Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Acta Physiol (Oxf) ; 216(2): 186-202, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26284956

ABSTRACT

Normal cardiac excitation involves orderly conduction of electrical activation and recovery dependent upon surface membrane, voltage-gated, sodium (Na(+) ) channel α-subunits (Nav 1.5). We summarize experimental studies of physiological and clinical consequences of loss-of-function Na(+) channel mutations. Of these conditions, Brugada syndrome (BrS) and progressive cardiac conduction defect (PCCD) are associated with sudden, often fatal, ventricular tachycardia (VT) or fibrillation. Mouse Scn5a(+/-) hearts replicate important clinical phenotypes modelling these human conditions. The arrhythmic phenotype is associated not only with the primary biophysical change but also with additional, anatomical abnormalities, in turn dependent upon age and sex, each themselves exerting arrhythmic effects. Available evidence suggests a unified binary scheme for the development of arrhythmia in both BrS and PCCD. Previous biophysical studies suggested that Nav 1.5 deficiency produces a background electrophysiological defect compromising conduction, thereby producing an arrhythmic substrate unmasked by flecainide or ajmaline challenge. More recent reports further suggest a progressive decline in conduction velocity and increase in its dispersion particularly in ageing male Nav 1.5 haploinsufficient compared to WT hearts. This appears to involve a selective appearance of slow conduction at the expense of rapidly conducting pathways with changes in their frequency distributions. These changes were related to increased cardiac fibrosis. It is thus the combination of the structural and biophysical changes both accentuating arrhythmic substrate that may produce arrhythmic tendency. This binary scheme explains the combined requirement for separate, biophysical and structural changes, particularly occurring in ageing Nav 1.5 haploinsufficient males in producing clinical arrhythmia.


Subject(s)
Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Heart/physiopathology , NAV1.5 Voltage-Gated Sodium Channel/genetics , Animals , Haploinsufficiency , Humans
3.
Acta Physiol (Oxf) ; 214(3): 361-75, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25850710

ABSTRACT

AIMS: Cardiac ryanodine receptor mutations are associated with catecholaminergic polymorphic ventricular tachycardia (CPVT), and some, including RyR2-P2328S, also predispose to atrial fibrillation. Recent work associates reduced atrial Nav 1.5 currents in homozygous RyR2-P2328S (RyR2(S/S) ) mice with slowed conduction and increased arrhythmogenicity. Yet clinically, and in murine models, the Nav 1.5 blocker flecainide reduces ventricular arrhythmogenicity in CPVT. We aimed to determine whether, and how, flecainide influences atrial arrhythmogenicity in RyR2(S/S) mice and their wild-type (WT) littermates. METHODS: We explored effects of 1 µm flecainide on WT and RyR2(S/S) atria. Arrhythmic incidence, action potential (AP) conduction velocity (CV), atrial effective refractory period (AERP) and AP wavelength (λ = CV × AERP) were measured using multi-electrode array recordings in Langendorff-perfused hearts; Na(+) currents (INa ) were recorded using loose patch clamping of superfused atria. RESULTS: RyR2(S/S) showed more frequent atrial arrhythmias, slower CV, reduced INa and unchanged AERP compared to WT. Flecainide was anti-arrhythmic in RyR2(S/S) but pro-arrhythmic in WT. It increased INa in RyR2(S/S) atria, whereas it reduced INa as expected in WT. It increased AERP while sparing CV in RyR2(S/S) , but reduced CV while sparing AERP in WT. Thus, RyR2(S/S) hearts have low λ relative to WT; flecainide then increases λ in RyR2(S/S) but decreases λ in WT. CONCLUSIONS: Flecainide (1 µm) rescues the RyR2-P2328S atrial arrhythmogenic phenotype by restoring compromised INa and λ, changes recently attributed to increased sarcoplasmic reticular Ca(2+) release. This contrasts with the increased arrhythmic incidence and reduced INa and λ with flecainide in WT.


Subject(s)
Atrial Fibrillation/metabolism , Flecainide/administration & dosage , Membrane Potentials/drug effects , NAV1.5 Voltage-Gated Sodium Channel/drug effects , Ryanodine Receptor Calcium Release Channel/genetics , Sodium/metabolism , Animals , Anti-Arrhythmia Agents/administration & dosage , Atrial Fibrillation/diagnosis , Ion Channel Gating/drug effects , Mice , Mutation , Treatment Outcome , Voltage-Gated Sodium Channel Blockers/administration & dosage
4.
Acta Physiol (Oxf) ; 211(4): 559-73, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24913289

ABSTRACT

AIM: To test a hypothesis attributing arrhythmia in Brugada Syndrome to right ventricular (RV) outflow tract (RVOT) conduction abnormalities arising from Nav 1.5 insufficiency and fibrotic change. METHODS: Arrhythmic properties of Langendorff-perfused Scn5a+/- and wild-type mouse hearts were correlated with ventricular effective refractory periods (VERPs), multi-electrode array (MEA) measurements of action potential (AP) conduction velocities and dispersions in conduction direction (CD), Nav 1.5 expression levels, and fibrotic change, as measured at the RVOT and RV. Two-way anova was used to test for both independent and interacting effects of anatomical region and genotype on these parameters. RESULTS: Scn5a+/- hearts showed greater arrhythmic frequencies during programmed electrical stimulation at the RVOT but not the RV. The Scn5a+/- genotype caused an independent increase of VERP regardless of whether the recording site was the RVOT or RV. Effective AP conduction velocities (CV†s), derived from fitting regression planes to arrays of observed local activation times were reduced in Scn5a+/- hearts and at the RVOT independently. AP conduction velocity magnitudes derived by averaging MEA results from local vector analyses, CV*, were reduced by the Scn5a+/- genotype alone. In contrast, dispersions in conduction direction, were greater in the RVOT than the RV, when the atrioventricular node was used as the pacing site. The observed reductions in Nav 1.5 expression were attributable to Scn5a+/-, whereas increased levels of fibrosis were associated with the RVOT. CONCLUSIONS: The Scn5a+/- RVOT recapitulates clinical findings of increased arrhythmogenicity through reduced CV† reflecting reduced CV* attributable to reduced Nav 1.5 expression and increased CD attributable to fibrosis.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Brugada Syndrome/physiopathology , Heart Conduction System/physiopathology , Heart/physiopathology , Action Potentials/physiology , Animals , Blotting, Western , Disease Models, Animal , Electrophysiology , Female , Male , Mice , Mice, Mutant Strains , NAV1.5 Voltage-Gated Sodium Channel/genetics , Organ Culture Techniques
5.
Acta Physiol (Oxf) ; 207(2): 308-23, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22958452

ABSTRACT

AIM: RyR2 mutations are associated with catecholaminergic polymorphic tachycardia, a condition characterized by ventricular and atrial arrhythmias. The present experiments investigate the atrial electrophysiology of homozygotic murine RyR2-P2328S (RyR2(S/S)) hearts for ectopic triggering events and for conduction abnormalities that might provide a re-entrant substrate. METHODS: Electrocardiograph recordings were made from regularly stimulated RyR2(S/S) and wild type (WT) hearts, perfused using a novel modified Langendorff preparation. This permitted the simultaneous use of either floating intracellular microelectrodes to measure action potential (AP) parameters, or a multielectrode array to measure epicardial conduction velocity (CV). RESULTS: RyR2(S/S) showed frequent sustained tachyarrhythmias, delayed afterdepolarizations and ectopic APs, increased interatrial conduction delays, reduced epicardial CVs and reduced maximum rates of AP depolarization ((dV/dt)(max)), despite similar effective refractory periods, AP durations and AP amplitudes. Effective interatrial CVs and (dV/dt)(max) values of APs following ectopic (S2) stimulation were lower than those of APs following regular stimulation and decreased with shortening S1S2 intervals. However, although RyR2(S/S) atria showed arrhythmias over a wider range of S1S2 intervals, the interatrial CV and (dV/dt)(max) of S2 APs provoking such arrhythmias were similar in RyR2(S/S) and WT. CONCLUSIONS: These results suggest that abnormal intracellular Ca(2+) homoeostasis produces both arrhythmic triggers and a slow-conducting arrhythmic substrate in RyR2(S/S) atria. A similar mechanism might also contribute to arrhythmogenesis in other conditions, associated with diastolic Ca(2+) release, such as atrial fibrillation.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Calcium/metabolism , Heart Atria/physiopathology , Ryanodine Receptor Calcium Release Channel/metabolism , Action Potentials/physiology , Animals , Arrhythmias, Cardiac/metabolism , Electrocardiography , Heart Atria/metabolism , Mice , Mice, Mutant Strains , Organ Culture Techniques , Ryanodine Receptor Calcium Release Channel/genetics
6.
Acta Physiol (Oxf) ; 206(3): 164-77, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22510251

ABSTRACT

AIM: QT interval prolongation reflecting delayed action potential (AP) repolarization is associated with polymorphic ventricular tachycardia and early after depolarizations potentially initiating extrasystolic APs if of sufficient amplitude. The current experiments explored contributions of altered re-excitation thresholds for, and conduction of, such extrasystolic APs to arrhythmogenesis in Langendorff-perfused, normokalaemic, control wild-type hearts and two experimental groups modelling long QT (LQT). The two LQT groups consisted of genetically modified, Scn5a(+/ΔKPQ) and hypokalaemic wild-type murine hearts. METHODS: Hearts were paced from their right ventricles and monophasic AP electrode recordings obtained from their left ventricular epicardia, with recording and pacing electrodes separated by 1 cm. An adaptive programmed electrical stimulation protocol applied pacing (S1) stimulus trains followed by premature (S2) extrastimuli whose amplitudes were progressively increased with progressive decrements in S1S2 interval to maintain stimulus capture. Such protocols culminated in either arrhythmic or refractory endpoints. RESULTS: Arrhythmic outcomes were associated with (1) lower conduction velocities in their initiating extrasystolic APs than refractory outcomes and (2) higher conduction velocities in the LQT groups than in controls. Furthermore, (3) the endpoints were reached at longer S1S2 coupling intervals and with smaller stimulus amplitudes in the LQT groups compared with controls. This was despite (4) similar relationships between conduction velocity and S1S2 coupling interval and between re-excitation thresholds and S1S2 coupling interval in all three experimental groups. CONCLUSIONS: Arrhythmias induced by extrasystolic APs in the LQT groups thus occur under conditions of higher conduction velocity and greater sensitivity to extrastimuli than in controls.


Subject(s)
Action Potentials/physiology , Arrhythmias, Cardiac/physiopathology , Long QT Syndrome/physiopathology , Models, Animal , Neural Conduction/physiology , Ventricular Premature Complexes/physiopathology , Animals , Arrhythmias, Cardiac/genetics , Electric Stimulation , Endpoint Determination , Female , Heart Conduction System/physiopathology , Hypokalemia/genetics , Hypokalemia/physiopathology , Long QT Syndrome/genetics , Male , Mice , Mice, Inbred Strains , Mice, Mutant Strains , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/physiology , Neural Conduction/genetics , Time Factors , Ventricular Premature Complexes/genetics
7.
Acta Physiol (Oxf) ; 200(2): 129-46, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20384594

ABSTRACT

AIM: clinical observations suggest that alternans in action potential (AP) characteristics presages breakdown of normal ordered cardiac electrical activity culminating in ventricular arrhythmogenesis. We compared such temporal nonuniformities in monophasic action potential (MAP) waveforms in left (LV) and right ventricular (RV) epicardia and endocardia of Langendorff-perfused murine wild-type (WT), and Scn5a(+/-) hearts modelling Brugada syndrome (BrS) for the first time. METHODS: a dynamic pacing protocol imposed successively incremented steady pacing rates between 5.5 and 33 Hz. A signal analysis algorithm detected sequences of >10 beats showing alternans. Results were compared before and following the introduction of flecainide (10 microm) and quinidine (5 microm) known to exert pro- and anti-arrhythmic effects in BrS. RESULTS: sustained and transient amplitude and duration alternans were both frequently followed by ventricular ectopic beats and ventricular tachycardia or fibrillation. Diastolic intervals (DIs) that coincided with onsets of transient (tr) or sustained (ss) alternans in MAP duration (DI*) and amplitude (DI') were determined. Kruskal-Wallis tests followed by Bonferroni-corrected Mann-Whitney U-tests were applied to these DI results sorted by recording site, pharmacological conditions or experimental populations. WT hearts showed no significant heterogeneities in any DI. Untreated Scn5a (+/-) hearts showed earlier onsets of transient but not sustained duration alternans in LV endocardium compared with RV endocardium or LV epicardium. Flecainide administration caused earlier onsets of both transient and sustained duration alternans selectively in the RV epicardium in the Scn5a (+/-) hearts. CONCLUSION: these findings in a genetic model thus implicate RV epicardial changes in the arrhythmogenicity produced by flecainide challenge in previously asymptomatic clinical BrS.


Subject(s)
Action Potentials/physiology , Heart/physiopathology , Sodium Channels/genetics , Animals , Brugada Syndrome/genetics , Brugada Syndrome/physiopathology , Cardiac Pacing, Artificial , Electrophysiology , Mice , Mice, Mutant Strains , NAV1.5 Voltage-Gated Sodium Channel , Organ Culture Techniques
8.
Acta Physiol (Oxf) ; 200(1): 23-33, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20331542

ABSTRACT

AIM: To investigate the interacting effects of age and sex on electrocardiographic (ECG) features of Scn5a(+/-) mice modelling Brugada syndrome. METHODS: Recordings were performed on anaesthetized wild-type (WT) and Scn5a(+/-) mice and differences attributable to these risk factors statistically stratified. RESULTS: Scn5a(+/-) exerted sex-dependent effects upon sino-atrial function that only became apparent with age. RR intervals were greater in old male than in old female Scn5a(+/-). Atrio-ventricular (AV) conduction was slower in young female mice, whether WT and Scn5a(+/-), than the corresponding young male WT and Scn5a(+/-). However, PR intervals lengthened with age in male but not in female Scn5a(+/-) giving the greatest PR intervals in old male Scn5a(+/-) compared with either old male WT or young male Scn5a(+/-) mice. In contrast, PR intervals were similar in old female Scn5a(+/-) and in old female WT. QTc was prolonged in Scn5a(+/-) compared with WT, and female Scn5a(+/-) compared with female WT. Age-dependent alterations in durations of ventricular repolarization relative to WT affected male but not female Scn5a(+/-). Thus, T-wave durations were greater in old male Scn5a(+/-) compared with old male WT, but indistinguishable between old female Scn5a(+/-) and old female WT. Finally, analysis for combined interactions of genotype, age and sex demonstrated no effects on P wave and QRS durations and QTc intervals. CONCLUSION: We demonstrate for the first time that age, sex and genotype exert both independent and interacting ECG effects. The latter suggest alterations in cardiac pacemaker function, atrio-ventricular conduction and ventricular repolarization greatest in ageing male Scn5a(+/-).


Subject(s)
Atrioventricular Node/physiopathology , Brugada Syndrome/physiopathology , Mutation , Sinoatrial Node/physiopathology , Sodium Channels/metabolism , Sodium/metabolism , Age Factors , Animals , Atrioventricular Node/metabolism , Brugada Syndrome/genetics , Brugada Syndrome/metabolism , Disease Models, Animal , Electrocardiography , Female , Genetic Predisposition to Disease , Male , Membrane Potentials , Mice , Mice, Knockout , NAV1.5 Voltage-Gated Sodium Channel , Phenotype , Sex Factors , Sinoatrial Node/metabolism , Sodium Channels/deficiency , Sodium Channels/genetics
9.
Acta Physiol (Oxf) ; 198(1): 47-59, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19796257

ABSTRACT

AIM: In contrast to extensive reports on the roles of Na(v)1.5 alpha-subunits, there have been few studies associating the beta-subunits with cardiac arrhythmogenesis. We investigated the sino-atrial and conduction properties in the hearts of Scn3b(-/-) mice. METHODS: The following properties were compared in the hearts of wild-type (WT) and Scn3b(-/-) mice: (1) mRNA expression levels of Scn3b, Scn1b and Scn5a in atrial tissue. (2) Expression of the beta(3) protein in isolated cardiac myocytes. (3) Electrocardiographic recordings in intact anaesthetized preparations. (4) Bipolar electrogram recordings from the atria of spontaneously beating and electrically stimulated Langendorff-perfused hearts. RESULTS: Scn3b mRNA was expressed in the atria of WT but not Scn3b(-/-) hearts. This was in contrast to similar expression levels of Scn1b and Scn5a mRNA. Immunofluorescence experiments confirmed that the beta(3) protein was expressed in WT and absent in Scn3b(-/-) cardiac myocytes. Lead I electrocardiograms from Scn3b(-/-) mice showed slower heart rates, longer P wave durations and prolonged PR intervals than WT hearts. Spontaneously beating Langendorff-perfused Scn3b(-/-) hearts demonstrated both abnormal atrial electrophysiological properties and evidence of partial or complete dissociation of atrial and ventricular activity. Atrial burst pacing protocols induced atrial tachycardia and fibrillation in all Scn3b(-/-) but hardly any WT hearts. Scn3b(-/-) hearts also demonstrated significantly longer sinus node recovery times than WT hearts. CONCLUSION: These findings demonstrate, for the first time, that a deficiency in Scn3b results in significant atrial electrophysiological and intracardiac conduction abnormalities, complementing the changes in ventricular electrophysiology reported on an earlier occasion.


Subject(s)
Arrhythmia, Sinus/metabolism , Heart Atria/metabolism , Myocytes, Cardiac/metabolism , Sodium Channels/metabolism , Animals , Electrocardiography , Female , Fluorescent Antibody Technique , Male , Mice , Mice, Knockout , NAV1.5 Voltage-Gated Sodium Channel , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/genetics
10.
Acta Physiol (Oxf) ; 198(2): 143-58, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19886909

ABSTRACT

AIM: To investigate the effect of increases in extracellular Ca(2+) entry produced by the L-type Ca(2+) channel agonist FPL-64176 (FPL) upon acute atrial arrhythmogenesis in intact Langendorff-perfused mouse hearts and its dependence upon diastolic Ca(2+) release from sarcoplasmic reticular Ca(2+) stores. METHODS: Confocal microscope studies of Fluo-3 fluorescence in isolated atrial myocytes were performed in parallel with electrophysiological examination of Langendorff-perfused mouse hearts. RESULTS: Atrial myocytes stimulated at 1 Hz and exposed to FPL (0.1 microm) initially showed (<10 min) frequent, often multiple, diastolic peaks following the evoked Ca(2+) transients whose amplitudes remained close to control values. With continued pacing (>10 min) this reverted to a regular pattern of evoked transients with increased amplitudes but in which diastolic peaks were absent. Higher FPL concentrations (1.0 microm) produced sustained and irregular patterns of cytosolic Ca(2+) activity, independent of pacing. Nifedipine (0.5 microm), and caffeine (1.0 mm) and cyclopiazonic acid (CPA) (0.15 microm) pre-treatments respectively produced immediate and gradual reductions in the F/F(0) peaks. Such nifedipine and caffeine, or CPA pre-treatments, abolished, or reduced, the effects of 0.1 and 1.0 microm FPL on cytosolic Ca(2+) signals. FPL (1.0 microm) increased the incidence of atrial tachycardia and fibrillation in intact Langendorff-perfused hearts without altering atrial effective refractory periods. These effects were inhibited by nifedipine and caffeine, and reduced by CPA. CONCLUSION: Enhanced extracellular Ca(2+) entry exerts acute atrial arrhythmogenic effects that is nevertheless dependent upon diastolic Ca(2+) release. These findings complement reports that associate established, chronic, atrial arrhythmogenesis with decreased overall inward Ca(2+) current.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Calcium/metabolism , Heart/drug effects , Aniline Compounds/pharmacology , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Caffeine/pharmacology , Calcium Channels, L-Type/metabolism , Calcium Signaling , Heart/physiopathology , Heart Atria/drug effects , Heart Atria/physiopathology , Mice , Myocytes, Cardiac/drug effects , Nifedipine/pharmacology , Pyrroles/pharmacology , Xanthenes/pharmacology
11.
Acta Physiol (Oxf) ; 197(1): 13-25, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19416122

ABSTRACT

AIM: The multifunctional signal molecule calmodulin kinase II (CaMKII) has been associated with cardiac arrhythmogenesis under conditions where its activity is chronically elevated. Recent studies report that its activity is also acutely elevated during acidosis. We test a hypothesis implicating CaMKII in the arrhythmogenesis accompanying metabolic acidification. METHODS: We obtained monophasic action potential recordings from Langendorff-perfused whole heart preparations and single cell action potentials (AP) using whole-cell patch-clamped ventricular myocytes. Spontaneous sarcoplasmic reticular (SR) Ca(2+)release events during metabolic acidification were investigated using confocal microscope imaging of Fluo-4-loaded ventricular myocytes. RESULTS: In Langendorff-perfused murine hearts, introduction of lactic acid into the Krebs-Henseleit perfusate resulted in abnormal electrical activity and ventricular tachycardia. The CaMKII inhibitor, KN-93 (2 microm), reversibly suppressed this spontaneous arrhythmogenesis during intrinsic rhythm and regular 8 Hz pacing. However, it failed to suppress arrhythmia evoked by programmed electrical stimulation. These findings paralleled a CaMKII-independent reduction in the transmural repolarization gradients during acidosis, which previously has been associated with the re-entrant substrate under other conditions. Similar acidification produced spontaneous AP firing and membrane potential oscillations in patch-clamped isolated ventricular myocytes when pipette solutions permitted cytosolic Ca(2+) to increase following acidification. However, these were abolished by both KN-93 and use of pipette solutions that held cytosolic Ca(2+) constant during acidosis. Acidosis also induced spontaneous Ca(2+) waves in isolated intact Fluo-4-loaded myocytes studied using confocal microscopy that were abolished by KN-93. CONCLUSION: These findings together implicate CaMKII-dependent SR Ca(2+) waves in spontaneous arrhythmic events during metabolic acidification.


Subject(s)
Acidosis/enzymology , Arrhythmias, Cardiac/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Myocytes, Cardiac/enzymology , Acidosis/complications , Action Potentials/drug effects , Action Potentials/physiology , Animals , Arrhythmias, Cardiac/etiology , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Female , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/enzymology , In Vitro Techniques , Male , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Protein Kinase Inhibitors/pharmacology , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Sulfonamides/pharmacology
12.
Acta Physiol (Oxf) ; 194(4): 311-23, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18616619

ABSTRACT

AIM: We compared the clinical and physiological consequences of the novel mutation R878C in a highly conserved pore residue in domain II (S5-S6) of human, hNa(v)1.5, cardiac Na(+) channels. METHODS: Full clinical evaluation of pedigree members through three generations of a Chinese family combined with SCN5A sequencing from genomic DNA was compared with patch and voltage-clamp results from two independent expression systems. RESULTS: The four mutation carriers showed bradycardia, and slowed sino-atrial, atrioventricular and intraventricular conduction. Two also showed sick sinus syndrome; two had ST elevation in leads V1 and V2. Unlike WT-hNa(v)1.5, whole-cell patch-clamped HEK293 cells expressing R878C-hNa(v)1.5 showed no detectable Na(+) currents (i(Na)), even with substitution of a similarly charged lysine residue. Voltage-clamped Xenopus oocytes injected with either 0.04 or 1.5 microg microL(-1) R878C-hNa(v)1.5 cRNA similarly showed no i(Na), yet WT-hNa(v)1.5 cRNA diluted to 0.0004-0.0008 ng microL(-1)resulted in expression of detectable i(Na). i(Na) was simply determined by the amount of injected WT-hNa(v)1.5: doubling the dose of WT-hNa(v)1.5 cRNA doubled i(Na). i(Na) amplitudes and activation and inactivation characteristics were similar irrespective of whether WT-hNa(v)1.5 cRNA was given alone or combined with equal doses of R878C-hNa(v)1.5 cRNA therefore excluding dominant negative phenotypic effects. Na(+) channel function in HEK293 cells transfected with R878C-hNa(v)1.5 was not restored by exposure to mexiletine (200 microM) and lidocaine (100 microM). Fluorescence confocal microscopy using E3-Nav1.5 antibody demonstrated persistent membrane expression of both WT and R878C-hNa(v)1.5. Modelling studies confirmed that such i(Na) reductions reproduced the SSS phenotype. CONCLUSION: Clinical consequences of the novel R878C mutation correlate with results of physiological studies.


Subject(s)
Arrhythmias, Cardiac/genetics , Mutation/genetics , Myocardium/metabolism , Sodium Channels/genetics , Adult , Animals , Arrhythmias, Cardiac/metabolism , Electrocardiography , Female , Heterozygote , Humans , Immunohistochemistry , Male , Microscopy, Confocal , Pedigree , Sick Sinus Syndrome/genetics , Sick Sinus Syndrome/metabolism , Xenopus
13.
Acta Physiol (Oxf) ; 194(2): 123-40, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18419777

ABSTRACT

AIM: To explore the physiological consequences of the ryanodine receptor (RyR2)-P2328S mutation associated with catecholaminergic polymorphic ventricular tachycardia (CPVT). METHODS: We generated heterozygotic (RyR2 p/s) and homozygotic (RyR2 s/s) transgenic mice and studied Ca2+ signals from regularly stimulated, Fluo-3-loaded, cardiac myocytes. Results were compared with monophasic action potentials (MAPs) in Langendorff-perfused hearts under both regular and programmed electrical stimulation (PES). RESULTS: Evoked Ca2+ transients from wild-type (WT), heterozygote (RyR2 p/s) and homozygote (RyR2 s/s) myocytes had indistinguishable peak amplitudes with RyR2 s/s showing subsidiary events. Adding 100 nm isoproterenol produced both ectopic peaks and subsidiary events in WT but not RyR2 p/s and ectopic peaks and reduced amplitudes of evoked peaks in RyR2 s/s. Regularly stimulated WT, RyR2 p/s and RyR2 s/s hearts showed indistinguishable MAP durations and refractory periods. RyR2 p/s hearts showed non-sustained ventricular tachycardias (nsVTs) only with PES. Both nsVTs and sustained VTs (sVTs) occurred with regular stimuli and PES with isoproterenol treatment. RyR2 s/s hearts showed higher incidences of nsVTs before but mainly sVTs after introduction of isoproterenol with both regular stimuli and PES, particularly at higher pacing frequencies. Additionally, intrinsically beating RyR2 s/s showed extrasystolic events often followed by spontaneous sVT. CONCLUSION: The RyR2-P2328S mutation results in marked alterations in cellular Ca2+ homeostasis and arrhythmogenic properties resembling CPVT with greater effects in the homozygote than the heterozygote demonstrating an important gene dosage effect.


Subject(s)
Mutation , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Tachycardia, Ventricular/genetics , Action Potentials , Animals , Calcium/metabolism , Cardiac Pacing, Artificial/methods , Cells, Cultured , Heterozygote , Homeostasis , Homozygote , Mice , Mice, Transgenic , Organ Culture Techniques , Reverse Transcriptase Polymerase Chain Reaction/methods , Ryanodine Receptor Calcium Release Channel/physiology , Tachycardia, Ventricular/metabolism
14.
Acta Physiol (Oxf) ; 192(4): 455-69, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18045245

ABSTRACT

Sudden cardiac death stemming from ventricular arrhythmogenesis is one of the major causes of mortality in the developed world. Congenital and acquired forms of long QT syndrome (LQTS) are in turn associated with life threatening arrhythmias. Over the past decade our understanding of arrhythmogenic mechanisms in the setting of these diseases has increased greatly due to the creation of a number of animal models. Of these, the genetically amenable mouse has proved to be a particularly powerful tool. This review summarizes the congenital and acquired LQTS and describes the various mouse models that have been created to further probe arrhythmogenic mechanisms.


Subject(s)
Arrhythmias, Cardiac/metabolism , Disease Models, Animal , Animals , Humans , Mice
15.
Acta Physiol (Oxf) ; 192(4): 505-17, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17973950

ABSTRACT

AIM: To perform an empirical, pharmacological, separation of early afterdepolarizations (EADs) and transmural gradients of repolarization in arrhythmogenesis in a genetically modified mouse heart modelling human long QT syndrome (LQT) 3. METHODS: Left ventricular endocardial and epicardial monophasic action potentials and arrhythmogenic tendency were compared in isolated wild type (WT) and Scn5a+/Delta hearts perfused with 0.1 and 1 microm propranolol and paced from the right ventricular epicardium. RESULTS: All spontaneously beating bradycardic Scn5a+/Delta hearts displayed EADs, triggered beats and ventricular tachycardia (VT; n = 7), events never seen in WT hearts (n = 5). Perfusion with 0.1 and 1 microm propranolol suppressed all EADs, triggered beats and episodes of VT. In contrast, triggering of VT persisted following programmed electrical stimulation in 6 of 12 (50%), one of eight (12.5%), but six of eight (75%) Scn5a+/Delta hearts perfused with 0, 0.1 and 1 microm propranolol respectively in parallel with corresponding alterations in repolarization gradients, reflected in action potential duration (DeltaAPD(90)) values. Thus 0.1 microm propranolol reduced epicardial but not endocardial APD(90) from 54.7 +/- 1.6 to 44.0 +/- 2.0 ms, restoring DeltaAPD(90) from -3.8 +/- 1.6 to 3.5 +/- 2.5 ms (all n = 5), close to WT values. However, 1 microm propranolol increased epicardial APD(90) to 72.5 +/- 1.2 ms and decreased endocardial APD(90) from 50.9 +/- 1.0 to 24.5 +/- 0.3 ms, increasing DeltaAPD(90) to -48.0 +/- 1.2 ms. CONCLUSION: These findings empirically implicate EADs in potentially initiating spontaneous arrhythmogenic phenomena and transmural repolarization gradients in the re-entrant substrate that would sustain such activity when provoked by extrasystolic activity in murine hearts modelling human LQT3 syndrome.


Subject(s)
Heart/drug effects , Heart/physiology , Long QT Syndrome/metabolism , Propranolol/pharmacology , Sodium Channels/metabolism , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Cardiac Electrophysiology , Dose-Response Relationship, Drug , Humans , Mice , NAV1.5 Voltage-Gated Sodium Channel
16.
Acta Physiol (Oxf) ; 193(1): 25-36, 2008 May.
Article in English | MEDLINE | ID: mdl-18005217

ABSTRACT

AIM: We explored the anti-arrhythmic efficacy of K(+) channel activation in the hypokalaemic murine heart using NS1643 and nicorandil, compounds which augment I(Kr) and I(KATP) respectively. METHODS: Left ventricular epicardial and endocardial monophasic action potentials were compared in normokalaemic and hypokalaemic preparations in the absence and presence of NS1643 (30 microM) and nicorandil (20 microM). RESULTS: Spontaneously beating hypokalaemic hearts (3 mM K(+)) all elicited early afterdepolarizations (EADs) and episodes of ventricular tachycardia (VT). Perfusion with NS1643 and nicorandil suppressed EADs and VT in 7 of 13 and five of six hypokalaemic hearts. Provoked arrhythmia studies using programmed electrical stimulation induced VT in all hypokalaemic hearts, but failed to do so in 7 of 13 and five of six hearts perfused with NS1643 and nicorandil respectively. These anti-arrhythmic effects were accompanied by reductions in action potential duration at 90% repolarization (APD(90)) and changes in the transmural gradient of repolarization, reflected in DeltaAPD(90). NS1643 and nicorandil reduced epicardial APD(90) from 68.3 +/- 1.1 to 56.5 +/- 4.1 and 51.5 +/- 1.5 ms, respectively, but preserved endocardial APD(90) in hypokalaemic hearts. NS1643 and nicorandil thus restored DeltaAPD(90) from -9.6 +/- 4.3 ms under baseline hypokalaemic conditions to 3.9 +/- 4.1 and 9.9 +/- 2.1 ms, respectively, close to normokalaemic values. CONCLUSION: These findings demonstrate, for the first time, the anti-arrhythmic efficacy of K(+) channel activation in the setting of hypokalaemia. NS1643 and nicorandil are anti-arrhythmic through the suppression of EADs, reductions in APD(90) and restorations of DeltaAPD(90).


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Heart/drug effects , Hypokalemia/physiopathology , Potassium Channels/drug effects , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/physiopathology , Cresols/pharmacology , Electric Stimulation , Female , Heart/physiopathology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Male , Mice , Nicorandil/pharmacology , Organ Culture Techniques , Phenylurea Compounds/pharmacology , Potassium Channels/physiology
17.
Acta Physiol (Oxf) ; 191(1): 43-58, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17524066

ABSTRACT

AIMS: We resolved roles for early afterdepolarizations (EADs) and transmural gradients of repolarization in arrhythmogenesis in Langendorff-perfused hypokalaemic murine hearts paced from the right ventricular epicardium. METHODS: Left ventricular epicardial and endocardial monophasic action potentials (MAPs) and arrhythmogenic tendency were compared in the presence and absence of the L-type Ca(2+) channel blocker nifedipine (10 nm-1 microm) and the calmodulin kinase type II inhibitor KN-93 (2 microm). RESULTS: All the hypokalaemic hearts studied showed prolonged epicardial and endocardial MAPs, decreased epicardial-endocardial APD(90) difference, EADs, triggered beats and ventricular tachycardia (VT) (n = 6). In all spontaneously beating hearts, 100 (but not 10) nm nifedipine reduced both the incidence of EADs and triggered beats from 66.9 +/- 15.7% to 28.3 +/- 8.7% and episodes of VT from 10.8 +/- 6.3% to 1.2 +/- 0.7% of MAPs (n = 6 hearts, P < 0.05); 1 microm nifedipine abolished all these phenomena (n = 6). In contrast programmed electrical stimulation (PES) still triggered VT in six of six hearts with 0, 10 and 100 nm but not 1 microm nifedipine. 1 microm nifedipine selectively reduced epicardial (from 66.1 +/- 3.4 to 46.2 +/- 2.5 ms) but not endocardial APD(90), thereby restoring DeltaAPD(90) from -5.9 +/- 2.5 to 15.5 +/- 3.2 ms, close to normokalaemic values. KN-93 similarly reduced EADs, triggered beats and VT in spontaneously beating hearts to 29.6 +/- 8.9% and 1.7 +/- 1.1% respectively (n = 6) yet permitted PES-induced VT (n = 6), in the presence of a persistently negative DeltaAPD(90). CONCLUSIONS: These findings empirically implicate both EADs and triggered beats alongside arrhythmogenic substrate of DeltaAPD(90) in VT pathogenesis at the whole heart level.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Hypokalemia/metabolism , Action Potentials/drug effects , Animals , Benzylamines/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Electric Stimulation , Female , Homeostasis , Male , Mice , Nifedipine/pharmacology , Perfusion , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology
18.
Acta Physiol (Oxf) ; 189(1): 33-46, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17280555

ABSTRACT

AIM: Hypokalaemia is associated with a lethal form of ventricular tachycardia (VT), torsade de pointes, through pathophysiological mechanisms requiring clarification. METHODS: Left ventricular endocardial and epicardial monophasic action potentials were compared in isolated mouse hearts paced from the right ventricular epicardium perfused with hypokalaemic (3 and 4 mm [K(+)](o)) solutions. Corresponding K(+) currents were compared in whole-cell patch-clamped epicardial and endocardial myocytes. RESULTS: Hypokalaemia prolonged epicardial action potential durations (APD) from mean APD(90)s of 37.2 +/- 1.7 ms (n = 7) to 58.4 +/- 4.1 ms (n =7) and 66.7 +/- 2.1 ms (n = 11) at 5.2, 4 and 3 mm [K(+)](o) respectively. Endocardial APD(90)s correspondingly increased from 51.6 +/- 1.9 ms (n = 7) to 62.8 +/- 2.8 ms (n = 7) and 62.9 +/- 5.9 ms (n = 11) giving reductions in endocardial-epicardial differences, DeltaAPD(90), from 14.4 +/- 2.6 to 4.4 +/- 5.0 and -3.4 +/- 6.0 ms respectively. Early afterdepolarizations (EADs) occurred in epicardia in three of seven spontaneously beating hearts at 4 mm [K(+)](o) with triggered beats followed by episodes of non-sustained VT in nine of 11 preparations at 3 mm. Programmed electrical stimulation never induced arrhythmic events in preparations perfused with normokalemic solutions yet induced VT in two of seven and nine of 11 preparations at 4 and 3 mm [K(+)](o) respectively. Early outward K(+) current correspondingly fell from 73.46 +/- 8.45 to 61.16+/-6.14 pA/pF in isolated epicardial but not endocardial myocytes (n = 9) (3 mm [K(+)](o)). CONCLUSIONS: Hypokalaemic mouse hearts recapitulate the clinical arrhythmogenic phenotype, demonstrating EADs and triggered beats that might initiate VT on the one hand and reduced transmural dispersion of repolarization reflected in DeltaAPD(90) suggesting arrhythmogenic substrate on the other.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Hypokalemia/physiopathology , Ventricular Dysfunction, Left/physiopathology , Action Potentials/physiology , Animals , Bradycardia/physiopathology , Electric Stimulation , Endocardium/physiopathology , Female , Hypokalemia/complications , Male , Mice , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Pericardium/physiopathology , Potassium/physiology , Risk Factors , Tachycardia, Ventricular/physiopathology , Torsades de Pointes/etiology , Torsades de Pointes/physiopathology
19.
Subcell Biochem ; 45: 539-62, 2007.
Article in English | MEDLINE | ID: mdl-18193652

ABSTRACT

Calcium transport and calcium signalling mechanisms in bone cells have, in many cases, been discovered by study of diseases with disordered bone metabolism. Calcium matrix deposition is driven primarily by phosphate production, and disorders in bone deposition include abnormalities in membrane phosphate transport such as in chondrocalcinosis, and defects in phosphate-producing enzymes such as in hypophosphatasia. Matrix removal is driven by acidification, which dissolves the mineral. Disorders in calcium removal from bone matrix by osteoclasts cause osteopetrosis. On the other hand, although bone is central to management of extracellular calcium, bone is not a major calcium sensing organ, although calcium sensing proteins are expressed in both osteoblasts and osteoclasts. Intracellular calcium signals are involved in secondary control including cellular motility and survival, but the relationship of these findings to specific diseases is not clear. Intracellular calcium signals may regulate the balance of cell survival versus proliferation or anabolic functional response as part of signalling cascades that integrate the response to primary signals via cell stretch, estrogen, tyrosine kinase, and tumor necrosis factor receptors.


Subject(s)
Bone Diseases/physiopathology , Calcium Signaling/physiology , Calcium/metabolism , Animals , Biological Transport , Calcification, Physiologic/physiology , Calcium/physiology , Cytosol/physiology , Humans , Osteoblasts/metabolism , Osteoclasts/metabolism , Protons , Receptors, Calcium-Sensing/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Vacuolar Proton-Translocating ATPases/physiology
20.
Cell Mol Biol (Noisy-le-grand) ; 52(3): 33-43, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-17535752

ABSTRACT

Calcitonin is a thirty-two amino acid peptide that contains an N-terminal disulphide bridge and a C-terminal prolineamide residue. It is released from thyroid parafollicular C-cells and its direct actions on the osteoclast account for its physiological effects whether as a hypocalcaemic agent and a potent inhibitor of bone resorption. These effects likely reflect actions upon a number of specific osteoclast cell surface receptors that initiate intracellular signaling events through both cyclic AMP and calcium mediated second messenger pathways. Studies of its potent anti-resorptive effects have significant translational implications in the management of Paget's bone disease, osteoporosis, and hypercalcaemia. This chapter summarizes major concepts in the synthesis and structure of calcitonin and then proceeds to outline its cellular, molecular actions and therapeutic applications, whilst seeking to provide a reference source. More detailed accounts have been given on different aspects of calcitonin physiology and biochemistry in a number of recent reviews by ourselves and others (155,157, Zaidi et al., 1994; 2002).


Subject(s)
Calcitonin/pharmacology , Calcitonin/physiology , Osteoclasts/physiology , Receptors, Calcitonin/metabolism , Bone Density/drug effects , Bone Resorption/drug therapy , Calcitonin/genetics , Calcitonin/therapeutic use , Humans , Osteitis Deformans/drug therapy , Osteoclasts/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...