Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cardiovasc Eng Technol ; 11(3): 316-327, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32356274

RESUMO

PURPOSE: Fibrocalcific aortic valve disease (CAVD) is caused by the deposition of calcific nodules in the aortic valve leaflets, resulting in progressive loss of function that ultimately requires surgical intervention. This process is actively mediated by the resident valvular interstitial cells (VICs), which, in response to oxidized lipids, transition from a quiescent to an osteoblast-like state. The purpose of this study was to examine if the ryanodine receptor, an intracellular calcium channel, could be therapeutically targeted to prevent this phenotypic conversion. METHODS: The expression of the ryanodine receptor in porcine aortic VICs was characterized by qRT-PCR and immunofluorescence. Next, the VICs were exposed to lysophosphatidylcholine, an oxidized lipid commonly found in low-density lipoprotein, while the activity of the ryanodine receptor was modulated with ryanodine. The cultures were analyzed for markers of cellular mineralization, alkaline phosphatase activity, proliferation, and apoptosis. RESULTS: Porcine aortic VICs predominantly express isoform 3 of the ryanodine receptors, and this protein mediates the cellular response to LPC. Exposure to LPC caused elevated intracellular calcium concentration in VICs, raised levels of alkaline phosphatase activity, and increased calcific nodule formation, but these changes were reversed when the activity of the ryanodine receptor was blocked. CONCLUSIONS: Our findings suggest blocking the activity of the ryanodine receptor can attenuate the valvular mineralization caused by LPC. We conclude that oxidized lipids, such as LPC, play an important role in the development and progression of CAVD and that the ryanodine receptor is a promising target for pharmacological intervention.


Assuntos
Valva Aórtica/efeitos dos fármacos , Calcinose/induzido quimicamente , Agonistas dos Canais de Cálcio/toxicidade , Cálcio/metabolismo , Lisofosfatidilcolinas/toxicidade , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Apoptose/efeitos dos fármacos , Calcinose/metabolismo , Calcinose/patologia , Calcinose/prevenção & controle , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Sus scrofa
2.
Heart Vessels ; 35(4): 593-602, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31628538

RESUMO

Torsade de pointes (TdP) occurred in a long QT syndrome type 3 (LQT3) patient after switching perospirone to blonanserin. We studied how their electropharmacological effects had induced TdP in the LQT3 patient. Perospirone hydrochloride (n = 4) or blonanserin (n = 4) of 0.01, 0.1, and 1 mg/kg, i.v. was cumulatively administered to the halothane-anesthetized dogs over 10 min. The low dose of perospirone decreased total peripheral vascular resistance, but increased heart rate and cardiac output, facilitated atrioventricular conduction, and prolonged J-Tpeakc. The middle dose decreased mean blood pressure and prolonged repolarization period, in addition to those observed after the low dose. The high dose further decreased mean blood pressure with the reduction of total peripheral vascular resistance; however, it did not increase heart rate or cardiac output. It tended to delay atrioventricular conduction and further delayed repolarization with the prolongation of Tpeak-Tend, whereas J-Tpeakc returned to its baseline level. Meanwhile, each dose of blonanserin decreased total peripheral vascular resistance, but increased heart rate, cardiac output and cardiac contractility in a dose-related manner. J-Tpeakc was prolonged by each dose, but Tpeak-Tend was shortened by the middle and high doses. These results indicate that perospirone and blonanserin may cause the hypotension-induced, reflex-mediated increase of sympathetic tone, leading to the increase of inward Ca2+ current in the heart except that the high dose of perospirone reversed them. Thus, blonanserin may have more potential to produce intracellular Ca2+ overload triggering early afterdepolarization than perospirone, which might explain the onset of TdP in the LQT3 patient.


Assuntos
Doença do Sistema de Condução Cardíaco/fisiopatologia , Antagonistas de Dopamina/toxicidade , Sistema de Condução Cardíaco/efeitos dos fármacos , Síndrome do QT Longo/fisiopatologia , Antagonistas da Serotonina/toxicidade , Torsades de Pointes/induzido quimicamente , Potenciais de Ação/efeitos dos fármacos , Anestésicos Inalatórios , Animais , Agonistas dos Canais de Cálcio/toxicidade , Delírio/tratamento farmacológico , Cães , Relação Dose-Resposta a Droga , Eletrocardiografia , Feminino , Halotano , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Isoindóis , Pessoa de Meia-Idade , Modelos Animais , Piperazinas , Piperidinas , Bloqueadores dos Canais de Potássio/toxicidade , Distúrbios do Início e da Manutenção do Sono/tratamento farmacológico , Tiazóis , Torsades de Pointes/metabolismo , Torsades de Pointes/fisiopatologia
3.
Toxicol Mech Methods ; 29(2): 138-145, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30394170

RESUMO

Timolol is a medication used widely to treat glaucoma. Regarding Ca2+ signaling, timolol was shown to modulate Ca2+-related physiology in various cell types, however, the effect of timolol on Ca2+ homeostasis and cell viability has not been explored in human prostate cancer cells. The aim of this study was to explore the effect of timolol on intracellular Ca2+ concentrations ([Ca2+]i) and viability in PC3 human prostate cancer cells. Timolol at concentrations of 100-1000 µM induced [Ca2+]i rises. The Ca2+ signal in Ca2+-containing medium was reduced by removal of extracellular Ca2+ by approximately 75%. Timolol (1000 µM) induced Mn2+ influx suggesting of Ca2+ entry. Timolol-induced Ca2+ entry was partially inhibited by three inhibitors of store-operated Ca2+ channels: nifedipine, econoazole and SKF96365, and by a protein kinase C (PKC) activator (phorbol 12-myristate 13 acetate [PMA]) or an inhibitor (GF109203X). In Ca2+-free medium, treatment with the endoplasmic reticulum Ca2+ pump inhibitor thapsigargin abolished timolol-evoked [Ca2+]i rises. Conversely, treatment with timolol abolished thapsigargin-evoked [Ca2+]i rises. Inhibition of phospholipase C (PLC) with U73122 abolished timolol-induced [Ca2+]i rises. Timolol at concentrations between 200 and 600 µM killed cells in a concentration-dependent fashion. Chelation of cytosolic Ca2+ with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid/AM (BAPTA/AM) did not reverse cytotoxicity of timolol. Together, in PC3 cells, timolol induced [Ca2+]i rises by evoking Ca2+release from the endoplasmic reticulum in a PLC-dependent manner, and Ca2+ influx via PKC-regulated store-operated Ca2+ entry. Timolol also caused cell death that was not linked to preceding [Ca2+]i rises.


Assuntos
Agonistas dos Canais de Cálcio/toxicidade , Canais de Cálcio/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Próstata/efeitos dos fármacos , Timolol/toxicidade , Canais de Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Humanos , Cinética , Masculino , Células PC-3 , Próstata/metabolismo , Próstata/patologia , Proteína Quinase C/metabolismo , Fosfolipases Tipo C/metabolismo
4.
J Med Chem ; 61(18): 8186-8201, 2018 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-30148953

RESUMO

Transient-receptor-potential melastatin 8 (TRPM8), the predominant mammalian cold-temperature thermosensor, is a nonselective cation channel expressed in a subpopulation of sensory neurons in the peripheral nervous system, including nerve circuitry implicated in migraine pathogenesis: the trigeminal and pterygopalatine ganglia. Genomewide association studies have identified an association between TRPM8 and reduced risk of migraine. This disclosure focuses on medicinal-chemistry efforts to improve the druglike properties of initial leads, particularly removal of CYP3A4-induction liability and improvement of pharmacokinetic properties. A novel series of biarylmethanamide TRPM8 antagonists was developed, and a subset of leads were evaluated in preclinical toxicology studies to identify a clinical candidate with an acceptable preclinical safety profile leading to clinical candidate AMG 333, a potent and highly selective antagonist of TRPM8 that was evaluated in human clinical trials.


Assuntos
Anticonvulsivantes/farmacologia , Descoberta de Drogas , Transtornos de Enxaqueca/prevenção & controle , Niacina/química , Convulsões/tratamento farmacológico , Canais de Cátion TRPM/antagonistas & inibidores , Animais , Anticonvulsivantes/química , Agonistas dos Canais de Cálcio/toxicidade , Humanos , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Modelos Moleculares , Estrutura Molecular , Pirimidinonas/toxicidade , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente
5.
J Toxicol Sci ; 41(3): 439-47, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27193735

RESUMO

Since amantadine-induced long QT syndrome has been clinically reported, we investigated its electropharmacological effects to estimate the extent of proarrhythmic risk by using the halothane-anesthetized beagle dogs (n = 4). Amantadine in doses of 0.1, 1 and 10 mg/kg was infused over 10 min with a pause of 20 min under the monitoring of multiple cardiovascular variables. J-Tpeak and Tpeak-Tend were separately measured on the lead II electrocardiogram to precisely analyze the net balance between inward and outward current modifications by amantadine. The low dose increased the ventricular contractile force, but suppressed the intraventricular conduction. The middle dose prolonged the QT interval besides enhancing the changes induced by the low dose. The high dose increased the mean blood pressure, left ventricular end-diastolic pressure and total peripheral resistance, and accelerated the atrioventricular nodal conduction, but decreased the cardiac output besides enhancing the changes induced by the middle dose. A reverse use-dependence was confirmed in the repolarization delay. Amantadine hardly affected the J-Tpeak, but prolonged the Tpeak-Tend. Amantadine can be considered to stimulate Ca(2+) channel but inhibit Na(+) and K(+) channels in the in situ heart. J-Tpeak and Tpeak-Tend analysis suggests that amantadine may possess modest risk for arrhythmia.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Amantadina/toxicidade , Anestésicos Inalatórios , Arritmias Cardíacas/induzido quimicamente , Agonistas dos Canais de Cálcio/toxicidade , Halotano , Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/toxicidade , Bloqueadores dos Canais de Sódio/toxicidade , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Pressão Arterial/efeitos dos fármacos , Estimulação Cardíaca Artificial , Cães , Relação Dose-Resposta a Droga , Eletrocardiografia , Técnicas Eletrofisiológicas Cardíacas , Feminino , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Masculino , Modelos Animais , Medição de Risco , Fatores de Tempo , Função Ventricular Esquerda/efeitos dos fármacos , Pressão Ventricular/efeitos dos fármacos
6.
Anesth Analg ; 122(5): 1360-9, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26771269

RESUMO

BACKGROUND: High systemic lidocaine concentrations exert well-known toxic effects on the central nervous system (CNS), including seizures, coma, and death. The underlying mechanisms are still largely obscure, and the actions of lidocaine on supraspinal neurons have received comparatively little study. We recently found that lidocaine at clinically neurotoxic concentrations increases excitability mediated by Na-independent, high-threshold (HT) action potential spikes in rat thalamocortical neurons. Our goal in this study was to characterize these spikes and test the hypothesis that they are generated by HT Ca currents, previously implicated in neurotoxicity. We also sought to identify and isolate the specific underlying subtype of Ca current. METHODS: We investigated the actions of lidocaine in the CNS-toxic concentration range (100 µM-1 mM) on ventrobasal thalamocortical neurons in rat brain slices in vitro, using whole-cell patch-clamp recordings aided by differential interference contrast infrared videomicroscopy. Drugs were bath applied; action potentials were generated using current clamp protocols, and underlying currents were identified and isolated with ion channel blockers and electrolyte substitution. RESULTS: Lidocaine (100 µM-1 mM) abolished Na-dependent tonic firing in all neurons tested (n = 46). However, in 39 of 46 (85%) neurons, lidocaine unmasked evoked HT action potentials with lower amplitudes and rates of de-/repolarization compared with control. These HT action potentials remained during the application of tetrodotoxin (600 nM), were blocked by Cd (50 µM), and disappeared after superfusion with an extracellular solution deprived of Ca. These features implied that the unmasked potentials were generated by high-voltage-activated Ca channels and not by Na channels. Application of the L-type Ca channel blocker, nifedipine (5 µM), completely blocked the HT potentials, whereas the N-type Ca channel blocker, ω-conotoxin GVIA (1 µM), had little effect. CONCLUSIONS: At clinically CNS-toxic concentrations, lidocaine unmasked in thalamocortical neurons evoked HT action potentials mediated by the L-type Ca current while substantially suppressing Na-dependent excitability. On the basis of the known role of an increase in intracellular Ca in the pathogenesis of local anesthetic neurotoxicity, this novel action represents a plausible contributing candidate mechanism for lidocaine's CNS toxicity in vivo.


Assuntos
Anestésicos Locais/toxicidade , Agonistas dos Canais de Cálcio/toxicidade , Canais de Cálcio Tipo L/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Lidocaína/toxicidade , Neurônios/efeitos dos fármacos , Núcleos Ventrais do Tálamo/efeitos dos fármacos , Potenciais de Ação , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Relação Dose-Resposta a Droga , Técnicas In Vitro , Neurônios/metabolismo , Neurônios/patologia , Ratos Sprague-Dawley , Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Fatores de Tempo , Núcleos Ventrais do Tálamo/metabolismo , Núcleos Ventrais do Tálamo/patologia
8.
Neurosci Lett ; 461(3): 271-4, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19545607

RESUMO

The TRPA1 agonist mustard oil (allyl isothiocyanate=AITC) induces heat hyperalgesia and mechanical allodynia in human skin and sensitizes rat spinal wide dynamic range (WDR) neuronal responses to noxious skin heating. We presently used electrophysiological methods to investigate if AITC affects the responsiveness of individual spinal WDR neurons to intense skin cooling. Recordings were made from cold-sensitive WDR neurons in lamina I and deeper dorsal horn; 21/23 also responded to noxious skin heating. Topical application of AITC excited 8/18 units and significantly enhanced their responses to noxious heat while not significantly affecting responses to the cold stimulus. Vehicle (mineral oil) had no effect on thermal responses. The data confirm a role for the TRPA1 agonist AITC in enhancing heat nociception without significantly affecting cold sensitivity.


Assuntos
Compostos Alílicos/toxicidade , Agonistas dos Canais de Cálcio/toxicidade , Temperatura Baixa/efeitos adversos , Temperatura Alta/efeitos adversos , Isocianatos/toxicidade , Neurônios/efeitos dos fármacos , Dor/fisiopatologia , Medula Espinal/efeitos dos fármacos , Potenciais de Ação , Animais , Anquirinas , Canais de Cálcio/fisiologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Masculino , Mostardeira/toxicidade , Neurônios/fisiologia , Dor/induzido quimicamente , Óleos de Plantas/toxicidade , Ratos , Pele/efeitos dos fármacos , Pele/fisiopatologia , Medula Espinal/fisiopatologia , Canal de Cátion TRPA1 , Canais de Cátion TRPC
9.
Basic Clin Pharmacol Toxicol ; 94(2): 79-88, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14748851

RESUMO

The present study will summarize our findings concerning the anticonvulsant properties of the Ca2+ channel blocker flunarizine in a variety of experimental models of epilepsy. Flunarizine exhibits anticonvulsant effects against tonic seizures induced by electroshock or various chemoconvulsants in mice, however, did not protect against pentylenetetrazol-induced clonic seizures. In the MES test, the efficacy of clinically established antiepileptics was increased by co-medication. In the rotarod test, a minimal "neurotoxic" dose (TD50) of 18.0 mg/kg intraperitoneally was determined. In models of complex partial seizures like the hippocampal stimulation and the amygdala kindling in rats, flunarizine showed only a moderate activity. Thus, it can be suggested that the anticonvulsant potency of flunarizine in various screening tests is lower than that of standard antiepileptics such as carbamazepine and phenytoin. Concerning the possible mode of action, whole-cell patch-clamp experiments with cultured neonatal rat cardiomyocytes showed that flunarizine depressed the fast inward Na+ current in a concentration- and frequency-dependent manner well comparable with the action of phenytoin. It is concluded that the use-dependent inhibition of voltage-dependent Na+ channels may essentially contribute to the anticonvulsant activity of flunarizine in models for generalized tonic-clonic seizures. The clinical efficacy as add-on therapy is critically discussed in view of the present data.


Assuntos
Anticonvulsivantes/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Flunarizina/farmacologia , Convulsões/tratamento farmacológico , Canais de Sódio/efeitos dos fármacos , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/toxicidade , Animais , Encéfalo/metabolismo , Agonistas dos Canais de Cálcio/toxicidade , Células Cultivadas , Convulsivantes/toxicidade , Relação Dose-Resposta a Droga , Interações Medicamentosas , Eletrochoque , Masculino , Camundongos , Pentilenotetrazol/toxicidade , Ratos , Ratos Wistar , Teste de Desempenho do Rota-Rod , Convulsões/induzido quimicamente , Convulsões/etiologia
10.
Proteins ; 40(3): 436-42, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10861934

RESUMO

We determined the structure in solution by (1)H two-dimensional NMR of Maurocalcine from the venom of Scorpio maurus. This toxin has been demonstrated to be a potent effector of ryanodyne-sensitive calcium channel from skeletal muscles. This is the first description of a scorpion toxin which folds following the Inhibitor Cystine Knot fold (ICK) already described for numerous toxic and inhibitory peptides, as well as for various protease inhibitors. Its three dimensional structure consists of a compact disulfide-bonded core from which emerge loops and the N-terminus. A double-stranded antiparallel beta-sheet comprises residues 20-23 and 30-33. A third extended strand (residues 9-11) is perpendicular to the beta-sheet. Maurocalcine structure mimics the activating segment of the dihydropyridine receptor II-III loop and is therefore potentially useful for dihydropyridine receptor/ryanodine receptor interaction studies. Proteins 2000;40:436-442.


Assuntos
Agonistas dos Canais de Cálcio/química , Venenos de Escorpião/química , Sequência de Aminoácidos , Agonistas dos Canais de Cálcio/toxicidade , Canais de Cálcio Tipo L/metabolismo , Simulação por Computador , Modelos Moleculares , Mimetismo Molecular , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Venenos de Escorpião/toxicidade , Homologia de Sequência de Aminoácidos
11.
J Ethnopharmacol ; 55(2): 113-8, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9032623

RESUMO

In order to elucidate a possible role for calcium on the negative cardiotropic effects of a garlic (Allium sativum L., Liliaceae) dialysate in rat atria we studied: (a) the effects of our extract 15 min after preincubation with high and low concentrations of extracellular calcium ([Ca2+]o) on left and right activity of rat atria. The negative inotropism of garlic dialysate increased with calcium 0.75 mM; in contrast, high level of calcium (4.5 mM) induced a significant reduction of this depressant effect. None of these treatments modified the negative chronotropism of garlic; (b) nifedipine (10(-9) to 10(-7) M, verapamil (10(-9) to 10(-7) M) and diltiazem (10(-9) to 10(-7) M) induced a concentration-dependent synergism of the log concentration-effect of garlic dialysate on left atria. Verapamil and diltiazem (10(-7)M), but not nifedipine increased the inhibitory chronotropism of garlic in right atria; (c) negative inotropic and chronotropic effects demonstrated by nifedipine (1 x 10(-10) to 1.1 x 10(-6) M) were antagonized as expected by preincubation with Bay K-8644. Depressant actions of garlic were not modified with this pretreatment. These results suggest that the negative inotropic effect of our garlic dialysate is related to [Ca2+]o availability. It is possible that a restriction of intracellular calcium contributes to this effect. However, the negative chronotropic effect of garlic is scarcely affected by these modifications.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Cálcio/metabolismo , Alho/metabolismo , Átrios do Coração/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Plantas Medicinais , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/toxicidade , Animais , Cálcio/toxicidade , Agonistas dos Canais de Cálcio/toxicidade , Diálise , Diltiazem/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Sinergismo Farmacológico , Átrios do Coração/metabolismo , Masculino , Nifedipino/farmacologia , Extratos Vegetais/administração & dosagem , Extratos Vegetais/metabolismo , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Ratos , Verapamil/farmacologia
12.
Pol J Pharmacol ; 49(5): 369-71, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9566038

RESUMO

The study was designed to investigate the effects of administration of cholinomimetic agents and nifedipine on seizures induced by BAY k-8644 in rats. Injection of pilocarpine (3 mg/kg) increased seizures induced by BAY k-8644 (2 mg/kg). Administration of nifedipine (10 mg/kg) did not affect the convulsions and mortality elicited by coadministration of BAY k-8644 and pilocarpine. However, the facilitating effect of pilocarpine on BAY k-8644-induced seizures was fully prevented by pretreatment of rats with a cholinergic antagonist atropine (5 mg/kg). No similar effects were observed after injection of another cholinomimetic agent physostigmine (0.3 mg/kg). This finding implies, that facilitation of convulsant action of BAY k-8644 by pilocarpine may be related to activity of cholinergic system, but not strictly to activity of voltage dependent calcium channels.


Assuntos
Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/toxicidade , Agonistas dos Canais de Cálcio/toxicidade , Canais de Cálcio/fisiologia , Convulsivantes/toxicidade , Receptores Colinérgicos/fisiologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Ativação do Canal Iônico , Masculino , Agonistas Muscarínicos/farmacologia , Nifedipino/farmacologia , Pilocarpina/farmacologia , Ratos , Ratos Wistar , Receptores Colinérgicos/efeitos dos fármacos , Convulsões/induzido quimicamente , Convulsões/fisiopatologia
13.
Nat Toxins ; 3(6): 419-27, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-8612004

RESUMO

Maitotoxin (MTX) is a 3,424 dalton polyether marine toxin that causes influx of calcium through type L voltage-dependent calcium channels (L-VDCC) in GH4C1 rat pituitary cells, presumably as the result of membrane depolarization. In this study we have investigated the ionic conductances responsible for MTX-induced depolarization under voltage clamp conditions using the perforated and ruptured patch methods. MTX-induced steady-state voltage independent currents of nearly 400 pS/pF within seconds of addition to the bath. Ion substitution experiments demonstrated these currents are consistent with the conductance of sodium and chloride, but not calcium, ions. MTX induction of the voltage-independent chloride conductance in GH4C1 cells occurred concurrently without modification of L-VDCC currents. Pretreatment with nimodipine eliminated voltage activation of L-VDCC, and reduced by two thirds the voltage independent current. Analysis as a function of time of MTX exposure revealed that the first 60 sec of MTX-induced currents were not affected by nimodipine pretreatment, but subsequent additional currents were prevented. This indicates that the initial currents induced by MTX occur independently of L-VDCC mediated calcium entry, but full activation of these currents by MTX likely requires the involvement L-VDCC. Taken together this work identifies a voltage-independent sodium/chloride conductance as an initial action of MTX, one that may promote the sequence of ionic events leading to activation of L-VDCC and massive calcium entry.


Assuntos
Agonistas dos Canais de Cloreto , Toxinas Marinhas/toxicidade , Oxocinas , Hipófise/efeitos dos fármacos , Agonistas de Canais de Sódio , Potenciais de Ação/efeitos dos fármacos , Animais , Agonistas dos Canais de Cálcio/toxicidade , Células Cultivadas , Técnicas de Patch-Clamp , Hipófise/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA