Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
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.
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
3.
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
4.
Neuroendocrinology ; 82(2): 87-102, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16424676

RESUMO

Insulin-like growth factor 1 (IGF-1) is a potent mitogenic and secretory factor that acts on voltage operated Ca(2+) channels (VOCCs). VOCCs are categorized into L-type channels (Ca(V)1.1-1.4), P/Q-type channels (Ca(V)2.1), N-type channels (Ca(V)2.2), R-type channels (Ca(V)2.3), and T-type channels (Ca(V)3.1-3.3). Aside from regulating membrane excitability, VOCCs influence chromogranin A (CgA) secretion in neuroendocrine tumor (NET) cells. It is not known, whether VOCCs play a role in the IGF-1-dependent regulation of CgA secretion in NET cells. We therefore studied the effects of IGF-1 on individual VOCC subtypes and characterized their role in mediating IGF-1-dependent regulation of CgA secretion in NET cells. Using specific modulators of VOCC subtypes, we identified the functional expression of L-, N-, P/Q- and R-type channels in primary as well as permanent models of NET. The IGF-1-induced intracellular Ca(2+) increase in NET cells was mainly due to the activation of R-type channel activity. The effects on intracellular calcium, observed in whole-cell patch-clamp recordings and fluorescence imaging, were partially blocked by the specific R-type channel blocker SNX-482 and antisense oligonucleotides against the alpha(1) subunit of this channel. IGF-1 potently induced CgA secretion. The effect of IGF-1 was reduced by both, inhibition of R-type channel activity and a reduction of R-type channel expression using antisense oligonucleotides. Since R-type channels exist in NET cells and couple to both, IGF-1 receptor signaling as well as CgA secretion, pharmacological interference with R-type channels may represent a new therapeutic option by blocking Ca(2+) signaling thereby abrogating IGF-1-dependent hypersecretion in NET disease.


Assuntos
Canais de Cálcio Tipo R/metabolismo , Cálcio/metabolismo , Cromograninas/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Biomarcadores Tumorais/metabolismo , Canais de Cálcio Tipo R/efeitos dos fármacos , Linhagem Celular Tumoral , Cromogranina A , Meios de Cultura , Eletrofisiologia , Imunofluorescência , Corantes Fluorescentes , Fura-2 , Humanos , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Patch-Clamp , Fenótipo
5.
J Pharmacol Sci ; 95(2): 240-7, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15215649

RESUMO

We examined if a range of carbonic anhydrase inhibitors (CAIs) interacted with the high-voltage activated voltage-sensitive calcium channels (VSCCs) encoded by the human alpha(1E) subunit. Whole-cell recordings were made from HEK293 cells stably expressing human alpha(1E)beta(3)-mediated calcium channels. SNX-482 (an alpha(1E) inhibitor) blocked alpha(1E)-mediated VSCCs with an IC(50) close to 10 nM. The anticonvulsant CAI ethoxyzolamide also inhibited these currents, with an IC(50) close to 1 microM, and produced an accompanying 20-mV hyperpolarizing shift in the steady-state inactivation profile. Other structurally diverse CAIs (e.g., acetazolamide and benzolamide) produced approximately 30 - 40% inhibition of alpha(1E)beta(3)-mediated Ca(2+) currents at 10 microM. Topiramate (10 microM), an anticonvulsant with CAI activity, inhibited these currents by 68 +/- 7%. This off-target activity of CAIs at VSCCs may contribute to some of the effects they produce both in vitro and in vivo.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Inibidores da Anidrase Carbônica/farmacologia , Proteínas de Transporte de Cátions/metabolismo , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/metabolismo , Linhagem Celular Tumoral , Eletrofisiologia , Etoxzolamida/farmacologia , Humanos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Proteínas Recombinantes , Venenos de Aranha/farmacologia
6.
Brain Res ; 992(2): 272-80, 2003 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-14625066

RESUMO

Elimination of adenosine by addition of adenosine deaminase (ADA) to the media leads to alterations in intracellular free calcium concentration ([Ca(2+)](i)) in cerebellar granular cells. Adenosine deaminase brings about increases or decreases in [Ca(2+)](i) depending on the previous activation state of the cell. These effects are dependent on the catalytic activity of adenosine deaminase, since its previous catalytic inactivation with Hg(2+) prevents the above-mentioned changes in intracellular calcium. Extracellular calcium is required for the increase in [Ca(2+)](i) promoted by ADA. This rise is insensitive to thapsigargin, but sensitive to micromolar concentrations of Ni(2+). Toxins specific for L, N and P/Q calcium channels do not overtly reduce this effect. N(6)-Cyclopentyl adenosine (CPA), an A(1) receptor agonist, produces a partial reversion of ADA effects, while CGS21680, A(2A)/A(2B) receptor agonist, slightly enhances them. Expression of A(1), A(2A), A(2B) and A(3) adenosine receptor mRNAs was detected in cerebellar granular cell cultures. These results suggest that adenosine modulate [Ca(2+)](i) in cerebellar granule cells through different adenosine receptor subtypes which, at least in part, seem to act through R-type calcium channels.


Assuntos
Adenosina/análogos & derivados , Adenosina/deficiência , Sinalização do Cálcio/fisiologia , Córtex Cerebelar/metabolismo , Neurônios/metabolismo , Receptores Purinérgicos P1/metabolismo , Adenosina/metabolismo , Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina , Adenosina Desaminase/farmacologia , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Córtex Cerebelar/citologia , Córtex Cerebelar/efeitos dos fármacos , Líquido Intracelular/efeitos dos fármacos , Líquido Intracelular/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Níquel/farmacologia , Fenetilaminas/farmacologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/metabolismo , Receptores Purinérgicos P1/efeitos dos fármacos , Receptores Purinérgicos P1/genética
7.
Br J Pharmacol ; 136(8): 1135-45, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12163346

RESUMO

1 Acetylcholine release at the neuromuscular junction relies on rapid, local and transient calcium increase at presynaptic active zones, triggered by the ion influx through voltage-dependent calcium channels (VDCCs) clustered on the presynaptic membrane. Pharmacological investigation of the role of different VDCC subtypes (L-, N-, P/Q- and R-type) in spontaneous and evoked acetylcholine (ACh) release was carried out in adult mouse neuromuscular junctions (NMJs) under normal and pathological conditions. 2 omega-Agatoxin IVA (500 nM), a specific P/Q-type VDCC blocker, abolished end plate potentials (EPPs) in normal NMJs. However, when neurotransmitter release was potentiated by the presence of the K(+) channel blocker 4-aminopyridine (4-AP), an omega-agatoxin IVA- and omega-conotoxin MVIIC-resistant component was detected. This resistant component was only partially sensitive to 1 micro M omega-conotoxin GVIA (N-type VDCC blocker), but insensitive to any other known VDCC blockers. Spontaneous release was dependent only on P/Q-type VDCC in normal NMJs. However, in the presence of 4-AP, it relied on L-type VDCCs too. 3 ACh release from normal NMJs was compared with that of NMJs of mice passively injected with IgGs obtained from patients with Lambert-Eaton myasthenic syndrome (LEMS), a disorder characterized by a compromised neurotransmitter release. Differently from normal NMJs, in LEMS IgGs-treated NMJs an omega-agatoxin IVA-resistant EPP component was detected, which was only partially blocked by calciseptine (1 micro M), a specific L-type VDCC blocker. 4 Altogether, these data demonstrate that multiple VDCC subtypes are present at the mouse NMJ and that a resistant component can be identified under 'pharmacological' and/or 'pathological' conditions.


Assuntos
4-Aminopiridina/farmacologia , Acetilcolina/metabolismo , Autoanticorpos/farmacologia , Canais de Cálcio/fisiologia , Síndrome Miastênica de Lambert-Eaton/imunologia , Junção Neuromuscular/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio Tipo N/efeitos dos fármacos , Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/efeitos dos fármacos , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/efeitos dos fármacos , Canais de Cálcio Tipo Q/fisiologia , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/fisiologia , Feminino , Humanos , Imunoglobulina G/farmacologia , Técnicas In Vitro , Masculino , Camundongos , Pessoa de Meia-Idade , Junção Neuromuscular/metabolismo , Junção Neuromuscular/fisiologia
8.
J Neurosci ; 19(21): 9235-41, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10531427

RESUMO

Multiple types of voltage-dependent Ca(2+) channels are involved in the regulation of neurotransmitter release (Tsien et al., 1991; Dunlap et al., 1995). In the nerve terminals of the neurohypophysis, the roles of L-, N-, and P/Q-type Ca(2+) channels in neuropeptide release have been identified previously (Wang et al., 1997a). Although the L- and N-type Ca(2+) currents play equivalent roles in both vasopressin and oxytocin release, the P/Q-type Ca(2+) current only regulates vasopressin release. An oxytocin-release and Ca(2+) current component is resistant to the L-, N-, and P/Q-type Ca(2+) channel blockers but is inhibited by Ni(2+). A new polypeptide toxin, SNX-482, which is a specific alpha(1E)-type Ca(2+) channel blocker (Newcomb et al., 1998), was used to characterize the biophysical properties of this resistant Ca(2+) current component and its role in neuropeptide release. This resistant component was dose dependently inhibited by SNX-482, with an IC(50) of 4.1 nM. Furthermore, SNX-482 did not affect the other Ca(2+) current types in these CNS terminals. Like the N- and P/Q-type Ca(2+) currents, this SNX-482-sensitive transient Ca(2+) current is high-threshold activated and shows moderate steady-state inactivation. At the same concentrations, SNX-482 blocked the component of oxytocin, but not of vasopressin, release that was resistant to the other channel blockers, indicating a preferential role for this type of Ca(2+) current in oxytocin release from neurohypophysial terminals. Our results suggest that an alpha(1E) or "R"-type Ca(2+) channel exists in oxytocinergic nerve terminals and, thus, functions in controlling only oxytocin release from the rat neurohypophysis.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo R/fisiologia , Terminações Nervosas/fisiologia , Ocitocina/metabolismo , Neuro-Hipófise/fisiologia , Venenos de Aranha/farmacologia , ômega-Conotoxinas , Animais , Arginina Vasopressina/metabolismo , Canais de Cálcio Tipo R/química , Canais de Cálcio Tipo R/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Terminações Nervosas/efeitos dos fármacos , Nicardipino/farmacologia , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Neuro-Hipófise/efeitos dos fármacos , Ratos , ômega-Agatoxina IVA/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA