais de Cálcio/fisiologia , Sinalização do Cálcio/fisiologia , Neurônios/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/fisiologia , Núcleo Supraquiasmático/metabolismo , Animais , Bário/metabolismo , Bário/farmacologia , Cálcio/metabolismo , Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Quelantes/farmacologia , Estimulação Elétrica , Líquido Extracelular/metabolismo , Magnésio/metabolismo , Magnésio/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo , Sódio/farmacologia , Núcleo Supraquiasmático/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA