Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Life Sci ; 90(15-16): 607-11, 2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-22406079

RESUMO

AIMS: In a previous study we found that A-935142 enhanced hERG current in a concentration-dependent manner by facilitating activation, reducing inactivation, and slowing deactivation (Su et al., 2009). A-935142 also shortened action potential duration (APD90) in canine Purkinje fibers and guinea pig atrial tissue. This study focused on the combined effects of the prototypical hERG enhancer, A-935142 and two hERG current blockers (sotalol and terfenadine). MAIN METHODS: The whole-cell voltage clamp method with HEK 293 cells heterologously expressing the hERG channel (Kv 11.1) was used. KEY FINDINGS: A-935142 did not compete with 3H-dofetilide binding, suggesting that A-935142 does not overlap the binding site of typical hERG blockers. In whole-cell voltage clamp studies we found: 1) 60 µM A-935142 enhanced hERG current in the presence of 150 µM sotalol (57.5±5.8%) to a similar extent as seen with A-935142 alone (55.6±5.1%); 2) 150 µM sotalol blocked hERG current in the presence of 60 µM A-935142 (43.5±1.5%) to a similar extent as that seen with sotalol alone (42.0±3.2%) and 3) during co-application, hERG current enhancement was followed by current blockade. Similar results were obtained with 60 nM terfenadine combined with A-935142. SIGNIFICANCE: These results suggest that the hERG enhancer, A-935142 does not compete with these two known hERG blockers at their binding site within the hERG channel. This selective hERG current enhancement may be useful as a treatment for inherited or acquired LQTS (Casis et al., 2006).


Assuntos
Acetatos/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Pirazóis/metabolismo , Análise de Variância , Células HEK293 , Humanos , Técnicas de Patch-Clamp , Fenetilaminas/metabolismo , Sotalol/farmacologia , Sulfonamidas/metabolismo , Terfenadina/farmacologia , Trítio
2.
Inflamm Res ; 60(7): 683-93, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21394563

RESUMO

OBJECTIVE: The aim of this study was to compare a diverse set of peptide and small-molecule calcium channel blockers for inactivated-state block of native and recombinant N-type calcium channels using fluorescence-based and automated patch-clamp electrophysiology assays. METHODS: The pharmacology of calcium channel blockers was determined at N-type channels in IMR-32 cells and in HEK cells overexpressing the inward rectifying K(+) channel Kir2.1. N-type channels were opened by increasing extracellular KCl. In the Kir2.1/N-type cell line the membrane potential could be modulated by adjusting the extracellular KCl, allowing determination of resting and inactivated-state block of N-type calcium channels. The potency and degree of state-dependent inhibition of these blockers were also determined by automated patch-clamp electrophysiology. RESULTS: N-type-mediated calcium influx in IMR-32 cells was determined for a panel of blockers with IC(50) values of 0.001-7 µM and this positively correlated with inactivated-state block of recombinant channels measured using electrophysiology. The potency of several compounds was markedly weaker in the state-dependent fluorescence-based assay compared to the electrophysiology assay, although the degree of state-dependent blockade was comparable. CONCLUSIONS: The present data demonstrate that fluorescence-based assays are suitable for assessing the ability of blockers to selectively interact with the inactivated state of the N-type channel.


Assuntos
Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio Tipo N/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo N/farmacologia , Linhagem Celular , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp
3.
J Pharmacol Toxicol Methods ; 64(1): 68-73, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21440075

RESUMO

INTRODUCTION: Delayed cardiac repolarization is an established risk factor for proarrhythmia and Torsades-de-Pointes (TdeP) that is typically measured in vitro during slow, regular stimulation. We have developed an alternative, novel, and rapid cellular-based approach for predicting drug-induced proarrhythmia that detects changes in electrical refractoriness based on mechanical responses (measured optically) during increasingly rapid trains of stimulation interspersed with pauses (mimicking the clinically observed short-long-short (SLS) stimulation sequence associated with the TdeP initiation). METHODS: Acutely isolated rabbit ventricular myocytes were superfused and electrically stimulated using an accelerating pacing protocol (APP) consisting of 12 consecutive pacing segments (10 beats per segment) with incrementally faster cycle lengths; trains were separated by pauses to identify loss of stimulus capture as well as to mimic clinically observed SLS sequences. Drug effects were evaluated based on a myocyte's ability to contract during progressively faster pacing segments (rate-adaptation); the earliest rate during which the myocyte fails to respond (longest cycle length with incomplete capture (CLIC)) was used to quantify electrophysiologic effects. RESULTS: Torsadogenic drugs known to delay repolarization during slow stimulation prolonged CLIC and dramatically limited the ability to respond to progressively rapid stimulation. The recognized proarrhythmic compounds E-4031, cisapride, grepafloxacin, and haloperidol rapidly prolonged CLIC at and above therapeutic concentrations in a concentration-dependent manner, while negative controls (captopril, indomethacin, and loratidine) do not affect rate-adaptation. DISCUSSION: Ventricular rate adaptation represents a novel approach for rapidly detecting drugs with torsadogenic risk using rapid rhythms that are typically not employed when evaluating proarrhythmic risk. This method is well suited for detecting and avoiding potential cardiac liabilities early in drug discovery ("frontloading") prior to final selection of candidate drugs.


Assuntos
Eletrocardiografia/efeitos dos fármacos , Coração/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Torsades de Pointes/induzido quimicamente , Torsades de Pointes/diagnóstico , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Estimulação Elétrica/métodos , Feminino , Coração/fisiopatologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Coelhos , Torsades de Pointes/fisiopatologia
4.
Curr Drug Saf ; 6(5): 277-84, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22424534

RESUMO

A-955840, a selective CB2 agonist, has been shown to elicit concentration-dependent decreases in cardiac contractility in the anesthetized dog (decreased maximal velocity of left ventricular pressure development [LV dP/dt max]). However, it is unknown whether this represents a direct effect or a response dependent on other factors (such as autonomic tone and neurohumoral factors) present in vivo. This study examined if A-955840 had a direct effect on contractility of isolated cardiac myocytes, and if so to determine the potential mechanisms. Contractility was assessed in vitro using percent changes in maximal shortening velocity of sarcomeres (dL/dt max) and fractional shortening of sarcomere length (FS) in rabbit left ventricular myocytes. L-type calcium current in myocytes was recorded using wholecell voltage-clamp techniques. A-955840 reduced dL/dt max and FS in a reversible and concentration-dependent manner with an IC50 of 11.4 µg/mL (based on dL/dt max) which is similar to the estimated IC50 value of 9.8 µg/mL based on the effects of A-955840 on LV dP/dt max in anesthetized dogs. A-955840 (4.1 µg/mL) reduced myocyte contractility (%FS) to a similar extent in the absence and presence of a CB2 antagonist, SR-2 (24.0 ± 3.4 vs 23.1 ± 3.0 %, n=5) or a CB1 antagonist, Rimonabant (18.8 ± 2.3 vs 19.8 ± 2.7 %, n=5). A-955840 (4.1 µg/mL) also reduced L-type calcium current of rabbit ventricular myocytes (1.05 ± 0.11 vs 0.70 ± 0.12 nA, n=5, P < 0.01). These results suggest that A-955840 exerts direct negative inotropic effects on isolated rabbit ventricular myocytes, which is mediated by neither CB2 nor CB1 receptors, and consistent with off-target negative inotropy mediated by inhibition of the cardiac L-type calcium current.


Assuntos
Benzamidas/farmacologia , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Receptor CB2 de Canabinoide/agonistas , Tiazóis/farmacologia , Animais , Benzamidas/administração & dosagem , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/metabolismo , Cães , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Concentração Inibidora 50 , Masculino , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Pirazóis/farmacologia , Coelhos , Receptor CB1 de Canabinoide/metabolismo , Rimonabanto , Sarcômeros/metabolismo , Tiazóis/administração & dosagem
5.
Biochem Pharmacol ; 80(7): 1000-6, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20599796

RESUMO

N'1-(3,3,6,8-tetramethyl-1-oxo-1,2,3,4-tetrahydronaphthalen-2-yliden)-2-cyanoethanohydrazide (TTYC) increases secretion of glucagon-like peptide-1 and intracellular Ca(2+) concentration in GLUTag cells. The purpose of the present study was to examine if TTYC exerts positive inotropic effects on isolated rabbit ventricular myocytes and in vivo heart in anesthetized rats, and if so to further define the potential mechanism of action. Contractility was assessed in vitro using changes in fractional shortening (FS) of myocyte sarcomere length and in vivo using changes in the velocity of left ventricular pressure. Changes in L-type Ca(2+) current of ventricular myocytes were evaluated using whole-cell voltage-clamp techniques. TTYC increased FS of myocyte sarcomere length in a concentration-dependent manner. The positive inotropic effect was not abrogated by beta-adrenergic blockade (propranolol) or protein kinase A inhibition. TTYC enhanced peak L-type Ca(2+) current in a voltage-dependent manner (current amplitudes increased by 4.0-fold at -10 mV and 1.5-fold at +10 mV). Voltage-dependence of steady-state activation of L-type Ca(2+) current was shifted by 15 mV in the negative direction. Inactivation time course of the L-type Ca(2+) currents at voltages of -10 to 20 mV was significantly slowed by 0.3 microM TTYC. In vivo studies demonstrated that TTYC increased cardiac contractility in a dose-dependent manner. In conclusion, TTYC is a novel L-type Ca(2+) current activator with positive cardiac inotropic effects. Negative shifting of the voltage-dependence of L-type Ca(2+) current activation and reduced inactivation are two mechanisms responsible for the enhanced L-type Ca(2+) current that contribute to the positive inotropic effects.


Assuntos
Cálcio/metabolismo , Contração Miocárdica/efeitos dos fármacos , Animais , Cálcio/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Coração/efeitos dos fármacos , Masculino , Células Musculares/metabolismo , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Coelhos , Ratos , Ratos Sprague-Dawley
6.
J Cardiovasc Pharmacol ; 49(5): 269-74, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17513944

RESUMO

Delayed cardiac repolarization and fatal proarrhythmia have been linked to block of the repolarizing current, Ikr or hERG (human ether-a-go-go related gene) current. Thus, determining the potency of hERG block is critical in evaluating cardiac safety during preclinical development. Hydroxypropyl beta-cyclodextrins (HbetaC) are cyclic oligosaccharides used to enhance drug solubility. To evaluate the utility of HbetaC to enhance drug solubility in hERG screening assays, we studied the effect of HbetaC on hERG current and the sensitivity of the hERG assay to 3 structurally different hERG blocking drugs using whole-cell voltage clamp technique and HEK-293 cells expressing the hERG channel. HbetaC inhibited hERG activation and tail current and accelerated current deactivation in a concentration-dependent manner. HbetaC (6%) reduced the apparent potency of block by terfenadine (IC50 12000 nM vs 45 nM), cisapride (IC50 281 nM vs 28 nM), and E-4031 (163 nM vs 26 nM). Reduced potency of block was consistent with loss of activity as a result of complexation with HbetaC by terfenadine and cisapride (demonstrated in solubility studies) and interactions with HbetaC by E-4031 (demonstrated in absorbance studies). These results demonstrate that HbetaC is an unsuitable agent for enhancing compound solubility in the in vitro hERG current assay and may mask drug effects, allowing potentially dangerous drugs to advance into clinical development.


Assuntos
Bioensaio , Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Excipientes/farmacologia , beta-Ciclodextrinas/farmacologia , 2-Hidroxipropil-beta-Ciclodextrina , Antiarrítmicos/farmacologia , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/fisiopatologia , Cisaprida/farmacologia , Relação Dose-Resposta a Droga , Eletrofisiologia , Fármacos Gastrointestinais/farmacologia , Antagonistas dos Receptores Histamínicos H1/efeitos adversos , Antagonistas dos Receptores Histamínicos H1/farmacologia , Humanos , Concentração Inibidora 50 , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Piperidinas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Piridinas/farmacologia , Solubilidade/efeitos dos fármacos , Terfenadina/efeitos adversos , Terfenadina/farmacologia , Fatores de Tempo
7.
Bioorg Med Chem Lett ; 17(8): 2365-71, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17350253

RESUMO

A series of potent 2-carboxychromone-based melanin-concentrating hormone receptor 1 (MCHr1) antagonists were synthesized and evaluated for hERG (human Ether-a-go-go Related Gene) channel affinity and functional blockade. Basic dialkylamine-terminated analogs were found to weakly bind the hERG channel and provided marked improvement in a functional patch-clamp assay versus previously reported antagonists of the series.


Assuntos
Amidas/farmacologia , Cromonas/farmacologia , Canais de Potássio Éter-A-Go-Go/metabolismo , Receptores do Hormônio Hipofisário/antagonistas & inibidores , Animais , Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Camundongos , Obesidade/tratamento farmacológico , Técnicas de Patch-Clamp , Farmacocinética
8.
J Pharmacol Toxicol Methods ; 56(1): 11-7, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17350293

RESUMO

INTRODUCTION: Drug-induced long QT syndrome (LQTS) has been linked to arrhythmias (including Torsades de Pointes and sudden cardiac death), and has led to an increased awareness of the potential risk of delayed repolarization in vitro and in vivo. However, in vitro assessments of delayed repolarization have not been fully predictive of in vivo effects. METHODS: To define the extent to which plasma protein binding (ppb) contributes to such disparities in repolarization studies, we compared drug-induced prolongation of the canine Purkinje fiber action potential duration (APD(90)) in vitro during superfusion with 100% Tyrode's solution (Tyrodes), canine plasma [50% plasma/50% Tyrodes] and a 5% solution of recombinant human serum albumin in Tyrodes (HSA). Drugs evaluated included cisapride (>98% ppb), risperidone (90% ppb), and d, l-sotalol (negligible ppb). Effects on APD were monitored using standard microelectrode techniques under physiologic conditions and temperature ([K(+)]=4 mM, 37 degrees C) during slow stimulation (2 s basic cycle length). RESULTS: The effects of cisapride and risperidone on Purkinje fiber APD(90) were significantly attenuated in the presence of plasma proteins. However, with cisapride, the extent of reduction with plasma proteins was significantly less than predicted based on calculated free drug levels. DISCUSSION: We conclude that while plasma protein binding does reduce APD prolongation seen with bound drugs, this effect is not well correlated with the calculated plasma protein binding or expected clinical free fraction. Because of the complex drug interactions that occur in plasma, the electrophysiological effects seen with bound drugs are not well correlated with the calculated free fraction and thus caution should be exercised when assigning a predictive safety window. Thus, the canine Purkinje fiber assay is useful for defining the modulation of delayed repolarization due to plasma protein binding of novel therapeutic agents.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Proteínas Sanguíneas/metabolismo , Cisaprida/metabolismo , Ramos Subendocárdicos/efeitos dos fármacos , Risperidona/metabolismo , Sotalol/metabolismo , Animais , Cisaprida/efeitos adversos , Cães , Humanos , Técnicas In Vitro , Soluções Isotônicas , Modelos Biológicos , Ligação Proteica , Ramos Subendocárdicos/fisiologia , Risperidona/efeitos adversos , Albumina Sérica/metabolismo , Albumina Sérica/farmacologia , Sotalol/efeitos adversos
9.
J Cardiovasc Pharmacol ; 48(5): 199-206, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17110801

RESUMO

Terodiline and tolterodine are drugs used to treat urinary incontinence. Terodiline was removed from the market in 1991 for proarrhythmia, whereas tolterodine has a generally benign clinical cardiac profile. To assess differences in the electrophysiologic actions of these drugs, we evaluated their effects on hERG current (HEK cells) and cardiac Purkinje fiber repolarization. The IC50 for hERG block (37 degrees C) by tolterodine was 9.6 nM and by terodiline was 375 nM, values near or below clinical concentrations. Tolterodine elicited concentration-dependent prolongation of the action potential duration (APD90). In contrast, terodiline depressed the action potential plateau and induced triangulation without affecting APD90. The triangulation ratios (normalized ratio of APD50 over APD90) for terodiline were 0.94 and 0.59 for 1.0 and 10 microM and for tolterodine, were 0.99 and 0.97 at 7 and 70 nM. In summary, tolterodine, a potent hERG blocker, has a benign clinical cardiac profile at therapeutic concentrations that may be due to its lack of triangulation, as well as extensive plasma protein binding. However, at supratherapeutic concentrations, preclinical data predict risk of QT prolongation. These data suggest that hERG block and triangulation are among multiple factors that must be considered in preclinical cardiac safety assessments.


Assuntos
Compostos Benzidrílicos/farmacologia , Butilaminas/farmacologia , Cresóis/farmacologia , Coração/efeitos dos fármacos , Fenilpropanolamina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Linhagem Celular , Cães , Avaliação Pré-Clínica de Medicamentos , Coração/fisiologia , Tartarato de Tolterodina
10.
J Med Chem ; 49(22): 6569-84, 2006 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-17064075

RESUMO

Evaluation of multiple structurally distinct series of melanin concentrating hormone receptor 1 antagonists in an anesthetized rat cardiovascualar assay led to the identification of a chromone-2-carboxamide series as having excellent safety against the chosen cardiovascular endpoints at high drug concentrations in the plasma and brain. Optimization of this series led to considerable improvements in affinity, functional potency, and pharmacokinetic profile. This led to the identification of a 7-fluorochromone-2-carboxamide (22) that was orally efficacious in a diet-induced obese mouse model, retained a favorable cardiovascular profile in rat, and demonstrated dramatic improvement in effects on mean arterial pressure in our dog cardiovascular model compared to other series reported by our group. However, this analogue also led to prolongation of the QT interval in the dog that was linked to affinity for hERG channel and unexpectedly potent functional blockade of this ion channel.


Assuntos
Benzodioxóis/farmacologia , Doenças Cardiovasculares/induzido quimicamente , Cromonas/farmacologia , Receptores de Somatostatina/antagonistas & inibidores , Acilação , Animais , Área Sob a Curva , Benzodioxóis/farmacocinética , Benzodioxóis/toxicidade , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Cálcio/metabolismo , Linhagem Celular , Cromonas/farmacocinética , Cromonas/toxicidade , Cães , Eletrocardiografia/efeitos dos fármacos , Feminino , Meia-Vida , Frequência Cardíaca/efeitos dos fármacos , Indicadores e Reagentes , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
11.
Clin Exp Pharmacol Physiol ; 33(11): 1059-65, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17042915

RESUMO

1. Drug-induced block of the rapidly activating delayed rectifier K+ current (I(Kr)), encoded by human ether-a-go-go-related gene (hERG), has been linked to acquired long QT syndrome (aLQTS). Hypokalaemia is a recognized risk factor in aLQTS. To further understand why hypokalaemia is a risk factor in aLQTS, we examined the effect of [K+]o on drug block of the hERG potassium channel stably expressed in human embryonic kidney (HEK-293) cells using whole-cell voltage-clamp techniques. 2. The effects of selected [K+]o (1-20 mmol/L) on hERG block with four structurally diverse compounds (dofetilide, mesoridazine, quinidine and terfenadine) from different therapeutic classes were evaluated. Reducing [K+]o from 20 to 1 mmol/L had little effect on IC50 values for hERG current block for all four compounds. For example, evaluating quinidine in external potassium concentrations of 20, 10, 5 and 1 mmol/L resulted in IC50 values of 1.82 +/- 0.33, 2.04 +/- 0.28, 1.57 +/- 0.52 and 1.14 +/- 0.21 mmol/L, respectively. No statistically significant difference (P > 0.35, anova) was observed between drug block of hERG in different external potassium concentrations. These data are in contrast with previously reported results examining hERG channel modulation expressed in AT-1 cells under similar experimental conditions. 3. These results demonstrate that [K+]o does not directly modulate drug block of hERG channels expressed in an HEK-293 cell line. The enhanced risk of Torsades de Pointes associated with hypokalaemia in aLQTS may be due to reduction of other (non-hERG) potassium currents, further reducing the repolarization reserve, and not due to direct modulation of hERG block by [K+]o.


Assuntos
Canais de Potássio Éter-A-Go-Go/metabolismo , Rim/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Potássio/farmacologia , Linhagem Celular , Canal de Potássio ERG1 , Humanos , Rim/citologia , Mesoridazina/farmacologia , Fenetilaminas/farmacologia , Quinidina/farmacologia , Sulfonamidas/farmacologia , Terfenadina/farmacologia
12.
Comb Chem High Throughput Screen ; 6(5): 481-8, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12871054

RESUMO

A biased chemical library containing 91 differentially substituted thiazolidinones was prepared in an effort to improve the pharmacology of a known anticonvulsant agent V102862. The collection was prepared in a single step multi-component condensation reaction that produced good yields and very high crude purity (75%-85%). Seven compounds, identified within the library were shown to be more potent than V102862, our parent reference compound, in an electrophysiological assay measuring sodium channel antagonism. The most potent compound, 3-(2-piperidinylethyl)-2-(3-(3-trifluoromethylphenoxy)phenyl)thiazolidinone, has a Ki of 90 nM.


Assuntos
Bloqueadores dos Canais de Sódio/síntese química , Bloqueadores dos Canais de Sódio/farmacologia , Animais , Técnicas de Química Combinatória , Feminino , Potenciais da Membrana/efeitos dos fármacos , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...