Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 172(12): 3112-25, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25684549

ABSTRACT

BACKGROUND AND PURPOSE: We aimed to characterize the pharmacology and electrophysiology of N-[3-(1H-benzimidazol-2-yl)-4-chloro-phenyl]pyridine-3-carboxamide (AZSMO-23), an activator of the human ether-a-go-go-related gene (hERG)-encoded K(+) channel (Kv 11.1). EXPERIMENTAL APPROACH: Automated electrophysiology was used to study the pharmacology of AZSMO-23 on wild-type (WT), Y652A, F656T or G628C/S631C hERG, and on other cardiac ion channels. Its mechanism of action was characterized with conventional electrophysiology. KEY RESULTS: AZSMO-23 activated WT hERG pre-pulse and tail current with EC50 values of 28.6 and 11.2 µM respectively. At 100 µM, pre-pulse current at +40 mV was increased by 952 ± 41% and tail current at -30 mV by 238 ± 13% compared with vehicle values. The primary mechanism for this effect was a 74.5 mV depolarizing shift in the voltage dependence of inactivation, without any shift in the voltage dependence of activation. Structure-activity relationships for this effect were remarkably subtle, with close analogues of AZSMO-23 acting as hERG inhibitors. AZSMO-23 blocked the mutant channel, hERG Y652A, but against another mutant channel, hERG F656T, its activator activity was enhanced. It inhibited activity of the G628C/S631C non-inactivating hERG mutant channel. AZSMO-23 was not hERG selective, as it blocked hKv 4.3-hKChIP2.2, hCav 3.2 and hKv 1.5 and activated hCav 1.2/ß2/α2δ channels. CONCLUSION AND IMPLICATIONS: The activity of AZSMO-23 and those of its close analogues suggest these compounds may be of value to elucidate the mechanism of type 2 hERG activators to better understand the pharmacology of this area from both a safety perspective and in relation to treatment of congenital long QT syndrome.


Subject(s)
Benzimidazoles/pharmacology , Electrophysiological Phenomena/drug effects , Ether-A-Go-Go Potassium Channels/drug effects , Ion Channels/drug effects , Niacinamide/analogs & derivatives , Animals , Benzimidazoles/chemistry , CHO Cells , Cell Line , Cricetinae , Cricetulus , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Ion Channels/metabolism , Niacinamide/chemistry , Niacinamide/pharmacology , Structure-Activity Relationship
2.
Toxicol Appl Pharmacol ; 279(2): 87-94, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24952337

ABSTRACT

Cardiovascular-related adverse drug effects are a major concern for the pharmaceutical industry. Activity of an investigational drug at the L-type calcium channel could manifest in a number of ways, including changes in cardiac contractility. The aim of this study was to define which of the two assay technologies - radioligand-binding or automated electrophysiology - was most predictive of contractility effects in an in vitro myocyte contractility assay. The activity of reference and proprietary compounds at the L-type calcium channel was measured by radioligand-binding assays, conventional patch-clamp, automated electrophysiology, and by measurement of contractility in canine isolated cardiac myocytes. Activity in the radioligand-binding assay at the L-type Ca channel phenylalkylamine binding site was most predictive of an inotropic effect in the canine cardiac myocyte assay. The sensitivity was 73%, specificity 83% and predictivity 78%. The radioligand-binding assay may be run at a single test concentration and potency estimated. The least predictive assay was automated electrophysiology which showed a significant bias when compared with other assay formats. Given the importance of the L-type calcium channel, not just in cardiac function, but also in other organ systems, a screening strategy emerges whereby single concentration ligand-binding can be performed early in the discovery process with sufficient predictivity, throughput and turnaround time to influence chemical design and address a significant safety-related liability, at relatively low cost.


Subject(s)
Calcium Channels, L-Type/drug effects , Drug Discovery/methods , High-Throughput Screening Assays , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Toxicity Tests/methods , Animals , Automation , Binding Sites , CHO Cells , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cricetinae , Cricetulus , Dogs , Female , Humans , Ligands , Membrane Potentials , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Predictive Value of Tests , Protein Binding , Radioligand Assay , Risk Assessment , Transfection
3.
Toxicol Appl Pharmacol ; 260(2): 162-72, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22373797

ABSTRACT

Measurement of cardiac contractility is a logical part of pre-clinical safety assessment in a drug discovery project, particularly if a risk has been identified or is suspected based on the primary- or non-target pharmacology. However, there are limited validated assays available that can be used to screen several compounds in order to identify and eliminate inotropic liability from a chemical series. We have therefore sought to develop an in vitro model with sufficient throughput for this purpose. Dog ventricular myocytes were isolated using a collagenase perfusion technique and placed in a perfused recording chamber on the stage of a microscope at ~36 °C. Myocytes were stimulated to contract at a pacing frequency of 1 Hz and a digital, cell geometry measurement system (IonOptix™) was used to measure sarcomere shortening in single myocytes. After perfusion with vehicle (0.1% DMSO), concentration-effect curves were constructed for each compound in 4-30 myocytes taken from 1 or 2 dog hearts. The validation test-set was 22 negative and 8 positive inotropes, and 21 inactive compounds, as defined by their effect in dog, cynolomolgous monkey or humans. By comparing the outcome of the assay to the known in vivo contractility effects, the assay sensitivity was 81%, specificity was 75%, and accuracy was 78%. With a throughput of 6-8 compounds/week from 1 cell isolation, this assay may be of value to drug discovery projects to screen for direct contractility effects and, if a hazard is identified, help identify inactive compounds.


Subject(s)
Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Animals , Dogs , Drug Discovery/methods , Female , Heart Ventricles/drug effects , In Vitro Techniques , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Reproducibility of Results , Sarcomeres/physiology , Sensitivity and Specificity , Video Recording
4.
Am J Physiol Heart Circ Physiol ; 302(7): H1466-80, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22198175

ABSTRACT

Cell lines expressing ion channels (IC) and the advent of plate-based electrophysiology device have enabled a molecular understanding of the action potential (AP) as a means of early QT assessment. We sought to develop an in silico AP (isAP) model that provides an assessment of the effect of a compound on the myocyte AP duration (APD) using concentration-effect curve data from a panel of five ICs (hNav1.5, hCav1.2, hKv4.3/hKChIP2.2, hKv7.1/hminK, hKv11.1). A test set of 53 compounds was selected to cover a range of selective and mixed IC modulators that were tested for their effects on optically measured APD. A threshold of >10% change in APD at 90% repolarization (APD(90)) was used to signify an effect at the top test concentration. To capture the variations observed in left ventricular midmyocardial myocyte APD data from 19 different dogs, the isAP model was calibrated to produce an ensemble of 19 model variants that could capture the shape and form of the APs and also quantitatively replicate dofetilide- and diltiazem-induced APD(90) changes. Provided with IC panel data only, the isAP model was then used, blinded, to predict APD(90) changes greater than 10%. At a simulated concentration of 30 µM and based on a criterion that six of the variants had to agree, isAP prediction was scored as showing greater than 80% predictivity of compound activity. Thus, early in drug discovery, the isAP model allows integrating separate IC data and is amenable to the throughput required for use as a virtual screen.


Subject(s)
Action Potentials/physiology , Cardiovascular Agents/pharmacology , Cardiovascular Agents/toxicity , Computer Simulation , Drug-Related Side Effects and Adverse Reactions , Heart/physiology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calibration , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , ERG1 Potassium Channel , Electrodes, Implanted , Electrophysiological Phenomena , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Female , Fluorescence , Fluorescent Dyes , Myocytes, Cardiac/physiology , Potassium Channel Blockers/pharmacology , Risk Assessment , Threshold Limit Values
5.
Br J Pharmacol ; 164(2b): 419-32, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21449979

ABSTRACT

BACKGROUND AND PURPOSE: The majority of human ether-a-go-go-related gene (hERG) screens aiming to minimize the risk of drug-induced long QT syndrome have been conducted using heterologous systems expressing the hERG 1a subunit, although both hERG 1a and 1b subunits contribute to the K+ channels producing the repolarizing current I(Kr) . We tested a range of compounds selected for their diversity to determine whether hERG 1a and 1a/1b channels exhibit different sensitivities that may influence safety margins or contribute to a stratified risk analysis. EXPERIMENTAL APPROACH: We used the IonWorks™ plate-based electrophysiology device to compare sensitivity of hERG 1a and 1a/1b channels stably expressed in HEK293 cells to 50 compounds previously shown to target hERG channels. Potency was determined as IC50 values (µM) obtained from non-cumulative, eight-point concentration-effect curves of normalized data, fitted to the Hill equation. To minimize possible sources of variability, compound potency was assessed using test plates arranged in alternating columns of cells expressing hERG 1a and 1a/1b. KEY RESULTS: Although the potency of most compounds was similar for the two targets, some surprising differences were observed. Fluoxetine (Prozac) was more potent at blocking hERG 1a/1b than 1a channels, yielding a corresponding reduction in the safety margin. In contrast, E-4031 was a more potent blocker of hERG 1a compared with 1a/1b channels, as previously reported, as was dofetilide, another high-affinity blocker. CONCLUSIONS AND IMPLICATIONS: The current assays may underestimate the risk of some drugs to cause torsades de pointes arrhythmia, and overestimate the risk of others.


Subject(s)
Drug Evaluation, Preclinical/methods , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Fluoxetine/pharmacology , Action Potentials/drug effects , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/genetics , Cell Line, Transformed , Ether-A-Go-Go Potassium Channels/genetics , HEK293 Cells , Humans , Inhibitory Concentration 50 , Long QT Syndrome/chemically induced , Long QT Syndrome/genetics , Piperidines/pharmacology , Protein Subunits , Pyridines/pharmacology , Sensitivity and Specificity , Torsades de Pointes/chemically induced , Torsades de Pointes/genetics
6.
Br J Pharmacol ; 159(1): 12-21, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20141516

ABSTRACT

Owing to its association with Torsades de Pointes, drug-induced QT interval prolongation has been and remains a significant hurdle to the development of safe, effective medicines. Genetic and pharmacological evidence highlighting the pivotal role the human ether-a-go-go-related gene (hERG) channel was a critical step in understanding how to start addressing this issue. It led to the development of hERG assays with the rapid throughput needed for the short timescales required in early drug discovery. The resulting volume of hERG data has fostered in silico models to help chemists design compounds with reduced hERG potency. In early drug discovery, a pragmatic approach based on exceeding a given potency value has been required to decide when a compound is likely to carry a low QT risk, to support its progression to late-stage discovery. At this point, the in vivo efficacy and metabolism characteristics of the potential drug are generally defined, as well its safety profile, which includes usually a dog study to assess QT interval prolongation risk. The hERG and in vivo QT data, combined with the likely indication and the estimated free drug level for efficacy, are put together to assess the risk that the potential drug will prolong QT in man. Further data may be required to refine the risk assessment before making the major investment decisions for full development. The non-clinical data are essential to inform decisions about compound progression and to optimize the design of clinical QT studies.


Subject(s)
Drug Design , Ether-A-Go-Go Potassium Channels/metabolism , Long QT Syndrome/chemically induced , Animals , Dogs , Drug-Related Side Effects and Adverse Reactions , Humans , Risk Assessment/methods , Torsades de Pointes/chemically induced
7.
J Pharmacol Toxicol Methods ; 57(1): 30-41, 2008.
Article in English | MEDLINE | ID: mdl-17980627

ABSTRACT

INTRODUCTION: The safety implications of blocking the human cardiac Na(+) channel (hNav1.5) make it prudent to test for this activity early in the drug discovery process and design-out any potential liability. This needs a method with adequate throughput and a demonstrable predictive value to effects in native cardiac tissues. Here we describe the validation of a method that combines the ability to screen tens of compounds a day, with direct assessment of channel function. METHODS: The electrophysiological and pharmacological properties of hNav1.5 were compared using two methods: conventional, low-throughput electrophysiology and planar-array-based, medium-throughput electrophysiology (IonWorks HT). A pharmacological comparison was also made between IonWorks HT and canine cardiac Purkinje Fibre action potential upstroke data. RESULTS: Activation curve parameters for hNav1.5 in IonWorks HT were not statistically different (p>0.05) from those generated using conventional electrophysiology. IonWorks HT V(1/2)=-22+/-0.8 mV, slope=6.9+/-0.2 (n=11); conventional electrophysiology V(1/2)=-20+/-1.6 mV, slope=6.4+/-0.3 (n=11). Potency values for a range of hNav1.5 blockers determined using IonWorks HT correlated closely with those obtained using conventional electrophysiology (R=0.967, p<0.001). The assay was able to distinguish between highly use-dependent blockers (e.g. tetracaine) and blockers that do not display strong use-dependence (e.g. quinidine). Comparison of the degree of hNav1.5 inhibition and decrease in canine Purkinje fibre action potential upstroke velocity (V(max)) showed that the IonWorks HT assay would have predicted the outcome in Purkinje fibres in the majority of cases, with false negative and positive rates estimated at 8 and 7%, respectively. Finally, hNav1.5 pharmacology was similar when determined using either IonWorks HT or IonWorks Quattro, although the latter yielded more consistent data. DISCUSSION: The assay described combines a functional assessment of hNav1.5 with medium-throughput. Furthermore the assay was able to reveal information on the use-dependency of compound block, as well as predicting Na(+) channel effects in more integrated systems such as the cardiac Purkinje fibre action potential. This makes it possible to determine quantitative potency data, and mechanistic information about use-dependence, in a timeframe short enough to influence medicinal chemistry.


Subject(s)
Drug Evaluation, Preclinical/methods , Electrophysiology , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/metabolism , Sodium Channels/metabolism , Animals , Biophysical Phenomena , Biophysics , CHO Cells , Cricetinae , Cricetulus , Dogs , Dose-Response Relationship, Drug , Female , Humans , Inhibitory Concentration 50 , Male , Membrane Potentials , NAV1.5 Voltage-Gated Sodium Channel , Predictive Value of Tests , Purkinje Fibers/drug effects , Reproducibility of Results , Sodium Channel Blockers/pharmacology
8.
J Pharmacol Toxicol Methods ; 54(2): 189-99, 2006.
Article in English | MEDLINE | ID: mdl-16563806

ABSTRACT

INTRODUCTION: Regulatory and competitive pressure to reduce the QT interval prolongation risk of potential new drugs has led to focus on methods to test for inhibition of the human ether-a-go-go-related gene (hERG)-encoded K+ channel, the primary molecular target underlying this safety issue. Here we describe the validation of a method that combines medium-throughput with direct assessment of channel function. METHODS: The electrophysiological and pharmacological properties of hERG were compared using two methods: conventional, low-throughput electrophysiology and planar-array-based, medium-throughput electrophysiology (IonWorks HT). A pharmacological comparison was also made between IonWorks HT and an indirect assay (Rb+ efflux). RESULTS: Basic electrophysiological properties of hERG were similar whether recorded conventionally (HEK cells) or using IonWorks HT (CHO cells): for example, tail current V1/2 -12.1+/-5.0 mV (32) for conventional and -9.5+/-6.0 mV (46) for IonWorks HT (mean+/-S.D. (n)). A key finding was that as the number of cells per well was increased in IonWorks HT, the potency reported for a given compound decreased. Using the lowest possible cell concentration (250,000 cells/ml) and 89 compounds spanning a broad potency range, the pIC50 values from IonWorks HT (CHO-hERG) were found to correlate well with those obtained using conventional methodology (HEK-hERG)(r=0.90; p<0.001). Further validation using CHO-hERG cells with both methods confirmed the correlation (r=0.94; p<0.001). In contrast, a comparison of IonWorks HT and Rb+ efflux data with 649 compounds using CHO-hERG cells showed that the indirect assay consistently reported compounds as being, on average, 6-fold less potent, though the differences varied depending on chemical series. DISCUSSION: The main finding of this work is that providing a relatively low cell concentration is used in IonWorks HT, the potency information generated correlates well with that determined using conventional electrophysiology. The effect on potency of increasing cell concentration may relate to a reduced free concentration of test compound owing to partitioning into cell membranes. In summary, the IonWorks HT hERG assay can generate pIC50 values based on a direct assessment of channel function in a timeframe short enough to influence chemical design.


Subject(s)
Electrophysiology/instrumentation , Ether-A-Go-Go Potassium Channels/drug effects , Patch-Clamp Techniques/instrumentation , Potassium Channel Blockers/pharmacology , Animals , CHO Cells , Cell Line , Cricetinae , ERG1 Potassium Channel , Humans , Reproducibility of Results , Rubidium/metabolism
9.
J Physiol ; 531(Pt 1): 117-30, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11179396

ABSTRACT

1. The role of the cGMP pathway in the modulation of the cardiac L-type Ca2+ current (ICa,L) by nitric oxide (NO) was examined in rat ventricular myocytes. 2. The NO donors DEANO, SIN-1, SNP, SNAP and GSNO had no significant effects on basal ICa,L. However, DEANO (100 microM) inhibited ICa,L after the current had been previously stimulated by either isoprenaline (Iso, 1-10 nM), a beta-adrenergic agonist, or isobutylmethyl-xanthine (IBMX, 10-80 microM), a wide spectrum phosphodiesterase (PDE) inhibitor. 3. The anti-adrenergic effect of DEANO on ICa,L was not mimicked by other NO donors (SIN-1, SNAP and SPNO). 4. The NO-sensitive guanylyl cyclase inhibitor ODQ (10 microM), antagonized the inhibitory effect of DEANO on ICa,L. Likewise, inhibitors of the cGMP-dependent protein kinase (cG-PK), Rp-8-chloro-phenylthio-cGMP (10 microM) and KT5823 (0.1 and 0.3 microM), also abolished the inhibitory effect of DEANO on Iso (1-10 nM)-stimulated ICa,L. 5. Intracellular dialysis with exogenous cAMP (10-100 microM) blunted the inhibitory effect of DEANO (10 and 100 microM) on ICa,L. SNAP and SNP also had no effect on the cAMP-stimulated ICa,L. 6. Pre-treatment of the myocytes with pertussis toxin (0.5 microg ml-1, 4-6 h at 37 degrees C) eliminated the inhibitory effect of DEANO (100 microM) on ICa,L, in the presence of either Iso (0.01 and 1 nM) or IBMX (10-80 microM). 7. These results demonstrate that DEANO produces anti-adrenergic effects in rat ventricular myocytes. This effect of DEANO occurs in a cGMP-dependent manner, and involves activation of cG-PK and regulation of a pertussis toxin-sensitive G protein.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , GTP-Binding Proteins/physiology , Heart/drug effects , Nitric Oxide Donors/pharmacology , Animals , Electrophysiology , Guanylate Cyclase/antagonists & inhibitors , Heart Ventricles/cytology , Heart Ventricles/drug effects , In Vitro Techniques , Male , Membrane Potentials/physiology , Microdialysis , Myocardium/cytology , Patch-Clamp Techniques , Rats , Rats, Wistar
10.
J Physiol ; 523 Pt 2: 377-89, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10699082

ABSTRACT

1. The biophysical and pharmacological characteristics of the hyperpolarization activated non- selective cation current (If) were recorded using whole-cell voltage clamp in embryonic stem (ES) cell-derived cardiomyocytes at different stages of development. 2. The cation current was detected in a large percentage (65 %) of early stage (EDS, differentiated for 7 + 3-4 days) cells at a current density of 11.4 +/- 0.6 pA pF-1 (n = 47). In late stage (LDS, differentiated for 7 + 9-12 days) cells the percentage of cells expressing If decreased (45 %), but If densities (15.5 +/- 0.9 pA pF-1, n = 20) were increased. 3. The muscarinic agonist carbachol (CCh, 1 microM) depressed basal If in EDS cells by 45.7 +/- 6.5 %, n = 5) and was without effect in LDS cardiomyocytes (n = 4). The beta-adrenoceptor agonist isoprenaline (ISO, 1 microM) stimulated If in LDS cells by 33 +/- 5.2 % (n = 6) but not in EDS cells (n = 5). 4. Cell infusion with the catalytic subunit of the cAMP-dependent protein kinase (PKA, 7 microM) stimulated If in EDS cells by 37.0 +/- 2.9 %, (n = 4), but subsequent superfusion of 8-bromo-cAMP (200 microM) was without effect. Intracellular perfusion of LDS cardiomyocytes with the highly selective peptide inhibitor of PKA (PKI, 20 microM) completely inhibited the stimulation of the L-type Ca2+ current (ICa,L) as well as of If by ISO (1 microM). 5. Extracellular superfusion with phosphodiesterase (PDE) inhibitors - IBMX, a non-selective antagonist, Erythro-9-(2-hydoxy-3-nonyl)adenine (EHNA), a PDE2 antagonist and rolipram, a PDE4 antagonist - resulted in stimulation of ICa,L and If in EDS cells. By contrast, milrinone and cilostamide, two PDE3 antagonists, stimulated ICa,L, but not If. 6. The present work demonstrates that If is functionally expressed during early cardiomyogenesis. Similar to ICa,L, If is regulated during embryonic development by phosphorylation via PKA. In contrast to ICa,L, If is not regulated by PDE3 suggesting different localization of these ion channels with respect to PDE3.


Subject(s)
Cations/metabolism , Heart/embryology , Ion Channels/biosynthesis , Myocardium/metabolism , Nerve Tissue Proteins , Stem Cells/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Cell Differentiation/physiology , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/pharmacology , Cyclic Nucleotide-Gated Cation Channels , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/agonists , Ion Channels/antagonists & inhibitors , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Muscarinic Agonists/pharmacology , Myocardium/cytology , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation/drug effects , Potassium Channels , Stem Cells/cytology
11.
Am J Respir Crit Care Med ; 160(4): 1196-204, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10508807

ABSTRACT

We report that in vivo injection of endotoxin (EDTX, 6 mg. kg(-)(1)) induces cardiovascular alterations in rats that closely mimic the clinical situation, as assessed by in vivo hemodynamic measurements in anesthetized and conscious, chronically instrumented animals. The patch-clamp technique was used to characterize the L-type calcium current (I(Ca)) and its autonomic regulation in isolated cardiac myocytes. The density of I(Ca) progressively decreased at 12 and 36 h after EDTX injection. However, the dihydropyridine (+/-)Bay K 8644 (100 nM) enhanced I(Ca) to levels similar to those in control and EDTX-treated myocytes. In addition, the net stimulatory effect of a beta-adrenergic agonist (isoproterenol) on I(Ca) was increased 12 h after EDTX injection. This change in the beta-adrenergic effect declined 24 h later. The potentiation in the beta-adrenergic stimulation of I(Ca) was mimicked by L858051 (10 microM), a direct activator of adenylyl cyclase, but not by IBMX (200 microM), a phosphodiesterase inhibitor. Besides, the antiadrenergic effect of acetylcholine on I(Ca) was unchanged 12 h after EDTX injection, but increased 36 h after EDTX injection. These results support the hypothesis that time-dependent changes in the adenylyl cyclase pathway in cardiac myocytes may contribute, via the autonomic regulation of I(Ca), to the severity of myocardial dysfunction during sepsis.


Subject(s)
Autonomic Nervous System/physiology , Endotoxins/pharmacology , Heart/innervation , Myocardium/cytology , 1-Methyl-3-isobutylxanthine/pharmacology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Acetylcholine/pharmacology , Acetylcholine/physiology , Adenylyl Cyclases/metabolism , Adrenergic Antagonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Calcium Channel Agonists/pharmacology , Calcium Channels, L-Type/metabolism , Colforsin/analogs & derivatives , Colforsin/pharmacology , Diterpenes , Electrophysiology , Endotoxemia/physiopathology , Endotoxins/administration & dosage , Enzyme Activators/pharmacology , Escherichia coli , Heart/physiology , Hemodynamics , In Vitro Techniques , Injections, Intravenous , Isoproterenol/pharmacology , Male , Myocardium/metabolism , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Rats , Sepsis/physiopathology
12.
Mol Pharmacol ; 52(3): 482-90, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9281611

ABSTRACT

We studied the mechanism of action of methylene blue (Mblue), a putative guanylyl cyclase inhibitor, on the L-type calcium current (ICa) and the muscarinic activated K+ current (IK,ACh) in rat ventricular and atrial myocytes, respectively, and on the binding of [3H]quinuclidinyl benzylate in rat ventricular membranes. Superfusion, but not internal dialysis, with 30 microM Mblue antagonized the inhibitory effect of acetylcholine (ACh, 1 microM) on beta-adrenergic stimulation of ICa with isoprenaline (Iso, 10 nM or 1 microM). However, Mblue had no effect on the basal ICa or on the stimulation of ICa by Iso in the absence of ACh. The activation of IK,ACh by 3 microM ACh was also antagonized by Mblue in a dose-dependent manner. In contrast, Mblue had no effect on the activation of IK,ACh by either guanosine-5'-O-(3-thio)triphosphate or guanosine-5'-(beta,gamma-imido)triphosphate. Chlorpromazine (CPZ), a piperazine derivative like Mblue, also inhibited the muscarinic activation of IK,ACh in a dose-dependent manner. The specific binding of [3H]QNB, a muscarinic ligand, to rat ventricular membranes was displaced in a dose-dependent manner by Mblue and CPZ. The piperazine derivatives behaved like competitive antagonists of [3H]QNB binding, exhibiting equilibrium dissociation constant (Ki) values of 187 nM for Mblue and 366 nM for CPZ. In conclusion, Mblue exerts antimuscarinic effects on ICa and IK,ACh in rat cardiac myocytes that are best explained by the binding of Mblue to the M2 subtype of muscarinic receptors. This property probably contributes to the antimuscarinic effect of the putative guanylyl cyclase inhibitor reported in previous studies.


Subject(s)
Heart/drug effects , Methylene Blue/pharmacology , Muscarinic Antagonists/pharmacology , Myocardium/ultrastructure , Acetylcholine/metabolism , Acetylcholine/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Calcium Channels/drug effects , Calcium Channels/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membranes/metabolism , Myocardium/cytology , Myocardium/metabolism , Potassium Channels/drug effects , Potassium Channels/physiology , Quinuclidinyl Benzilate/metabolism , Rats , Rats, Wistar , Receptors, Muscarinic/metabolism , Tritium
13.
Br J Pharmacol ; 121(7): 1369-77, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9257916

ABSTRACT

1. To investigate the participation of guanylyl cyclase in the muscarinic regulation of the cardiac L-type calcium current (ICa), we examined the effects of three guanylyl cyclase inhibitors, 1H-[1,2,4]oxidiazo-lo[4,3-a]quinoxaline-1-one (ODQ), 6-anilino-5,8-quinolinedione (LY 83583), and methylene blue (MBlue), on the beta-adrenoceptor; muscarinic receptor and nitric oxide (NO) regulation of ICa and on the muscarinic activated potassium current I(K,ACh), in frog atrial and ventricular myocytes. 2. ODQ (10 microM) and LY 83583 (30 microM) antagonized the inhibitory effect of an NO-donor (S-nitroso-N-acetylpenicillamine, SNAP, 1 microM) on the isoprenaline (Iso)-stimulated ICa which was consistent with their inhibitory action on guanylyl cyclase. However, MBlue (30 microM) had no effect under similar conditions. 3. In the absence of SNAP, LY 83583 (30 microM) potentiated the stimulations of ICa by either Iso (20 nM), forskolin (0.2 microM) or intracellular cyclic AMP (5-10 microM). ODQ (10 microM) had no effect under these conditions, while MBlue (30 microM) inhibited the Iso-stimulated ICa. 4. LY 83583 and MBlue, but not ODQ, reduced the inhibitory effect of up to 10 microM acetylcholine (ACh) on ICa. 5. MBlue, but not LY 83583 and ODQ, antagonized the activation of I(K,ACh) by ACh in the presence of intracellular GTP, and this inhibition was weakened when I(K,ACh) was activated by intracellular GTPgammaS. 6. The potentiating effect of LY 83583 on Iso-stimulated ICa was absent in the presence of either DL-dithiothreitol (DTT, 100 microM) or a combination of superoxide dismutase (150 u ml(-1)) and catalase (100 u ml(-1)). 7. All together, our data demonstrate that, among the three compounds tested, only ODQ acts in a manner which is consistent with its inhibitory action on the NO-sensitive guanylyl cyclase. The two other compounds produced severe side effects which may involve superoxide anion generation in the case of LY 83583 and alteration of beta-adrenoceptor and muscarinic receptor-coupling mechanisms in the case of M Blue.


Subject(s)
Calcium Channels/drug effects , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Heart/drug effects , Potassium Channels/drug effects , Aminoquinolines/pharmacology , Animals , Calcium Channels, L-Type , Cyclic AMP/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Methylene Blue/pharmacology , Nitric Oxide/physiology , Nitroprusside/pharmacology , Rana esculenta , Receptors, Adrenergic, beta/physiology , Receptors, Muscarinic/physiology , Superoxides/metabolism
14.
Life Sci ; 60(13-14): 1113-20, 1997.
Article in English | MEDLINE | ID: mdl-9121355

ABSTRACT

Muscarinic agonists regulate the L-type calcium current in isolated cardiac myocytes. The second messengers pathways involved in this regulation are discussed briefly, with particular emphasis on the involvement of cAMP and cGMP pathways.


Subject(s)
Calcium Channels/drug effects , Heart/drug effects , Muscarinic Agonists/pharmacology , Animals , Cyclic AMP/biosynthesis , Cyclic GMP/biosynthesis , Humans , Nitric Oxide/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...