Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
1.
Acta Physiol (Oxf) ; 221(3): 157-159, 2017 11.
Article in English | MEDLINE | ID: mdl-28613434
2.
Herz ; 41(8): 677-683, 2016 Dec.
Article in German | MEDLINE | ID: mdl-27847992

ABSTRACT

The first European Society of Cardiology (ESC) guidelines on atrial fibrillation (AF) developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS) were published in August 2016. These guidelines replace the revised guidelines from 2012 and contain some interesting new aspects. The topics range from the pathophysiology through diagnostics, therapy and stroke prevention up to special clinical situations, such as atrial fibrillation in cardiopathy, sport and pregnancy. Early screening, patient informed consent, individualized therapy and the modification of factors promoting atrial fibrillation are of particular importance. The guidelines recommend the establishment of AF heart teams, containing specialists from various disciplines. The guidelines also underline the importance of non-vitamin K­dependent oral anticoagulants (NOAC) for stroke prevention compared to standard anticoagulants with vitamin K antagonists. For symptomatic and especially paroxysmal atrial fibrillation, the guidelines emphasize the importance of an antiarrhythmic treatment with catheter ablation and/or pharmaceutical antiarrhythmic therapy in addition to a frequency regulating therapy.


Subject(s)
Atrial Fibrillation/diagnosis , Atrial Fibrillation/therapy , Cardiology/standards , Diagnostic Techniques, Cardiovascular/standards , Practice Guidelines as Topic , Stroke/prevention & control , Anticoagulants/administration & dosage , Atrial Fibrillation/complications , Cardiac Pacing, Artificial , Catheter Ablation/standards , Europe , Evidence-Based Medicine/standards , Guideline Adherence/standards , Humans , Stroke/diagnosis , Stroke/etiology
3.
Int J Cardiol ; 203: 22-9, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26490502

ABSTRACT

Atrial fibrillation (AF) is the most frequently encountered cardiac arrhythmia. The trigger for initiation of AF is generally an enhanced vulnerability of pulmonary vein cardiomyocyte sleeves to either focal or re-entrant activity. The maintenance of AF is based on a "driver" mechanism in a vulnerable substrate. Cardiac mapping technology is providing further insight into these extremely dynamic processes. AF can lead to electrophysiological and structural remodelling, thereby promoting the condition. The management includes prevention of stroke by oral anticoagulation or left atrial appendage (LAA) occlusion, upstream therapy of concomitant conditions, and symptomatic improvement using rate control and/or rhythm control. Nonpharmacological strategies include electrical cardioversion and catheter ablation. There are substantial geographical variations in the management of AF, though European data indicate that 80% of patients receive adequate anticoagulation and 79% adequate rate control. High rates of morbidity and mortality weigh against perceived difficulties in management. Clinical research and growing experience are helping refine clinical indications and provide better technical approaches. Active research in cardiac electrophysiology is producing new antiarrhythmic agents that are reaching the experimental clinical arena, inhibiting novel ion channels. Future research should give better understanding of the underlying aetiology of AF and identification of drug targets, to help the move toward patient-specific therapy.


Subject(s)
Atrial Fibrillation/physiopathology , Atrial Fibrillation/therapy , Global Health , Humans
4.
Basic Res Cardiol ; 110(2): 5, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25600224

ABSTRACT

The sustained component of the K(+) outward current in human atrial myocytes is believed to be due to the slowly inactivating ultra-rapid potassium current I Kur and not to the fast inactivating transient outward current Ito. Here we provide evidence for contribution of Ito to this late current due to the effects of dipeptidyl peptidase-like protein (DPP) 10 (DPP10a) interacting with Kv4.3 channels. We studied the late current component of Ito in human atrial myocytes and CHO cells co-expressing Kv4.3 or Kv4.3/KChIP2 (control) and DPP proteins using voltage-clamp technique and a pharmacological approach. A voltage dependent and slowly inactivating late current (43% of peak amplitude) could be observed in atrial myocytes. We found a similar current in CHO cells expressing Kv4.3/KChIP2 + DPP10a, but not in cells co-expressing Kv4.3 + DPP or Kv4.3/KChIP2 + DPP6-S. Assuming that DPP10a influences atrial Ito, we detected DPP10 expression of three alternatively spliced mRNAs, DPP10 protein and colocalization of Kv4.3 and DPP10 proteins in human atrial myocytes. DPP10a did not affect properties of expressed Kv1.5 excluding a contribution to the sustained IKur in atrial cells. To test for the contribution of Kv4-based Ito on sustained K(+) outward currents in human atrial myocytes, we used 4-AP to block IKur, in combination with Heteropoda toxin 2 to block Kv4 channels. We could clearly separate an Ito fraction of about 19% contributing to the late current in atrial myocytes. Thus, the interaction of DPP10a, expressed in human atrium, with Kv4.3 channels generates a sustained current component of Ito, which may affect late repolarization phase of atrial action potentials.


Subject(s)
Action Potentials/physiology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Myocytes, Cardiac/metabolism , Shal Potassium Channels/metabolism , Aged , Aged, 80 and over , Animals , CHO Cells , Cricetulus , Female , Fluorescent Antibody Technique , Heart Atria/metabolism , Humans , Male , Mice , Middle Aged , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Transfection
5.
J Mol Cell Cardiol ; 56: 8-18, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23291429

ABSTRACT

In human atrial myocytes the transient outward current I(to) develops a conspicuous faster inactivation with increasing temperatures. Since ß-subunits are known to modulate I(to) current kinetics, we hypothesized that the temperature sensitivity of I(to) is not only determined by the property of the ion-passing α-subunit Kv4.3 but also by its interaction with accessory ß-subunits. We therefore studied the influence of the transmembrane ß-subunits KCNE1, KCNE2 and DPP6 on Kv4.3/KChIP2 channels in CHO cells at room temperature and at physiological temperature. Exposure to 37°C caused a significant acceleration of the channel kinetics, whereas current densities and voltage dependences remained unaltered at 37°C compared to 23°C. However, Kv4.3/KChIP2 channels without transmembrane ß-subunits showed the strongest temperature sensitivity with considerably increased rates of activation and inactivation at 37°C. KCNE2 significantly slowed the current kinetics at 37°C compared to Kv4.3/KChIP2 channels, whereas KCNE1 did not influence the channel properties at both temperatures. Interestingly, the accelerating effects of DPP6 on current kinetics described at 23°C were diminished at physiological temperature, thus at 37°C current kinetics became remarkably similar for channel complexes Kv4.3/KChIP2 with and without DPP6 isoforms. A Markov state model was developed on the basis of experimental measurements to simulate the influence of ß-subunits on Kv4.3 channel complex at both temperatures. In conclusion, the remarkably fast kinetics of the native I(to) at 37°C could be reproduced by co-expressing Kv4.3, KChIP2, KCNE2 and DPP6 in CHO cells, whereas the high temperature sensitivity of human I(to) could be not mimicked.


Subject(s)
Protein Subunits/physiology , Shal Potassium Channels/metabolism , Action Potentials , Animals , CHO Cells , Cricetinae , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/physiology , Humans , Ion Channel Gating , Kinetics , Markov Chains , Models, Biological , Nerve Tissue Proteins/physiology , Patch-Clamp Techniques , Potassium Channels/physiology , Potassium Channels, Voltage-Gated/physiology , Protein Stability , Thermodynamics
6.
Prog Biophys Mol Biol ; 107(1): 4-10, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21745496

ABSTRACT

Cardiac experimental electrophysiology is in need of a well-defined Minimum Information Standard for recording, annotating, and reporting experimental data. As a step towards establishing this, we present a draft standard, called Minimum Information about a Cardiac Electrophysiology Experiment (MICEE). The ultimate goal is to develop a useful tool for cardiac electrophysiologists which facilitates and improves dissemination of the minimum information necessary for reproduction of cardiac electrophysiology research, allowing for easier comparison and utilisation of findings by others. It is hoped that this will enhance the integration of individual results into experimental, computational, and conceptual models. In its present form, this draft is intended for assessment and development by the research community. We invite the reader to join this effort, and, if deemed productive, implement the Minimum Information about a Cardiac Electrophysiology Experiment standard in their own work.


Subject(s)
Electrophysiological Phenomena , Heart/physiology , Information Dissemination/methods , Models, Biological , Research Design/standards , Animals , Humans , Reference Standards , Reproducibility of Results
7.
Thromb Haemost ; 105(6): 1010-23, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21544322

ABSTRACT

Atrial fibrillation (AF) patients may receive treatment from specialists or from general medicine physicians representing different levels of care within a structured health care system. This "choice" is influenced by patient flow within a health care system, patient preference, and individual access to health care resources. We analysed how the postgraduate training and work environment of treating physicians affects management decisions in AF patients. Patient characteristics and treatment decisions were analysed at the time of enrolment into the registry of the German Atrial Fibrillation NETwork (AFNET). A total of 9,577 patients were enrolled from 2004 to 2006 in 191 German centres that belonged to the following four levels of care: 13 tertiary care centres (TCC) enrolled 3,795 patients (39.6%), 58 district hospitals (DH) enrolled 2,339 patients (24.4%), 62 office-based cardiologists (OC) enrolled 2,640 patients (27.6%), and 58 general practitioners or internists (GP) enrolled 803 patients (8.4%). Patients with new-onset AF were often treated in DH. TCC treated younger patients who more often presented with paroxysmal AF. Older patients and patients in permanent AF more often received outpatient care. Consistent with recommendations, younger patients and patients with non-permanent AF received rhythm control therapy more often. In addition, the type of centre affected the decision for rhythm control. Stroke risk was similar between centre types (mean CHADS2 scores 1.6 -1.9). TCC (68.8%) and OC (73.6%) administered adequate antithrombotic therapy more often than DH (55.1%) or GP (52.0%, p<0.001 between groups). Upon multivariate analysis, enrolment by TCC or OC was associated with a 1.60 (1.20-2.12, p=0.001) fold chance for adequate antithrombotic treatment. This difference between centre types was consistent irrespective of the type of stroke risk estimation (ESC 2001 guidelines, CHADS2 score), and also consistent when the recently suggested CHA2DS2-VASc score was used to estimate stroke risk. In conclusion, management decisions in AF are influenced by the education and clinical background of treating physicians in Germany. Inpatients receive more rhythm control therapy. Adequate antithrombotic therapy is more often administered in specialist (cardiologist) centres.


Subject(s)
Atrial Fibrillation/epidemiology , Atrial Fibrillation/therapy , Cardiology , Fibrinolytic Agents/therapeutic use , Professional Practice/statistics & numerical data , Ambulatory Care/statistics & numerical data , Atrial Fibrillation/physiopathology , Disease Progression , Education, Medical, Graduate , General Practitioners , Germany , Health Services Accessibility/standards , Hospitals , Humans , Practice Patterns, Physicians' , Recurrence , Registries
9.
Herzschrittmacherther Elektrophysiol ; 21(4): 217-21, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21082185

ABSTRACT

Atrial fibrillation (AF) is accompanied by a high risk of thromboembolic complications necessitating anticoagulation therapy. Arrhythmias have a high tendency to become persistent. Catheter ablation techniques are highly effective in the treatment of AF; however, these procedures are far too costly and time-consuming for the routine treatment of large numbers of AF patients. Moreover, many patients prefer drug treatment although conventional antiarrhythmic drugs are moderately effective and are burdened with severe cardiac and noncardiac side effects. New antifibrillatory drugs developed for the treatment of AF include multichannel blockers with a high degree of atrial selectivity. The rationale of this approach is to induce antiarrhythmic actions only in the atria without conferring proarrhythmic effects in the ventricles.Atrial selective drug action is expected with ion channel blockers targeting ion channels that are expressed predominantly in the atria, i.e., Kv1.5 (I(Kur)), or Kir 3.1 and Kir 3.4 (I(K,ACh)). Na(+) channel blockers that dissociate rapidly may exert atrial selectivity because of subtle differences in atrial and ventricular action potentials. Finally, atrial-selective targets may evolve due to disease-specific processes (e.g., rate-dependent Na(+) channel blockers, selective drugs against constitutively active I(K,ACh) channels).


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Heart Atria/drug effects , Acetylcholine/physiology , Anti-Arrhythmia Agents/adverse effects , Atrial Fibrillation/complications , Catheter Ablation , Drug-Related Side Effects and Adverse Reactions , Electrocardiography/drug effects , Heart Ventricles/drug effects , Humans , Potassium Channels/drug effects , Sodium Channels/drug effects , Thromboembolism/prevention & control
10.
Herzschrittmacherther Elektrophysiol ; 21(3): 153-9, 2010 Sep.
Article in German | MEDLINE | ID: mdl-20676664

ABSTRACT

The German Competence Network on Atrial Fibrillation (AFNET) is a national interdisciplinary research network funded by the Federal Ministry of Education and Research (BMBF). AFNET was initiated in 2003 and aims at improving treatment of atrial fibrillation (AF), the most frequent sustained cardiac arrhythmia. AFNET has established a nationwide patient registry on diagnostics, therapy, course and complications of AF in Germany. The data analyzed to date demonstrate that patients with AF are likely to have multiple co-morbidities, such as hypertension, valvular heart disease, coronary artery disease, diabetes mellitus and advanced age. Oral anticoagulation is provided to the majority of patients in accordance with the recommendations given by guidelines. Further areas of research deal with the optimal duration of antiarrhythmic therapy following electrical cardioversion of atrial fibrillation and the value of strategies to prevent arrhythmogenic changes, such as fibrosis in the atria, for prevention of further episodes of atrial fibrillation. Additional registry projects were established for patients with catheter-based interventional therapy of atrial fibrillation and surgical ablation to define success, complications and long term results of these recently developed procedures more clearly. Data and insights gathered from these projects were used to further develop standards of care in two international conferences.


Subject(s)
Atrial Fibrillation/therapy , Quality Assurance, Health Care/organization & administration , Registries , Aged , Anti-Arrhythmia Agents/therapeutic use , Anticoagulants/therapeutic use , Atrial Fibrillation/complications , Atrial Fibrillation/diagnosis , Atrial Fibrillation/etiology , Biomedical Research , Cardiovascular Diseases/complications , Catheter Ablation , Combined Modality Therapy , Comorbidity , Cooperative Behavior , Electric Countershock , Evidence-Based Medicine , Female , Germany , Humans , Interdisciplinary Communication , Male , Outcome and Process Assessment, Health Care , Practice Guidelines as Topic , Randomized Controlled Trials as Topic
11.
Br J Pharmacol ; 154(4): 774-86, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18536731

ABSTRACT

BACKGROUND AND PURPOSE: The human cardiac transient outward potassium current (Ito) is believed to be composed of the pore-forming Kv4.3 alpha-subunit, coassembled with modulatory beta-subunits as KChIP2, MiRP1 and DPP6 proteins. beta-Subunits can alter the pharmacological response of Ito; therefore, we analysed the effects of flecainide on Kv4.3/KChIP2 channels coassembled with MiRP1 and/or DPP6 beta-subunits. EXPERIMENTAL APPROACH: Currents were recorded in Chinese hamster ovary cells stably expressing K(V)4.3/KChIP2 channels, and transiently transfected with either MiRP1, DPP6 or both, using the whole-cell patch-clamp technique. KEY RESULTS: In control conditions, Kv4.3/KChIP2/MiRP1 channels exhibited the slowest activation and inactivation kinetics and showed an 'overshoot' in the time course of recovery from inactivation. The midpoint values (Vh) of the activation and inactivation curves for Kv4.3/KChIP2/DPP6 and Kv4.3/KChIP2/MiRP1/DPP6 channels were approximately 10 mV more negative than Vh values for Kv4.3/KChIP2 and Kv4.3/KChIP2/MiRP1 channels. Flecainide (0.1-100 microM) produced a similar concentration-dependent blockade of total integrated current flow (IC50 approximately 10 microM) in all the channel complexes. However, the IC50 values for peak current amplitude and inactivated channel block were significantly different. Flecainide shifted the Vh values of both the activation and inactivation curves to more negative potentials and apparently accelerated inactivation kinetics in all channels. Moreover, flecainide slowed recovery from inactivation in all the channel complexes and suppressed the 'overshoot' in Kv4.3/KChIP2/MiRP1 channels. CONCLUSIONS AND IMPLICATIONS: Flecainide directly binds to the Kv4.3 alpha-subunit when the channels are in the open and inactivated state and the presence of the beta-subunits modulates the blockade by altering the gating function.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Flecainide/pharmacology , Protein Subunits , Shal Potassium Channels/drug effects , Animals , Anti-Arrhythmia Agents/administration & dosage , CHO Cells , Cricetinae , Cricetulus , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Dose-Response Relationship, Drug , Female , Flecainide/administration & dosage , Humans , Inhibitory Concentration 50 , Kv Channel-Interacting Proteins/metabolism , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Peptide Hydrolases/metabolism , Potassium Channels/metabolism , Potassium Channels, Voltage-Gated/metabolism , Shal Potassium Channels/metabolism , Transfection
12.
Br J Pharmacol ; 154(8): 1619-30, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18536759

ABSTRACT

BACKGROUND AND PURPOSE: This study was designed to establish the pathology-specific inhibitory effects of the IKur/Ito/IK,ACh blocker AVE0118 on atrium-selective channels and its corresponding effects on action potential shape and effective refractory period in patients with chronic AF (cAF). EXPERIMENTAL APPROACH: Outward K+-currents of right atrial myocytes and action potentials of atrial trabeculae were measured with whole-cell voltage clamp and microelectrode techniques, respectively. Outward currents were dissected by curve fitting. KEY RESULTS: Four components of outward K+-currents and AF-specific alterations in their properties were identified. Ito was smaller in cAF than in SR, and AVE0118 (10 microM) apparently accelerated its inactivation in both groups without reducing its amplitude. Amplitudes of rapidly and slowly inactivating components of IKur were lower in cAF than in SR. The former was abolished by AVE0118 in both groups, the latter was partially blocked in SR, but not in cAF, even though its inactivation was apparently accelerated in cAF. The large non-inactivating current component was similar in magnitude in both groups, but decreased by AVE0118 only in SR. AVE0118 strongly suppressed AF-related constitutively active IK,ACh and prolonged atrial action potential and effective refractory period exclusively in cAF. CONCLUSIONS AND IMPLICATIONS: In atrial myocytes of cAF patients, we detected reduced function of distinct IKur components that possessed decreased component-specific sensitivity to AVE0118 most likely as a consequence of AF-induced electrical remodelling. Inhibition of profibrillatory constitutively active IK,ACh may lead to pathology-specific efficacy of AVE0118 that is likely to contribute to its ability to convert AF into SR.


Subject(s)
Atrial Fibrillation/drug therapy , Biphenyl Compounds/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Action Potentials/drug effects , Aged , Atrial Fibrillation/physiopathology , Chronic Disease , Electrophysiology , Female , Heart Atria/cytology , Heart Atria/pathology , Humans , In Vitro Techniques , Male , Microelectrodes , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Patch-Clamp Techniques , Potassium Channels/metabolism
13.
Naunyn Schmiedebergs Arch Pharmacol ; 378(3): 253-60, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18542927

ABSTRACT

Chronic treatment with cyclosporine A (CyA) is often complicated by severe hypertension. If activation of the beta-adrenergic-receptor-linked adenylyl cyclase (AC) system contributes to hypertension is unresolved. Rats were treated with CyA (20 mg kg(-1) day(-1)) for 7 days. beta-adrenergic, muscarinic, and alpha-adrenergic receptors, G-proteins, and the activity of AC were determined in cardiac and pulmonary plasma membranes. The density of cardiac beta-adrenergic receptors, muscarinic receptors, alpha-adrenergic receptors, G(alphas) and, G(alphai) remained unchanged after treatment with CyA. However, CyA increased the responsiveness of AC to different stimulators. The responsiveness of AC was even more pronounced after solubilization and partial purification, suggesting a direct modulation of the enzyme. These data suggest that CyA modulates the activity of the sympathoadrenergic system by a direct, receptor-independent sensitization of AC, suggesting that this pathway contributes to hypertension in patients treated with CyA.


Subject(s)
Adenylyl Cyclases/metabolism , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Receptors, Adrenergic, beta/metabolism , Adenylyl Cyclases/biosynthesis , Adenylyl Cyclases/isolation & purification , Adrenergic beta-Agonists/pharmacology , Animals , Arrestin/biosynthesis , Cell Membrane/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Heart/drug effects , In Vitro Techniques , Isoproterenol/pharmacology , Lung/drug effects , Male , Myocardial Contraction/drug effects , Radioligand Assay , Rats , Rats, Inbred WKY , Receptors, G-Protein-Coupled/drug effects , Receptors, Muscarinic/drug effects , beta-Adrenergic Receptor Kinases/biosynthesis
15.
Br J Pharmacol ; 150(3): 261-70, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17179950

ABSTRACT

BACKGROUND AND PURPOSE: Adrenoceptors can associate with cardiac caveolae. To investigate the function of vascular caveolae, adrenoceptor-mediated effects were compared in the saphenous artery of caveolin-1 knockout (cav-1KO) and wild-type (WT) mice. EXPERIMENTAL APPROACH: Electronmicroscopy was used to detect caveolae. Real-Time quantitative PCR was used for adrenoceptor subtypes. Catecholamine-evoked contractions and relaxations were studied in arterial segments. KEY RESULTS: Caveolae were found in arterial smooth muscle from WT but not from cav-1KO mice. Arterial mRNA levels for the adrenoceptors alpha1A, alpha1B, alpha1D, beta1, beta2 and beta3 were similar in cav-1KO and WT. (-)-Noradrenaline contracted cav-1KO (-log EC50M=7.1) and WT (-log EC50M=7.3) arteries through prazosin-sensitive receptors. Maximum (-)-noradrenaline-evoked contractions were greater in cav-1KO than WT arteries. (-)-Isoprenaline relaxed WT arteries (-log EC50M=7.3) more potently than cav-1KO arteries (-log EC50M=6.8); the effects were antagonized partially and similarly by the beta2-selective antagonist ICI118551 (50 nM). The (-)-isoprenaline-evoked relaxation was partially antagonized by the beta1-adrenoceptor-selective antagonist CGP20712 (300 nM) in WT but not cav-1KO arteries. The beta3-adrenoceptor-selective antagonist L748337 (100 nM) partially antagonized the relaxant effects of (-)-isoprenaline in cav-1KO but not in WT arteries. BRL37344 partially relaxed arteries through beta3-adrenoceptors in cav-1KO but not WT. The relaxant effects of BRL37344 were decreased by the NO synthase inhibitor OmegaL-nitroarginine. CONCLUSIONS AND IMPLICATIONS: The function of arterial alpha1- and beta2-adrenoceptors is similar in cav-1KO and WT mice. beta1-adrenoceptor-mediated relaxation in WT is lost in cav-1KO and replaced by the appearance of beta3-adrenoceptors.


Subject(s)
Caveolin 1/physiology , Mice, Knockout/physiology , Receptors, Adrenergic, alpha/physiology , Receptors, Adrenergic, beta/physiology , Animals , Arteries , Cell Membrane/physiology , Cell Membrane/ultrastructure , Hindlimb/blood supply , Mice , Microscopy, Electron , Polymerase Chain Reaction , Receptors, Adrenergic, alpha/genetics , Receptors, Adrenergic, beta/genetics
16.
Naunyn Schmiedebergs Arch Pharmacol ; 374(3): 249-53, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17106669

ABSTRACT

Activation of both beta(1)- and beta(2)-adrenoceptors increases the contractility of human atrial myocardium through cyclic AMP-dependent pathways. Cyclic AMP is hydrolised by phosphodiesterases, but little is known about which isoenzymes catalyse inotropically relevant cyclic AMP accumulated upon stimulation of beta-adrenoceptor subtypes. We have compared the positive inotropic effects of (-)-noradrenaline and (-)-adrenaline, mediated through beta(1)- and beta(2)-adrenoceptors, respectively, in the absence and presence of the PDE3 inhibitor cilostamide (300 nM) or PDE4 inhibitor rolipram (1 muM) on human atrial trabeculae from non-failing hearts. Cilostamide, but not rolipram, potentiated the effects of both (-)-noradrenaline and (-)-adrenaline. Cilostamide increased the -logEC(50)M of (-)-adrenaline more than of (-)-noradrenaline (P < 0.05), regardless of whether or not the patients had been chronically treated with beta-blockers. The results are consistent with a greater PDE3-catalysed hydrolysis of inotropically relevant cyclic AMP produced through beta(2)-adrenoceptors than beta(1)-adrenoceptors in human atrium.


Subject(s)
Cyclic AMP/metabolism , Phosphodiesterase Inhibitors/pharmacology , Quinolones/pharmacology , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-2/drug effects , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adrenergic beta-Antagonists , Aged , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Dose-Response Relationship, Drug , Epinephrine/pharmacology , Female , Heart Atria/drug effects , Humans , Hydrolysis , In Vitro Techniques , Male , Middle Aged , Myocardial Contraction/drug effects , Myocardium , Norepinephrine/pharmacology , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Rolipram/pharmacology
18.
Herzschrittmacherther Elektrophysiol ; 17(2): 64-72, 2006 Jun.
Article in German | MEDLINE | ID: mdl-16786464

ABSTRACT

During the last ten years we have made substantial progress in our understanding of the underlying mechanisms of atrial fibrillation. The high rate associated alterations in electrical and structural properties of the atria, referred to as atrial remodeling, promote the progression of atrial fibrillation. The development of new therapeutic approaches addresses three different directions: (i) prevention of atrial remodeling, especially of structural remodeling; (ii) increase of long-term efficacy of currently used drugs and improvement of their side-effect profile; and (iii) design of atria- and pathology-specific antiarrhythmic drugs without concomitant proarrhythmic effects in the ventricles. The current review outlines the pathophysiology of atrial fibrillation and focuses on electrical remodeling. The properties of new antiarrhythmic drugs for atrial fibrillation are discussed in detail.


Subject(s)
Anti-Arrhythmia Agents/administration & dosage , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Calcium Channel Blockers/administration & dosage , Calcium Channels/drug effects , Heart Conduction System/physiopathology , Animals , Heart Conduction System/drug effects , Humans , Ion Channel Gating/drug effects , Models, Cardiovascular , Practice Guidelines as Topic , Practice Patterns, Physicians' , Treatment Outcome
20.
Circulation ; 112(24): 3697-706, 2005 Dec 13.
Article in English | MEDLINE | ID: mdl-16330682

ABSTRACT

BACKGROUND: The molecular mechanism of increased background inward rectifier current (IK1) in atrial fibrillation (AF) is not fully understood. We tested whether constitutively active acetylcholine (ACh)-activated I(K,ACh) contributes to enhanced basal conductance in chronic AF (cAF). METHODS AND RESULTS: Whole-cell and single-channel currents were measured with standard voltage-clamp techniques in atrial myocytes from patients with sinus rhythm (SR) and cAF. The selective I(K,ACh) blocker tertiapin was used for inhibition of I(K,ACh). Whole-cell basal current was larger in cAF than in SR, whereas carbachol (CCh)-activated I(K,ACh) was lower in cAF than in SR. Tertiapin (0.1 to 100 nmol/L) reduced I(K,ACh) in a concentration-dependent manner with greater potency in cAF than in SR (-logIC50: 9.1 versus 8.2; P<0.05). Basal current contained a tertiapin-sensitive component that was larger in cAF than in SR (tertiapin [10 nmol/L]-sensitive current at -100 mV: cAF, -6.7+/-1.2 pA/pF, n=16/5 [myocytes/patients] versus SR, -1.7+/-0.5 pA/pF, n=24/8), suggesting contribution of constitutively active I(K,ACh) to basal current. In single-channel recordings, constitutively active I(K,ACh) was prominent in cAF but not in SR (channel open probability: cAF, 5.4+/-0.7%, n=19/9 versus SR, 0.1+/-0.05%, n=16/9; P<0.05). Moreover, IK1 channel open probability was higher in cAF than in SR (13.4+/-0.4%, n=19/9 versus 11.4+/-0.7%, n=16/9; P<0.05) without changes in other channel characteristics. CONCLUSIONS: Our results demonstrate that larger basal inward rectifier K+ current in cAF consists of increased IK1 activity and constitutively active I(K,ACh). Blockade of I(K,ACh) may represent a new therapeutic target in AF.


Subject(s)
Atrial Fibrillation/etiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , GTP-Binding Protein beta Subunits/genetics , Acetylcholine/pharmacology , Aged , Atrial Appendage/cytology , Bee Venoms/pharmacology , Carbachol/pharmacology , Chronic Disease , Electrophysiology , Female , GTP-Binding Protein beta Subunits/physiology , Genotype , Humans , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Polymorphism, Single Nucleotide , Potassium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL