Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Chin Med ; 41(4): 789-800, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23895152

RESUMO

Ginkgo biloba has been used for medical purposes for centuries in traditional Chinese medicine. Ginkgo biloba extract 50 (GBE50) is a new standardized GBE product that matches the standardized German product as EGb761. This paper is aimed at studying the cardio-protection effects of GBE50 Salvia miltiorrhiza on myocardial function, area at risk, myocardial ultra-structure, and expression of calcium handling proteins in rat ischemic myocardium. Myocardium ischemia was induced by the left anterior descending (LAD) coronary artery occlusion and myocardial function was recorded by a transducer advanced into the left ventricle on a computer system. In vitro myocardial infarction was measured by 2,3,5-triphenyltetrazolium chloride (TTC) and Evans blue staining of heart sections. Morphological change was evaluated by electric microscopy and Western blotting was used for protein expression. Hemodynamic experiments in vivo showed that postischemic cardiac contractile function was reduced in ischemic rats. Salvia miltiorrhiza (7.5 g/kg/d×7) and Ginkgo biloba extract 50 (GBE50) (100 mg/kg/d×7) improved post-schemic cardiac diastolic dysfunction while not affecting the systolic function. In hearts of GBE50 group and Salvia miltiorrhiza (SM) group, the area at risk was significantly reduced and myocardial structure was better-preserved. Moreover, Na⁺-Ca²âº exchanger (NCX) expression increase and sarcoplasmic reticulum Ca²âº-ATPase 2 (SERCA2), LTCC, and ryanodine receptor 2 (RyR2) expression decreases were smaller than those in ischemia group. There was a significant difference between the GBE50 and ischemia group in NCX expression. GBE50 could improve recovery in contractile function and prevent myocardium from ischemia damage, which may be caused by attenuating the abnormal expression of NCX.


Assuntos
Cardiotônicos/farmacologia , Ginkgo biloba , Coração/efeitos dos fármacos , Infarto do Miocárdio , Fitoterapia/métodos , Extratos Vegetais/farmacologia , Salvia miltiorrhiza , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Masculino , Microscopia Eletrônica , Contração Miocárdica/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica , Miocárdio/ultraestrutura , Ratos , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/efeitos dos fármacos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Trocador de Sódio e Cálcio/efeitos dos fármacos , Trocador de Sódio e Cálcio/metabolismo
2.
Anesthesiology ; 119(1): 111-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23459219

RESUMO

BACKGROUND: Mutations within the gene encoding the skeletal muscle calcium channel ryanodine receptor can result in malignant hyperthermia. Although it is important to characterize the functional effects of candidate mutations to establish a genetic test for diagnosis, ex vivo methods are limited because of the low incidence of the disorder and sample unavailability. More than 250 candidate mutations have been identified, but only a few mutations have been functionally characterized. METHODS: The human skeletal muscle ryanodine receptor complementary DNA was cloned with or without a disease-related variant. Wild-type and mutant calcium channel proteins were transiently expressed in human embryonic kidney-293 cells expressing the large T-antigen of simian virus 40, and functional analysis was carried out using calcium imaging with fura-2 AM. Six human malignant hyperthermia-related mutants such as R44C, R163C, R401C, R533C, R533H, and H4833Y were analyzed. Cells were stimulated with a specific ryanodine receptor agonist 4-chloro-m-cresol, and intracellular calcium mobility was analyzed to determine the functional aspects of mutant channels. RESULTS: Mutant proteins that contained a variant linked to malignant hyperthermia showed higher sensitivity to the agonist. Compared with the wild type (EC50=453.2 µM, n=18), all six mutants showed a lower EC50 (21.2-170.4 µM, n=12-23), indicating susceptibility against triggering agents. CONCLUSIONS: These six mutations cause functional abnormality of the calcium channel, leading to higher sensitivity to a specific agonist, and therefore could be considered potentially causative of malignant hyperthermia reactions.


Assuntos
Hipertermia Maligna/genética , Mutação/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Cálcio/metabolismo , DNA Complementar/genética , DNA Complementar/metabolismo , Imunofluorescência , Células HEK293 , Humanos , Hipertermia Maligna/fisiopatologia , Mutação/fisiologia , Neuroimagem , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia
3.
Alcohol Clin Exp Res ; 37(5): 771-83, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23278119

RESUMO

BACKGROUND: Ryanodine receptors (RyRs) amplifying activity-dependent calcium influx via calcium-induced calcium release play an important role in central nervous system functions including learning, memory, and drug abuse. In this study, we investigated the role and the regulatory mechanisms of RyR expression under continuous exposure of mice to ethanol (EtOH) vapor for 9 days. METHODS: The model of EtOH physical dependence was prepared as follows: 8-week-old male ddY mice were exposed to EtOH vapor for 9 days. Protein and mRNA of RyR-1, RyR-2, and RyR-3 in the frontal cortex and limbic forebrain were determined by Western blot and real-time RT-PCR analysis, respectively. RESULTS: Exposure of mice to EtOH vapor for 9 days induced significant withdrawal signs when estimated with withdrawal score, which was dose-dependently suppressed by intracerebroventricular administration of dantrolene, an RyR antagonist. Protein levels of RyR-1 and RyR-2 in the frontal cortex and limbic forebrain significantly increased during EtOH vapor exposure for 9 days with increased expression of their mRNA, whereas that of RyR-3 in these 2 brain regions showed no changes. Increased proteins and mRNA of RyR-1 and RyR-2 were completely abolished by SCH23390, a selective antagonist of dopamine D1 receptors (D1DRs), but not by sulpiride, a selective antagonist of D2DRs. CONCLUSIONS: RyRs play a critical role in the development of EtOH physical dependence and that the up-regulation of RyRs in the brain of mouse, showing EtOH physical dependence is regulated by D1DRs.


Assuntos
Alcoolismo/metabolismo , Encéfalo/metabolismo , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Receptores de Dopamina D1/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Benzazepinas/farmacologia , Western Blotting , Dantroleno/farmacologia , Modelos Animais de Doenças , Lobo Frontal/metabolismo , Masculino , Camundongos , Prosencéfalo/metabolismo , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética
4.
Eur J Pharmacol ; 697(1-3): 81-7, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23051676

RESUMO

Salviae miltiorrhizae, a traditional Chinese medicine, is widely used in the treatment of cardiovascular and cerebrovascular diseases. Salvianolic acid B is identified as one of the most important water-soluble active ingredients in Salviae miltiorrhizae and associated with the activation of Ca(2+) channel of cytomembrane. But the further mechanism of action was not very clearly. In our study, we investigated the vasodilation activity of salvianolic acid B using the isolated thoracic aortic rings from Japanese white rabbit. Salvianolic acid B significantly released the contraction of the isolated thoracic aortic rings induced by phenylephrine and CaCl(2) while had no effects on the aortic rings with KCl stimulated. Different with Di-ao-xin-xue-kang capsule, salvianolic acid B caused an increase of Ca(2+) in cytoplasm from not only activation of Ca(2+) channel in cytomembrane but also release of endogenous Ca(2+). Then, a series of endogenous Ca(2+) inhibitors were pretreated to explore the mechanism of salvianolic acid B, and the results provided further evidences that salvianolic acid B causes intracellular calcium release in ryanodine receptors-dependent manners. Moreover, combining l-arginine (l-Arg) with salvianolic acid B promoted the vasodilation activity suggesting a relationship with nitric oxide (NO). To further investigated its mechanism, both guanylate cyclase (GC) inhibitor and NO Synthase inhibitor were used and demonstrated to block vasodilation activity of the aortic rings. Our findings reveal a NO-sGC-cGMP signals dependence mechanism of salvianolic acid B on its vasodilation activity which provide an evidence for its subsequent application in clinic.


Assuntos
Aorta Torácica/efeitos dos fármacos , Benzofuranos/farmacologia , Óxido Nítrico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Aorta Torácica/metabolismo , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , GMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Coelhos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Guanilil Ciclase Solúvel , Vasoconstritores/farmacologia
5.
Heart Rhythm ; 9(6): 953-60, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22245792

RESUMO

BACKGROUND: Ranolazine (Ran) is known to inhibit multiple targets, including the late Na(+)current, the rapid delayed rectifying K(+)current, the L-type Ca(2+)current, and fatty acid metabolism. Functionally, Ran suppresses early afterdepolarization (EADs) and torsades de pointes (TdP) in drug-induced long QT type 2 (LQT2) presumably by decreasing intracellular [Na(+)](i) and Ca(2+)overload. However, simulations of EADs in LQT2 failed to predict their suppression by Ran. OBJECTIVE: To elucidate the mechanism(s) whereby Ran alters cardiac action potentials (APs) and cytosolic Ca(2+)transients and suppresses EADs and TdP in LQT2. METHODS: The known effects of Ran were included in simulations (Shannon and Mahajan models) of rabbit ventricular APs and Ca(2+)transients in control and LQT2 models and compared with experimental optical mapping data from Langendorff rabbit hearts treated with E4031 (0.5 µM) to block the rapid delayed rectifying K(+)current. Direct effects of Ran on cardiac ryanodine receptors (RyR2) were investigated in single channels and changes in Ca(2+)-dependent high-affinity ryanodine binding. RESULTS: Ran (10 µM) alone prolonged action potential durations (206 ± 4.6 to 240 ± 7.8 ms; P <0.05); E4031 prolonged action potential durations (204 ± 6 to 546 ± 35 ms; P <0.05) and elicited EADs and TdP that were suppressed by Ran (10 µM; n = 7 of 7 hearts). Simulations (Shannon but not Mahajan model) closely reproduced experimental data except for EAD suppression by Ran. Ran reduced open probability (P(o)) of RyR2 (half maximal inhibitory concentration = 10 ± 3 µM; n = 7) in bilayers and shifted half maximal effective concentration for Ca(2+)-dependent ryanodine binding from 0.42 ± 0.02 to 0.64 ± 0.02 µM with 30 µM Ran. CONCLUSIONS: Ran reduces P(o) of RyR2, desensitizes Ca(2+)-dependent RyR2 activation, and inhibits Ca(i) oscillations, which represents a novel mechanism for its suppression of EADs and TdP.


Assuntos
Acetanilidas/farmacologia , Síndrome do QT Longo/complicações , Miocárdio/metabolismo , Piperazinas/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Torsades de Pointes/tratamento farmacológico , Potenciais de Ação/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Técnicas Eletrofisiológicas Cardíacas/métodos , Inibidores Enzimáticos/farmacologia , Feminino , Seguimentos , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/fisiopatologia , Coelhos , Ranolazina , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Torsades de Pointes/etiologia , Torsades de Pointes/fisiopatologia , Resultado do Tratamento
6.
Zhonghua Er Ke Za Zhi ; 49(6): 433-8, 2011 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-21924056

RESUMO

OBJECTIVE: To explore change of ryanodine receptor (RyR) in junior mouse with heart failure (HF) and the effect of ß-adrenoreceptor blocker and Radix astragali on RyR in HF in this experiment. METHOD: The animal model of congestive heart failure was established by coarctation of abdominal aorta. Five weeks old mice were randomly divided into 4 groups: (1) HF group without treatment (n = 30); (2) HF group treated with carvedilol (n = 30); (3) HF group treated with carvedilol and Radix astragali(n = 30); (4) Sham-operated group (n = 30). Carvedilol and Radix astragali were administered through direct gastric gavage. After 4 weeks of treatment the high frequency ultrasound was performed. Myocardial sarcoplasmic reticulum (SR) was fractionated with ultra centrifugation. The time courses of Ca(2+) uptake and leak were determined by fluorescent spectrophotometry. The levels of expression of RyR2 in the 4 groups were detected by semi-quantitative reverse transcription-polymerase chain reaction. RESULT: Compared with the sham-operated group, left ventricular diastolic dimension (LVEDD) (P < 0.05), left ventricular systolic dimension (LVESD), interventricular septal thickness at end-diastole (IVSTd), interventricular septal thickness at end-systole (IVSTs), left ventricular posterior wall thickness at end-diastole (LVPWTd), and left ventricular posterior wall thickness at endsystole (LVPWTs) were all significantly increased (P < 0.01), ejection fraction (EF)(%) (HF group without treatment 51.60 ± 1.15, HF treated with carvedilol 72.06 ± 1.39, HF treated with carvedilol and Radix astragali 79.06 ± 1.09, sham-operated group 85.86 ± 1.45) and fractional shortening (FS) (HF group without treatment 44.55 ± 1.20, HF treated with carvedilol 44.55 ± 1.20, HF treated with carvedilol and Radix astragali 53.58 ± 1.30, sham-operated group 59.03 ± 1.67) were decreased (P < 0.01) in HF group without treatment. LVEDD (P < 0.05), LVESD, IVSTd, IVSTs, LVPWTd and LVPWTs were all significantly decreased (P < 0.01), EF and FS were increased (P < 0.01) in the cases with HF treated with carvedilol and carvedilol and Radix astragali when compared with HF group without treatment. EF and FS were much more increased in the group treated with carvedilol and Radix astragali than in those treated with carvedilol (P < 0.05). After adding thapsigargin to the buffer including SR of the four groups, there were fewer Ca(2+) leak (%) in sham-operated group (11.5 ± 4.3), HF group treated with carvedilol (15.6 ± 5.8) and treated with carvedilol and Radix astragali (13.6 ± 4.8) than that of HF group without treatment (65.6 ± 6.2) (P < 0.01), while after adding FK506 and thapsigargin together to the buffer including SR of four groups, there were marked Ca(2+) leak in sham-operated group (60.6 ± 7.8), HF group treated with carvedilol (66.2 ± 4.5)and those treated with carvedilol and Radix astragali (70.2 ± 5.5, P < 0.01). However, there was no additional increase in Ca(2+) leak in HF group (67.3 ± 7.5) compared with that of the group where only thapsigargin was added (P > 0.05). The levels of expression of RyR2 were significantly decreased in HF group and increased in the group treated with carvedilol and the group treated with carvedilol and Radix astragali. CONCLUSION: There was more cardiac Ca(2+) leak and the expression of RyR2 mRNA decreased in HF. Carvedilol and Radix astragali can increase expression of RyR2 mRNA and inhibit Ca(2+) leak by restoring the binding of FKBP12.6 back to RyR in HF to improve cardiac function and prevent left ventricle from remodeling.


Assuntos
Carbazóis/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Insuficiência Cardíaca/metabolismo , Propanolaminas/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Antagonistas Adrenérgicos beta/farmacologia , Animais , Astrágalo , Astragalus propinquus , Carvedilol , Masculino , Ratos , Ratos Wistar , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
7.
J Physiol Sci ; 61(5): 363-72, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21691940

RESUMO

Oxymatrine is one of the alkaloids extracted from the Chinese herb Sophora japonica (Sophora flavescens Ait.) with anti-inflammatory, immune reaction inhibiting, antiviral, and hepatocyte and antihepatic fibrosis protective activities. However, the effect of oxymatrine on heart failure is not yet known. In this study, the effect of oxymatrine on heart failure was investigated using a Sprague-Dawley rat model of chronic heart failure. Morphological findings showed that in the group treated with 50 and 100 mg/kg of oxymatrine; intermyofibrillar lysis disappeared, myofilaments were orderly, closely and evenly arranged; and mitochondria contained tightly packed cristae compared with the heart failure group. We investigated the cytosolic Ca(2+) transients and sarcoplasmic reticulum (SR) Ca(2+) content, and assessed the expression of ryanodine receptor (RyR2), SR-Ca(2+) ATPase (SERCA2a), and L-type Ca(2+) channel (dihydropyridine receptor, DHPR). We found that the cytosolic Ca(2+) transients were markedly increased in amplitude in the medium- (ΔF/F (0) = 26.22 ± 2.01) and high-dose groups (ΔF/F (0) = 29.49 ± 1.17) compared to the heart failure group (ΔF/F (0) = 12.12 ± 1.35, P < 0.01), with changes paralleled by a significant increase in the SR Ca(2+) content (medium-dose group: ΔF/F (0) = 32.20 ± 1.67, high-dose group: ΔF/F (0) = 32.57 ± 1.29, HF: ΔF/F (0) = 17.26 ± 1.05, P < 0.01). Moreover, we demonstrated that the expression of SERCA2a and cardiac DHPR was significantly increased in the medium- and high-dose group compared with the heart failure rats. These findings suggest that oxymatrine could improve heart failure by improving the cardiac function and that this amelioration is associated with upregulation of SERCA2a and DHPR.


Assuntos
Alcaloides/uso terapêutico , Antiarrítmicos/uso terapêutico , Insuficiência Cardíaca/tratamento farmacológico , Quinolizinas/uso terapêutico , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/biossíntese , Canais de Cálcio Tipo L/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Doença Crônica , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Masculino , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/patologia , Ratos , Ratos Sprague-Dawley , Canal de Liberação de Cálcio do Receptor de Rianodina/biossíntese , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/biossíntese , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/efeitos dos fármacos
8.
J Pharmacol Exp Ther ; 334(3): 1042-50, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20551293

RESUMO

Our previous studies have demonstrated that an increase in intracellular levels of Ca(2+) in neurons is an important component of both the antinociception produced by morphine and morphine's tolerance. The present study tested the hypothesis that the Ca(2+) signaling second messenger, cyclic ADP-ribose (cADPR), derived from CD38 activation participates in morphine antinociception and tolerance. We first showed that morphine's antinociceptive potency was increased by the intracerebroventricular injection of CD38 substrate beta-NAD(+) in mice. Furthermore, morphine tolerance was reversed by intracerebroventricular administration of each of three different inhibitors of the CD38-cADPR-ryanodine receptor Ca(2+) signaling pathway. These inhibitors were the ADP-ribosylcyclase inhibitor nicotinamide, cADPR analog 8-bromo-cADPR, and a large dose of ryanodine (>50 muM) that blocks the ryanodine receptor. In CD38 gene knockout [CD38(-/-)] mice, the antinociceptive action of morphine was found to be less potent compared with wild-type (WT) mice, as measured by tail-flick response, hypothermia assay, and observations of straub tail. However, there was no difference in locomotor activation between CD38(-/-) and WT animals. It was also found that less tolerance to morphine developed in CD38(-/-) mice compared with WT animals. These results indicate that cADRP-ryanodine receptor Ca(2+) signaling associated with CD38 plays an important role in morphine tolerance.


Assuntos
ADP-Ribosil Ciclase 1/genética , ADP-Ribosil Ciclase 1/fisiologia , Analgésicos Opioides/farmacologia , Morfina/farmacologia , Dor/tratamento farmacológico , Animais , ADP-Ribose Cíclica/metabolismo , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Temperatura Alta , Hipotermia/induzido quimicamente , Hipotermia/fisiopatologia , Imersão/fisiopatologia , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , NAD/farmacologia , Niacinamida/farmacologia , Medição da Dor/efeitos dos fármacos , Tempo de Reação , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos
9.
Biochem Pharmacol ; 80(4): 512-21, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20471964

RESUMO

Catechins, polyphenols extracted from green tea leaves, have a broad range of biological activities although the specific molecular mechanisms responsible are not known. At the high experimental concentrations typically used polyphenols bind to membrane phospholipid and also are easily auto-oxidized to generate superoxide anion and semiquinones, and can adduct to protein thiols. We report that the type 1 ryanodine receptor (RyR1) is a molecular target that responds to nanomolar (-)-epigallocatechin-3-gallate (EGCG) and (-)-epicatechin-3-gallate (ECG). Single channel analyses demonstrate EGCG (5-10nM) increases channel open probability (Po) twofold, by lengthening open dwell time. The degree of channel activation is concentration-dependent and is rapidly and fully reversible. Four related catechins, EGCG, ECG, EGC ((-)-epigallocatechin) and EC ((-)-epicatechin) showed a rank order of activity toward RyR1 (EGCG>ECG>>EGC>>>EC). EGCG and ECG enhance the sensitivity of RyR1 to activation by < or =100microM cytoplasmic Ca(2+) without altering inhibitory potency by >100microM Ca(2+). EGCG as high as 10microM in the extracellular medium potentiated Ca(2+) transient amplitudes evoked by electrical stimuli applied to intact myotubes and adult FDB fibers, without eliciting spontaneous Ca(2+) release or slowing Ca(2+) transient recovery. The results identify RyR1 as a sensitive target for the major tea catechins EGCG and ECG, and this interaction is likely to contribute to their observed biological activities.


Assuntos
Catequina/análogos & derivados , Catequina/farmacologia , Extratos Vegetais/química , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Potenciais de Ação , Adulto , Anticarcinógenos , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Humanos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/fisiologia , Transdução de Sinais/fisiologia , Relação Estrutura-Atividade , Transmissão Sináptica/efeitos dos fármacos , Chá/química
10.
J Neurochem ; 106(6): 2385-94, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18643873

RESUMO

Elevation of baseline intracellular calcium levels was observed in platelets or lymphoblasts of patients with bipolar affective disorders suggesting an altered intracellular Ca(2+) homeostasis in the pathophysiology of mood disorders. The role of supraspinal endoplasmic ryanodine receptors (RyRs), which allow mobilization of intracellular Ca(2+) stores, in the modulation of depressive states was, then, investigated. Ryanodine and FK506 reduced the immobility time in the mouse forced swimming test showing an antidepressant-like profile comparable with that produced by amitriptyline and clomipramine. We generated types 1, 2, and 3 RyR knockdown mice by using selective antisense oligonucleotides (aODN) to investigate the role of each RyR isoform. A gene-specific cerebral RyR protein level reduction in knockdown animals was demonstrated by immunoblotting, immunoprecipitation, and immunohistochemical experiments. Repeated intracerebroventricular administration of aODNs complementary to the sequence of the types 1, 2, or 3 RyR produced an antidepressant-like response in the forced swimming test. The aODN-induced reduction of immobility time was temporary and reversible and did not impair motor coordination, spontaneous mobility, and exploratory activity. These findings identify cerebral RyRs as critical targets underlying depressive states and should facilitate the comprehension of the pathophysiology of mood disorders and help developing of new therapeutical strategies.


Assuntos
Encéfalo/metabolismo , Transtorno Depressivo/genética , Transtorno Depressivo/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Animais , Antidepressivos Tricíclicos/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Química Encefálica/efeitos dos fármacos , Química Encefálica/genética , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Transtorno Depressivo/tratamento farmacológico , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Imunossupressores/farmacologia , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Oligonucleotídeos Antissenso/farmacologia , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Tacrolimo/farmacologia
11.
Am J Physiol Heart Circ Physiol ; 295(2): H851-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18567703

RESUMO

Ginseng botanicals are increasingly used as complementary or alternative medicines for a variety of cardiovascular diseases, yet little is known about their cellular actions in cardiac muscle. Electromechanical alternans (EMA) is a proarrhythmic cardiac abnormality that results from disturbances of intracellular Ca(2+) homeostasis. This study sought to determine whether a purified ginsenoside extract of ginseng, Re, exerts effects to suppress EMA and to gain insight into its mechanism of action. Alternans was induced by electrically pacing cardiomyocytes at room temperature. Re (> or = 10 nM) reversibly suppressed EMA recorded from cat ventricular and atrial myocytes and Langendorff-perfused cat hearts. In cat ventricular myocytes, Re reversibly suppressed intracellular Ca(2+) concentration ([Ca(2+)](i)) transient alternans. Re exerted no significant effects on baseline action potential configuration or sarcolemmal L-type Ca(2+) current (I(Ca,L)), Na(+) current, or total K(+) conductance. In human atrial myocytes, Re suppressed mechanical alternans and exerted no effect on I(Ca,L). In cat ventricular myocytes, Re increased [Ca(2+)](i) transient amplitude and decreased sarcoplasmic reticulum (SR) Ca(2+) content, resulting in an increase in fractional SR Ca(2+) release. In SR microsomes isolated from cat ventricles, Re had no effect on SR Ca(2+) uptake. Re increased the open probability of ryanodine receptors (RyRs), i.e., SR Ca(2+)-release channels, isolated from cat ventricles and incorporated into planar lipid bilayers. We concluded that ginsenoside Re suppresses EMA in cat atrial and ventricular myocytes, cat ventricular muscle, and human atrial myocytes. The effects of Re are not mediated via actions on sarcolemmal ion channels or action potential configuration. Re acts via a subcellular mechanism to enhance the opening of RyRs and thereby overcome the impaired SR Ca(2+) release underlying EMA.


Assuntos
Antiarrítmicos/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Ginsenosídeos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Potenciais de Ação , Animais , Canais de Cálcio Tipo L/metabolismo , Estimulação Cardíaca Artificial , Gatos , Relação Dose-Resposta a Droga , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Humanos , Técnicas In Vitro , Miócitos Cardíacos/metabolismo , Potássio/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Sódio/metabolismo , Fatores de Tempo
12.
Learn Mem ; 15(5): 315-23, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18441289

RESUMO

The administration of the ryanodine receptor (RyR) agonist 4-Cmc (0.003-9 nmol per mouse intracerebroventricularly [i.c.v.]) ameliorated memory functions, whereas the RyR antagonist ryanodine (0.0001-1 nmol per mouse i.c.v.) induced amnesia in the mouse passive avoidance test. The role of the type 1, 2, and 3 RyR isoforms in memory processes was then evaluated by inhibiting the expression of the three RyR proteins in the mouse brain. A selective knockdown of the RyR isoforms was obtained by the i.c.v. administration of antisense oligonucleotides (aODNs) complementary to the sequence of RyR1, RyR2 and RyR3 proteins, as demonstrated by immunoblotting experiments. RyR1 (5-9 nmol per mouse i.c.v.) knockdown mice did not show any memory dysfunction. Conversely, RyR2 (1-7 nmol per mouse i.c.v.) and RyR3 (1-7 nmol per mouse i.c.v.) knockdown animals showed an impairment of memory processes. This detrimental effect was temporary and reversible, disappearing 7 d after the end of the aODN treatment. At the highest effective doses, none of the compounds used impaired motor coordination, as revealed by the rota rod test, nor modified spontaneous mobility and inspection activity, as revealed by the hole-board test. In conclusion, the lack of any involvement of cerebral RyR1 was demonstrated. These findings also showed the involvement of type 2 and type 3 RyR in the modulation of memory functions identifying these cerebral RyR isoforms as critical targets underlying memory processes.


Assuntos
Ventrículos Cerebrais/efeitos dos fármacos , Cresóis/efeitos adversos , Transtornos da Memória/induzido quimicamente , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Cresóis/administração & dosagem , Immunoblotting , Injeções , Transtornos da Memória/fisiopatologia , Camundongos , Oligonucleotídeos Antissenso/genética , Isoformas de Proteínas/efeitos dos fármacos , Transtornos Psicomotores/induzido quimicamente , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Fatores de Tempo
13.
J Membr Biol ; 222(2): 65-77, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18418540

RESUMO

Ryanodine receptor 2 (RyR2) cDNA has been available for more than 15 years; however, due to the complex nature of ligand gating in this channel, many aspects of recombinant RyR2 function have been unresearched. We established a stable, inducible HEK 293 cell line expressing full-length rabbit RyR2 cDNA and assessed the single-channel properties of the recombinant RyR2, with particular reference to ligand regulation with Ca2+ as the permeant ion. We found that the single-channel conductances of recombinant RyR2 and RyR2 isolated from cardiac muscle are essentially identical, as is irreversible modification by ryanodine. Although it is known that RyR2 expressed in HEK 293 cells is not associated with FKBP12.6, we demonstrate that these channels do not exhibit any discernable disorganized gating characteristics or subconductance states. We also show that the gating of recombinant RyR2 is indistinguishable from that of channels isolated from cardiac muscle when activated by cytosolic Ca2+, caffeine or suramin. The mechanisms underlying ATP activation are also similar; however, the experiments highlighted a novel effect of ATP at physiologically relevant concentrations of 5-10 mM. With Ca2+ as permeant ion, 5-10 mM ATP consistently inactivated recombinant channels (15/16 experiments). Such inactivation was rarely observed with native RyR2 isolated from cardiac muscle (1 in 16 experiments). However, if the channels were purified, inactivation by ATP was then revealed in all experiments. This action of ATP may be relevant for inactivation of sarcoplasmic reticulum Ca2+ release during cardiac excitation-contraction coupling or may represent unnatural behavior that is revealed when RyR2 is purified or expressed in noncardiac systems.


Assuntos
Trifosfato de Adenosina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cafeína/farmacologia , Cálcio/metabolismo , Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , DNA Complementar/genética , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ligantes , Coelhos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Suramina/farmacologia
14.
Curr Pharm Des ; 13(31): 3195-211, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18045169

RESUMO

Sudden cardiac death (SCD) remains a major cause of mortality, and despite our knowledge of the causative genetic, molecular and biochemical cellular mechanisms involved, effective therapeutic strategies are lacking. Perturbations in cardiac Ca2+ handling promote arrhythmias and there is enormous interest in developing new anti-arrhythmics aimed at correcting Ca2+ release dysfunction. In particular, abnormal Ca2+ release arising as a result of acquired or genetic defects in cardiac ryanodine receptors (RyR2) has emerged as an important arrhythmogenic trigger in heart failure, and in a devastating genetic arrhythmia syndrome termed catecholaminergic polymorphic ventricular tachycardia (CPVT). Here, we evaluate how experimental insights into RyR2 structure-function are unravelling the precise molecular basis of channel dysfunction and are advancing the development of new therapeutic strategies. We also discuss the functional role of RyR2 in the context of the exquisite synergism existing between numerous cellular components involved in cardiac Ca2+ signalling, and how these complex interactions may be used to design new anti-arrhythmic approaches that target multiple facets of RyR2 regulation.


Assuntos
Antiarrítmicos/administração & dosagem , Morte Súbita Cardíaca/etiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Antiarrítmicos/farmacologia , Sinalização do Cálcio/fisiologia , Catecolaminas/metabolismo , Ensaios Clínicos como Assunto , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Humanos , Polimorfismo Genético , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Taquicardia Ventricular/genética , Taquicardia Ventricular/fisiopatologia
15.
Biochem Biophys Res Commun ; 354(4): 929-36, 2007 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-17276394

RESUMO

Aconitine is an effective ingredient in Aconite tuber, an important traditional Chinese medicine. Aconitine is also known to be a highly toxic diterpenoid alkaloid with arrhythmogenic effects. In the present study, we have characterized the properties of arrhythmic cytotoxicity and explored the possible mechanisms of aconitine-induced cardiomyocytes. Results show that aconitine induces significant abnormity in the spontaneous beating rate, amplitude of spontaneous oscillations and the relative intracellular Ca(2+) concentration. Also, mRNA transcription levels and protein expressions of SR Ca(2+) release channel RyR(2) and sarcolemmal NCX were elevated in aconitine-induced cardiomyocytes. However, co-treatment with ruthenium red (RR), a RyR channel inhibitor, could reverse the aconitine-induced abnormity in intracellular Ca(2+) signals. These results demonstrate that disruption of intracellular Ca(2+) homeostasis in the cardiac excitation-contraction coupling (EC coupling) is a crucial mechanism of arrhythmic cytotoxicity in aconitine-induced cardiomyocytes. Moreover, certain inhibitors appear to play an important role in the detoxification of aconitine-induced Ca(2+)-dependent arrhythmias.


Assuntos
Aconitina/toxicidade , Arritmias Cardíacas/induzido quimicamente , Cálcio/fisiologia , Homeostase/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Frequência Cardíaca/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Rutênio Vermelho/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/biossíntese , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , Trocador de Sódio e Cálcio/biossíntese , Tapsigargina/farmacologia
16.
Am J Physiol Cell Physiol ; 292(5): C1591-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17182726

RESUMO

Malignant hyperthermia (MH) is a pharmacogenetic disorder of skeletal muscle triggered in susceptible individuals by inhalation anesthetics and depolarizing skeletal muscle relaxants. This syndrome has been linked to a missense mutation in the type 1 ryanodine receptor (RyR1) in more than 50% of cases studied to date. Using double-barreled Ca(2+) microelectrodes in myotubes expressing wild-type RyR1 ((WT)RyR1) or RyR1 with one of four common MH mutations ((MH)RyR1), we measured resting intracellular Ca(2+) concentration ([Ca(2+)](i)). Changes in resting [Ca(2+)](i) produced by several drugs known to modulate the RyR1 channel complex were investigated. We found that myotubes expressing any of the (MH)RyR1s had a 2.0- to 3.7-fold higher resting [Ca(2+)](i) than those expressing (WT)RyR1. Exposure of myotubes expressing (MH)RyR1s to ryanodine (500 microM) or (2,6-dichloro-4-aminophenyl)isopropylamine (FLA 365; 20 microM) had no effects on their resting [Ca(2+)](i). However, when myotubes were exposed to bastadin 5 alone or to a combination of ryanodine and bastadin 5, the resting [Ca(2+)](i) was significantly reduced (P < 0.01). Interestingly, the percent decrease in resting [Ca(2+)](i) in myotubes expressing (MH)RyR1s was significantly greater than that for (WT)RyR1. From these data, we propose that the high resting myoplasmic [Ca(2+)](i) in (MH)RyR1 expressing myotubes is due in part to a related structural conformation of (MH)RyR1s that favors "passive" calcium leak from the sarcoplasmic reticulum.


Assuntos
Cálcio/metabolismo , Hipertermia Maligna/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Regulação Alostérica , Animais , Linhagem Celular , DNA Complementar , Éteres Difenil Halogenados , Humanos , Eletrodos Seletivos de Íons , Hipertermia Maligna/genética , Microeletrodos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Mutação , Fenetilaminas/farmacologia , Éteres Fenílicos/farmacologia , Conformação Proteica/efeitos dos fármacos , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Retículo Sarcoplasmático/efeitos dos fármacos , Transfecção
17.
Anesth Analg ; 99(1): 103-107, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15281512

RESUMO

Malignant hyperthermia (MH) is an inherited skeletal muscle disorder triggered by commonly used anesthetics. Mutated ryanodine receptors have been identified as molecular targets. The sensitivity of myotubes from individuals classified by the in vitro contracture test as MH susceptible (MHS), normal (MHN), and equivocal (MHEH) was assessed for the Ca2+-releasing activity of 4-chloro-m-cresol (4-CmC) and caffeine. In this study, we sought to determine whether 4-CmC can differentiate the MH status of an individual on the basis of the release of intracellular Ca2+, particularly in regard to MHEH diagnosis. Intracellular Ca2+ concentration was determined photometrically with Fura2. Regions of the ryanodine receptor 1 harboring most of the described MH mutations were sequenced from MHS and MHEH cells. One MH mutation (Gly2434Arg) was found in one MHS individual. Results of the caffeine-induced Ca2+ release in MHS and MHN cells correlated well with the in vitro contracture test results. MHS cells showed a higher sensitivity against caffeine and, to a lesser extent, against 4-CmC. Cells of MHEH individuals showed low sensitivities against both caffeine and 4-CmC, comparable to those of the MHN group. Therefore, with myotubes, caffeine was able to discriminate between MHS and MHN cells, but both caffeine and 4-CmC failed to detect MHEH cells.


Assuntos
Cresóis , Hipertermia Maligna/diagnóstico , Hipertermia Maligna/patologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Cafeína/farmacologia , Cálcio/metabolismo , Calibragem , Células Cultivadas , Estimulantes do Sistema Nervoso Central/farmacologia , Análise Mutacional de DNA , DNA Complementar/genética , Corantes Fluorescentes , Fura-2 , Humanos , Hipertermia Maligna/genética , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Contração Muscular/efeitos dos fármacos , RNA Mensageiro/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos
18.
Jpn J Pharmacol ; 89(1): 21-8, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12083739

RESUMO

We determined the effect of 9-hydroxyellipticine (9HE) on ryanodine receptor (RyR) and cardiac function after global ischemia in isolated rat hearts. The binding of [3H]-ryanodine in rabbit cardiac sarcoplasmic reticulum was displaced by 9HE in a biphasic manner corresponding to the two sites model with IC50 values of 6.1 microM and 55 mM. The increase of the intracellular Ca2+ concentration induced by caffeine in CHO cells expressing cardiac-type RyR was suppressed by 9HE in a concentration-dependent manner. Pretreatment of the heart with 9HE decreased the total duration of reperfusion-induced ventricular fibrillation (VF) and delayed the onset of VF. There was also a significant recovery of contractile force of ischemic hearts following 9HE. Unlike nifedipine, an L-type Ca2+-channel blocker, 9HE did not suppress the contraction of rat papillary muscles. Thus, 9HE exerts the cardioprotective effects against ischemia /reperfusion injury without changing hemodynamic indices.


Assuntos
Antiarrítmicos/uso terapêutico , Arritmias Cardíacas/prevenção & controle , Elipticinas/uso terapêutico , Isquemia Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Antiarrítmicos/farmacologia , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Ligação Competitiva , Células CHO , Cálcio/metabolismo , Cricetinae , Elipticinas/farmacologia , Técnicas In Vitro , Masculino , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Nifedipino/farmacologia , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Fatores de Tempo
19.
Biophys J ; 82(5): 2428-35, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11964231

RESUMO

Although an elevation in myoplasmic Ca2+ can activate the skeletal muscle ryanodine receptor (RyR1), the function of this Ca2+ activation is unclear because extracellular Ca2+ influx is unnecessary for skeletal-type EC coupling. To determine whether Ca2+ activation of RyR1 is necessary for the initiation of skeletal-type EC coupling, we examined the behavior of RyR1 with glutamate 4032 mutated to alanine (E4032A-RyR1) because this mutation had been shown to dramatically reduce activation by Ca2+. Proc. Natl. Acad. Sci. USA. 98:2865-2870). Analysis after reconstitution into planar lipid bilayers revealed that E4032A-RyR1 was negligibly activated by 100 microM Ca2+ (P(o) too low to be measured). Even in the presence of both 2 mM caffeine and 2 mM ATP, P(o) remained low for E4032A-RyR1 (ranging from <0.0001 in 100 microM free Ca2+ to 0.005 in 2 mM free Ca2+). Thus, the E4032A mutation caused a nearly complete suppression of activation of RyR1 by Ca2+. Depolarization of E4032A-RyR1-expressing myotubes elicited L-type Ca2+ currents of approximately normal size and myoplasmic Ca2+ transients that were skeletal-type, but about fivefold smaller than those for wild-type RyR1. The reduced amplitude of the Ca2+ transient is consistent either with the possibility that Ca2+ activation amplifies Ca2+ release during EC coupling, or that the E4032A mutation generally inhibits activation of RyR1. In either case, Ca2+ activation of RyR1 does not appear to be necessary for the initiation of Ca2+ release during EC coupling in skeletal muscle.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/fisiologia , Contração Muscular/fisiologia , Músculo Esquelético/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Trifosfato de Adenosina/metabolismo , Substituição de Aminoácidos , Animais , Cafeína/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , DNA Complementar , Bicamadas Lipídicas/química , Mamíferos , Músculo Esquelético/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética
20.
J Med Chem ; 42(14): 2527-34, 1999 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10411473

RESUMO

Caffeine and other methylxanthines are known to induce Ca(2+)-release from intracellular stores via the ryanodine receptor. In the present work, a range of caffeine analogues, in which methyl groups at the 1 and 7 positions were replaced with alkyl chains containing different functional groups (oxo, hydroxyl, propargyl, ester, and acids), were synthesized. These compounds were then screened for their ability to potentiate Ca(2+)-release induced by cADPR (an endogenous modulator of ryanodine receptors) in sea urchin egg homogenates. Two of the synthesized methylxanthines, 1, 3-dimethyl-7-(7-hydroxyoctyl)xanthine (37) and 3-methyl-7-(7-oxooctyl)-1-propargylxanthine (66), were shown to be more potent than caffeine in potentiating cADPR-induced Ca(2+)-release, while 1,3-dimethyl-7-(5-ethylcarboxypentyl)xanthine (14) was shown to be more efficacious. The development of new methylxanthine analogues may lead to a better understanding of ryanodine receptor function and could possibly provide novel therapeutic agents.


Assuntos
Adenosina Difosfato Ribose/análogos & derivados , Cafeína/análogos & derivados , Cálcio/metabolismo , Xantinas/síntese química , Adenosina Difosfato Ribose/metabolismo , Animais , Cafeína/síntese química , Cafeína/química , Cafeína/farmacologia , ADP-Ribose Cíclica , Avaliação Pré-Clínica de Medicamentos , Técnicas In Vitro , Óvulo/efeitos dos fármacos , Óvulo/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Ouriços-do-Mar , Relação Estrutura-Atividade , Xantinas/química , Xantinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA