Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Cardiovasc Med ; 9: 956538, 2022.
Article in English | MEDLINE | ID: mdl-35990966

ABSTRACT

Background: The phosphoinositide 3-kinase (PI3K) signaling pathway is an interesting target in cancer treatment. The awareness of the proarrhythmic risk of PI3K inhibitors was raised because PI3K is also involved in regulating signaling toward cardiac ion channels. Canine cardiomyocytes treated with PI3K inhibitors show an increased action potential duration and reduced cardiac repolarizing currents. Now, the potential proarrhythmic effect of chronic treatment of PI3K/mTOR inhibitor GSK2126458 (omipalisib) was investigated in the atrioventricular (AV) block dog model. Methods: Purpose-bred Mongrel dogs received complete AV block by ablation of the bundle of His and their hearts were paced in the right ventricular apex at VDD-mode (RVA-VDD). In this way, sinus rhythm was maintained for 15 ± 1 days and thereby bradycardia-induced cardiac remodeling was prevented. Dogs received 1 mg/kg omipalisib once (n = 3) or twice (n = 10) a day via oral administration for 7 days. Under standardized conditions (anesthesia, bradycardia at 60 beats/min, and a dofetilide challenge), potential proarrhythmic effects of omipalisib were investigated. Results: Twice daily dosing of omipalisib increased accumulative plasma levels compared to once daily dosing accompanied with adverse events. Omipalisib prolonged the QT interval at baseline and more strongly after the dofetilide challenge (490 ± 37 to 607 ± 48 ms). The arrhythmic outcome after omipalisib resulted in single ectopic beats in 30% of dogs perpetuating in multiple ectopic beats and TdP arrhythmia in 20% of dogs. Isolated ventricular cardiomyocytes from omipalisib-treated dogs showed a diminished IKs current density. Conclusion: Chronic treatment of PI3K/mTOR inhibitor omipalisib prolonged the QT interval in a preclinical model under standardized proarrhythmic conditions. Furthermore, this study showed that electrical remodeling induced by omipalisib had a mild proarrhythmic outcome.

2.
Br J Pharmacol ; 169(6): 1322-34, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23586323

ABSTRACT

BACKGROUND AND PURPOSE: Drug interference with normal hERG protein trafficking substantially reduces the channel density in the plasma membrane and thereby poses an arrhythmic threat. The chemical substructures important for hERG trafficking inhibition were investigated using pentamidine as a model drug. Furthermore, the relationship between acute ion channel block and correction of trafficking by dofetilide was studied. EXPERIMENTAL APPROACH: hERG and K(IR)2.1 trafficking in HEK293 cells was evaluated by Western blot and immunofluorescence microscopy after treatment with pentamidine and six pentamidine analogues, and correction with dofetilide and four dofetilide analogues that displayed different abilities to inhibit IKr . Molecular dynamics simulations were used to address mode, number and type of interactions between hERG and dofetilide analogues. KEY RESULTS: Structural modifications of pentamidine differentially affected plasma membrane levels of hERG and K(IR)2.1. Modification of the phenyl ring or substituents directly attached to it had the largest effect, affirming the importance of these chemical residues in ion channel binding. PA-4 had the mildest effects on both ion channels. Dofetilide corrected pentamidine-induced hERG, but not K(IR)2.1 trafficking defects. Dofetilide analogues that displayed high channel affinity, mediated by pi-pi stacks and hydrophobic interactions, also restored hERG protein levels, whereas analogues with low affinity were ineffective. CONCLUSIONS AND IMPLICATIONS: Drug-induced trafficking defects can be minimized if certain chemical features are avoided or 'synthesized out'; this could influence the design and development of future drugs. Further analysis of such features in hERG trafficking correctors may facilitate the design of a non-blocking corrector for trafficking defective hERG proteins in both congenital and acquired LQTS.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Ether-A-Go-Go Potassium Channels/metabolism , Membrane Transport Modulators/pharmacology , Pentamidine/pharmacology , Phenethylamines/pharmacology , Potassium Channel Blockers/pharmacology , Shab Potassium Channels/metabolism , Sulfonamides/pharmacology , Animals , Anti-Arrhythmia Agents/chemistry , Antiprotozoal Agents/adverse effects , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/metabolism , Cell Membrane/drug effects , Dogs , ERG1 Potassium Channel , Endocytosis/drug effects , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/genetics , HEK293 Cells , Humans , Kinetics , Membrane Transport Modulators/adverse effects , Membrane Transport Modulators/chemistry , Mice , Molecular Dynamics Simulation , Pentamidine/adverse effects , Pentamidine/analogs & derivatives , Pentamidine/chemistry , Phenethylamines/chemistry , Potassium Channel Blockers/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Shab Potassium Channels/chemistry , Shab Potassium Channels/genetics , Structure-Activity Relationship , Sulfonamides/chemistry
3.
Br J Pharmacol ; 165(2): 467-78, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21718297

ABSTRACT

BACKGROUND AND PURPOSE: Drug development requires the testing of new chemical entities for adverse effects. For cardiac safety screening, improved assays are urgently needed. Isolated adult cardiomyocytes (CM) and human embryonic stem cell-derived cardiomyocytes (hESC-CM) could be used to identify pro-arrhythmic compounds. In the present study, five assays were employed to investigate their sensitivity and specificity for evaluating the pro-arrhythmic properties of I(Kr) blockers, using moxifloxacin (safe compound) and dofetilide or E-4031 (unsafe compounds). EXPERIMENTAL APPROACH: Assays included the anaesthetized remodelled chronic complete AV block (CAVB) dog, the anaesthetized methoxamine-sensitized unremodelled rabbit, multi-cellular hESC-CM clusters, isolated CM obtained from CAVB dogs and isolated CM obtained from the normal rabbit. Arrhythmic outcome was defined as Torsade de Pointes (TdP) in the animal models and early afterdepolarizations (EADs) in the cell models. KEY RESULTS: At clinically relevant concentrations (5-12 µM), moxifloxacin was free of pro-arrhythmic properties in all assays with the exception of the isolated CM, in which 10 µM induced EADs in 35% of the CAVB CM and in 23% of the rabbit CM. At supra-therapeutic concentrations (≥100 µM), moxifloxacin was pro-arrhythmic in the isolated rabbit CM (33%), in the hESC-CM clusters (18%), and in the methoxamine rabbit (17%). Dofetilide and E-4031 induced EADs or TdP in all assays (50-83%), and the induction correlated with a significant increase in beat-to-beat variability of repolarization. CONCLUSION AND IMPLICATIONS: Isolated cardiomyocytes lack specificity to discriminate between TdP liability of the I(Kr) blocking drugs moxifloxacin and dofetilide or E4031.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Aza Compounds/pharmacology , Myocytes, Cardiac/drug effects , Phenethylamines/pharmacology , Piperidines/pharmacology , Potassium Channel Blockers/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology , Sulfonamides/pharmacology , Torsades de Pointes/chemically induced , Action Potentials/drug effects , Animals , Cell Line , Disease Models, Animal , Dogs , Embryonic Stem Cells/cytology , Female , Fluoroquinolones , Heart/drug effects , Heart/physiopathology , Heart Block/physiopathology , Humans , Methoxamine , Moxifloxacin , Myocytes, Cardiac/physiology , Rabbits , Torsades de Pointes/physiopathology , Ventricular Remodeling/drug effects
4.
Acta Physiol (Oxf) ; 199(3): 243-56, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20331539

ABSTRACT

Inward rectifier currents based on K(IR)2.x subunits are regarded as essential components for establishing a stable and negative resting membrane potential in many excitable cell types. Pharmacological inhibition, null mutation in mice and dominant positive and negative mutations in patients reveal some of the important functions of these channels in their native tissues. Here we review the complex mammalian expression pattern of K(IR)2.x subunits and relate these to the outcomes of functional inhibition of the resultant channels. Correlations between expression and function in muscle and bone tissue are observed, while we recognize a discrepancy between neuronal expression and function.


Subject(s)
Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/physiology , Animals , Humans , Mice , Mice, Knockout , Mutation/physiology , Phenotype , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/biosynthesis , Potassium Channels, Inwardly Rectifying/drug effects
5.
Br J Pharmacol ; 159(7): 1532-41, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20180941

ABSTRACT

BACKGROUND AND PURPOSE: Pentamidine is a drug used in treatment of protozoal infections. Pentamidine treatment may cause sudden cardiac death by provoking cardiac arrhythmias associated with QTc prolongation and U-wave alterations. This proarrhythmic effect was linked to inhibition of hERG trafficking, but not to acute block of ion channels contributing to the action potential. Because the U-wave has been linked to the cardiac inward rectifier current (I(K1)), we examined the action and mechanism of pentamidine-mediated I(K1) block. EXPERIMENTAL APPROACH: Patch clamp measurements of I(K1) were made on cultured adult canine ventricular cardiomyocytes, K(IR)2.1-HEK293 cells and K(IR)2.x inside-out patches. Pentamidine binding to cytoplasmic amino acid residues of K(IR)2.1 channels was studied by molecular modelling. KEY RESULTS: Pentamidine application (24 h) decreased I(K1) in cultured canine cardiomyocytes and K(IR)2.1-HEK293 cells under whole cell clamp conditions. Pentamidine inhibited I(K1) in K(IR)2.1-HEK293 cells 10 min after application. When applied to the cytoplasmic side under inside-out patch clamp conditions, pentamidine block of I(K1) was acute (IC(50)= 0.17 microM). Molecular modelling predicted pentamidine-channel interactions in the cytoplasmic pore region of K(IR)2.1 at amino acids E224, D259 and E299. Mutation of these conserved residues to alanine reduced pentamidine block of I(K1). Block was independent of the presence of spermine. K(IR)2.2, and K(IR)2.3 based I(K1) was also sensitive to pentamidine blockade. CONCLUSIONS AND IMPLICATIONS: Pentamidine inhibits cardiac I(K1) by interacting with three negatively charged amino acids in the cytoplasmic pore region. Our findings may provide new insights for development of specific I(K1) blocking compounds.


Subject(s)
Antiprotozoal Agents/pharmacology , Cytoplasm/drug effects , Pentamidine/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Animals , Blotting, Western , Cell Line , Cytoplasm/metabolism , Dogs , Humans , Mutation , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...