Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Am Heart Assoc ; 7(10)2018 05 02.
Article in English | MEDLINE | ID: mdl-29720499

ABSTRACT

BACKGROUND: Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a familial arrhythmogenic syndrome characterized by sudden death. There are several genetic forms of CPVT associated with mutations in genes encoding the cardiac ryanodine receptor (RyR2) and its auxiliary proteins including calsequestrin (CASQ2) and calmodulin (CaM). It has been suggested that impairment of the ability of RyR2 to stay closed (ie, refractory) during diastole may be a common mechanism for these diseases. Here, we explore the possibility of engineering CaM variants that normalize abbreviated RyR2 refractoriness for subsequent viral-mediated delivery to alleviate arrhythmias in non-CaM-related CPVT. METHODS AND RESULTS: To that end, we have designed a CaM protein (GSH-M37Q; dubbed as therapeutic CaM or T-CaM) that exhibited a slowed N-terminal Ca dissociation rate and prolonged RyR2 refractoriness in permeabilized myocytes derived from CPVT mice carrying the CASQ2 mutation R33Q. This T-CaM was introduced to the heart of R33Q mice through recombinant adeno-associated viral vector serotype 9. Eight weeks postinfection, we performed confocal microscopy to assess Ca handling and recorded surface ECGs to assess susceptibility to arrhythmias in vivo. During catecholamine stimulation with isoproterenol, T-CaM reduced isoproterenol-promoted diastolic Ca waves in isolated CPVT cardiomyocytes. Importantly, T-CaM exposure abolished ventricular tachycardia in CPVT mice challenged with catecholamines. CONCLUSIONS: Our results suggest that gene transfer of T-CaM by adeno-associated viral vector serotype 9 improves myocyte Ca handling and alleviates arrhythmias in a calsequestrin-associated CPVT model, thus supporting the potential of a CaM-based antiarrhythmic approach as a therapeutic avenue for genetically distinct forms of CPVT.


Subject(s)
Calmodulin/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Heart Rate , Tachycardia, Ventricular/therapy , Animals , Calcium Signaling , Calmodulin/biosynthesis , Calsequestrin/deficiency , Calsequestrin/genetics , Disease Models, Animal , Genetic Predisposition to Disease , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Phenotype , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology
2.
Eur Heart J ; 36(11): 686-97, 2015 Mar 14.
Article in English | MEDLINE | ID: mdl-24216388

ABSTRACT

AIMS: Loss-of-function mutations in Calsequestrin 2 (CASQ2) are associated with catecholaminergic polymorphic ventricular tachycardia (CPVT). CPVT patients also exhibit bradycardia and atrial arrhythmias for which the underlying mechanism remains unknown. We aimed to study the sinoatrial node (SAN) dysfunction due to loss of CASQ2. METHODS AND RESULTS: In vivo electrocardiogram (ECG) monitoring, in vitro high-resolution optical mapping, confocal imaging of intracellular Ca(2+) cycling, and 3D atrial immunohistology were performed in wild-type (WT) and Casq2 null (Casq2(-/-)) mice. Casq2(-/-) mice exhibited bradycardia, SAN conduction abnormalities, and beat-to-beat heart rate variability due to enhanced atrial ectopic activity both at baseline and with autonomic stimulation. Loss of CASQ2 increased fibrosis within the pacemaker complex, depressed primary SAN activity, and conduction, but enhanced atrial ectopic activity and atrial fibrillation (AF) associated with macro- and micro-reentry during autonomic stimulation. In SAN myocytes, CASQ2 deficiency induced perturbations in intracellular Ca(2+) cycling, including abnormal Ca(2+) release, periods of significantly elevated diastolic Ca(2+) levels leading to pauses and unstable pacemaker rate. Importantly, Ca(2+) cycling dysfunction occurred not only at the SAN cellular level but was also globally manifested as an increased delay between action potential (AP) and Ca(2+) transient upstrokes throughout the atrial pacemaker complex. CONCLUSIONS: Loss of CASQ2 causes abnormal sarcoplasmic reticulum Ca(2+) release and selective interstitial fibrosis in the atrial pacemaker complex, which disrupt SAN pacemaking but enhance latent pacemaker activity, create conduction abnormalities and increase susceptibility to AF. These functional and extensive structural alterations could contribute to SAN dysfunction as well as AF in CPVT patients.


Subject(s)
Atrial Fibrillation/genetics , Bradycardia/genetics , Calsequestrin/genetics , Gene Deletion , Sarcoplasmic Reticulum/metabolism , Sinoatrial Node/physiology , Action Potentials/physiology , Animals , Atrial Function/genetics , Calcium/metabolism , Calsequestrin/deficiency , Cardiomegaly/genetics , Fibrosis/genetics , Gene Knockout Techniques , Mice, Transgenic , Sinoatrial Node/pathology
3.
Biochem J ; 461(1): 99-106, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24758151

ABSTRACT

CASQ2 (cardiac calsequestrin) is commonly believed to serve as the SR (sarcoplasmic reticulum) luminal Ca2+ sensor. Ablation of CASQ2 promotes SCWs (spontaneous Ca2+ waves) and CPVT (catecholaminergic polymorphic ventricular tachycardia) upon stress but not at rest. How SCWs and CPVT are triggered by stress in the absence of the CASQ2-based luminal Ca2+ sensor is an important unresolved question. In the present study, we assessed the role of the newly identified RyR2 (ryanodine receptor 2)-resident luminal Ca2+ sensor in determining SCW propensity, CPVT susceptibility and cardiac hypertrophy in Casq2-KO (knockout) mice. We crossbred Casq2-KO mice with RyR2 mutant (E4872Q+/-) mice, which lack RyR2-resident SR luminal Ca2+ sensing, to generate animals with both deficiencies. Casq2+/- and Casq2-/- mice showed stress-induced VTs (ventricular tachyarrhythmias), whereas Casq2+/-/E4872Q+/- and Casq2-/-/E4872Q+/- mice displayed little or no stress-induced VTs. Confocal Ca2+ imaging revealed that Casq2-/- hearts frequently exhibited SCWs after extracellular Ca2+ elevation or adrenergic stimulation, whereas Casq2-/-/E4872Q+/- hearts had few or no SCWs under the same conditions. Cardiac hypertrophy developed and CPVT susceptibility increased with age in Casq2-/- mice, but not in Casq2-/-/E4872Q+/- mice. However, the amplitudes and dynamics of voltage-induced Ca2+ transients in Casq2-/- and Casq2-/-/E4872Q+/- hearts were not significantly different. Our results indicate that SCWs, CPVT and hypertrophy in Casq2-null cardiac muscle are governed by the RyR2-resident luminal Ca2+ sensor. This implies that defects in CASQ2-based lumi-nal Ca2+ sensing can be overridden by the RyR2-resident luminal Ca2+ sensor. This makes this RyR2-resident sensor a promising molecular target for the treatment of Ca2+-mediated arrhythmias.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Calsequestrin/deficiency , Cardiomegaly/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Tachycardia, Ventricular/metabolism , Animals , Calsequestrin/genetics , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Mice , Mice, Knockout , Mutation/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/physiopathology
4.
PLoS One ; 8(10): e74919, 2013.
Article in English | MEDLINE | ID: mdl-24098358

ABSTRACT

Mitochondrial calcium handling and its relation with calcium released from sarcoplasmic reticulum (SR) in muscle tissue are subject of lively debate. In this study we aimed to clarify how the SR determines mitochondrial calcium handling using dCASQ-null mice which lack both isoforms of the major Ca(2+)-binding protein inside SR, calsequestrin. Mitochondrial free Ca(2+)-concentration ([Ca(2+)]mito) was determined by means of a genetically targeted ratiometric FRET-based probe. Electron microscopy revealed a highly significant increase in intermyofibrillar mitochondria (+55%) and augmented coupling (+12%) between Ca(2+) release units of the SR and mitochondria in dCASQ-null vs. WT fibers. Significant differences in the baseline [Ca(2+)]mito were observed between quiescent WT and dCASQ-null fibers, but not in the resting cytosolic Ca(2+) concentration. The rise in [Ca(2+)]mito during electrical stimulation occurred in 20-30 ms, while the decline during and after stimulation was governed by 4 rate constants of approximately 40, 1.6, 0.2 and 0.03 s(-1). Accordingly, frequency-dependent increase in [Ca(2+)]mito occurred during sustained contractions. In dCASQ-null fibers the increases in [Ca(2+)]mito were less pronounced than in WT fibers and even lower when extracellular calcium was removed. The amplitude and duration of [Ca(2+)]mito transients were increased by inhibition of mitochondrial Na(+)/Ca(2+) exchanger (mNCX). These results provide direct evidence for fast Ca(2+) accumulation inside the mitochondria, involvement of the mNCX in mitochondrial Ca(2+)-handling and a dependence of mitochondrial Ca(2+)-handling on intracellular (SR) and external Ca(2+) stores in fast skeletal muscle fibers. dCASQ-null mice represent a model for malignant hyperthermia. The differences in structure and in mitochondrial function observed relative to WT may represent compensatory mechanisms for the disease-related reduction of calcium storage capacity of the SR and/or SR Ca(2+)-leakage.


Subject(s)
Calcium/metabolism , Calsequestrin/deficiency , Calsequestrin/genetics , Gene Deletion , Mitochondria/metabolism , Muscle Fibers, Skeletal/cytology , Animals , Cytosol/metabolism , Electric Stimulation , Kinetics , Mice , Mice, Inbred C57BL
5.
PLoS One ; 8(10): e77633, 2013.
Article in English | MEDLINE | ID: mdl-24143248

ABSTRACT

Store-operated calcium entry (SOCE) channels play an important role in Ca(2+) signaling. Recently, excessive SOCE was proposed to play a central role in the pathogenesis of malignant hyperthermia (MH), a pharmacogenic disorder of skeletal muscle. We tested this hypothesis by characterizing SOCE current (ISkCRAC) magnitude, voltage dependence, and rate of activation in myotubes derived from two mouse models of anesthetic- and heat-induced sudden death: 1) type 1 ryanodine receptor (RyR1) knock-in mice (Y524S/+) and 2) calsequestrin 1 and 2 double knock-out (dCasq-null) mice. ISkCRAC voltage dependence and magnitude at -80 mV were not significantly different in myotubes derived from wild type (WT), Y524S/+ and dCasq-null mice. However, the rate of ISkCRAC activation upon repetitive depolarization was significantly faster at room temperature in myotubes from Y524S/+ and dCasq-null mice. In addition, the maximum rate of ISkCRAC activation in dCasq-null myotubes was also faster than WT at more physiological temperatures (35-37°C). Azumolene (50 µM), a more water-soluble analog of dantrolene that is used to reverse MH crises, failed to alter ISkCRAC density or rate of activation. Together, these results indicate that while an increased rate of ISkCRAC activation is a common characteristic of myotubes derived from Y524S/+ and dCasq-null mice and that the protective effects of azumolene are not due to a direct inhibition of SOCE channels.


Subject(s)
Acceleration , Anesthetics/adverse effects , Calcium/metabolism , Death, Sudden/etiology , Electrophysiological Phenomena/drug effects , Hot Temperature/adverse effects , Muscle Fibers, Skeletal/metabolism , Animals , Calcium Channels/metabolism , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Calsequestrin/deficiency , Calsequestrin/genetics , Disease Models, Animal , Gene Knock-In Techniques , Gene Knockout Techniques , Imidazoles/adverse effects , Mice , Muscle Fibers, Skeletal/drug effects , Oxazoles/adverse effects , Ryanodine Receptor Calcium Release Channel/genetics
6.
Circ Res ; 112(4): 689-97, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23295832

ABSTRACT

RATIONALE: Catecholaminergic polymorphic ventricular tachycardia (CPVT) is caused by mutations in cardiac ryanodine receptor (RyR2) or calsequestrin (Casq2) genes. Sinoatrial node dysfunction associated with CPVT may increase the risk for ventricular arrhythmia (VA). OBJECTIVE: To test the hypothesis that CPVT is suppressed by supraventricular overdrive stimulation. METHODS AND RESULTS: Using CPVT mouse models (Casq2(-/-) and RyR2(R4496C/+) mice), the effect of increasing sinus heart rate was tested by pretreatment with atropine and by atrial overdrive pacing. Increasing intrinsic sinus rate with atropine before catecholamine challenge suppressed ventricular tachycardia in 86% of Casq2(-/-) mice (6/7) and significantly reduced the VA score (atropine: 0.6±0.2 versus vehicle: 1.7±0.3; P<0.05). Atrial overdrive pacing completely prevented VA in 16 of 19 (84%) Casq2(-/-) and in 7 of 8 (88%) RyR2(R4496C/+) mice and significantly reduced ventricular premature beats in both CPVT models (P<0.05). Rapid pacing also prevented spontaneous calcium waves and triggered beats in isolated CPVT myocytes. In humans, heart rate dependence of CPVT was evaluated by screening a CPVT patient registry for antiarrhythmic drug-naïve individuals that reached >85% of their maximum-predicted heart rate during exercise testing. All 18 CPVT patients who fulfilled the inclusion criteria exhibited VA before reaching 87% of maximum heart rate. In 6 CPVT patients (33%), VA were paradoxically suppressed as sinus heart rates increased further with continued exercise. CONCLUSIONS: Accelerated supraventricular rates suppress VAs in 2 CPVT mouse models and in a subset of CPVT patients. Hypothetically, atrial overdrive pacing may be a therapy for preventing exercise-induced ventricular tachycardia in treatment-refractory CPVT patients.


Subject(s)
Heart Rate , Adult , Animals , Atropine/pharmacology , Atropine/therapeutic use , Bradycardia/genetics , Bradycardia/physiopathology , Caffeine/toxicity , Calcium Signaling/physiology , Calsequestrin/deficiency , Calsequestrin/genetics , Calsequestrin/physiology , Cardiac Pacing, Artificial , Exercise Test , Heart Rate/drug effects , Humans , Isoproterenol/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Random Allocation , Ryanodine Receptor Calcium Release Channel/deficiency , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/physiology , Sinoatrial Node/physiopathology , Sympathectomy, Chemical , Tachycardia, Ventricular , Vagus Nerve/drug effects , Vagus Nerve/physiopathology , Ventricular Premature Complexes/etiology , Ventricular Premature Complexes/prevention & control
7.
Cardiovasc Res ; 98(2): 297-306, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23135969

ABSTRACT

AIMS: Although aberrant Ca(2+) release (i.e. Ca(2+) 'leak') from the sarcoplasmic reticulum (SR) through cardiac ryanodine receptors (RyR2) is linked to heart failure (HF), it remains unknown whether and under what conditions SR-derived Ca(2+) can actually cause HF. We tested the hypothesis that combining dysregulated RyR2 function with facilitated Ca(2+) uptake into SR will exacerbate abnormal SR Ca(2+) release and induce HF. We also examined the mechanisms for these alterations. METHODS AND RESULTS: We crossbred mice deficient in expression of cardiac calsequestrin (CASQ2) with mice overexpressing the skeletal muscle isoform of SR Ca(2+)ATPase (SERCA1a). The new double-mutant strains displayed early mortality, congestive HF with left ventricular dilated hypertrophy, and decreased ejection fraction. Intact right ventricular muscle preparations from double-mutant mice preserved normal systolic contractile force but were susceptible to spontaneous contractions. Double-mutant cardiomyocytes while preserving normal amplitude of systolic Ca(2+) transients displayed marked disturbances in diastolic Ca(2+) handling in the form of multiple, periodic Ca(2+) waves and wavelets. Dysregulated myocyte Ca(2+) handling and structural and functional cardiac pathology in double-mutant mice were associated with increased rate of apoptotic cell death. Qualitatively similar results were obtained in a hybrid strain created by crossing CASQ2 knockout mice with mice deficient in phospholamban. CONCLUSION: We demonstrate that enhanced SR Ca(2+) uptake combined with dysregulated RyR2s results in sustained diastolic Ca(2+) release causing apoptosis, dilated cardiomyopathy, and early mortality. Our data also suggest that up-regulation of SERCA activity must be advocated with caution as a therapy for HF in the context of abnormal RyR2 function.


Subject(s)
Calcium/metabolism , Calsequestrin/physiology , Cardiomegaly/etiology , Myocardial Contraction , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Apoptosis , Arrhythmias, Cardiac/etiology , Calsequestrin/deficiency , Cardiomegaly/metabolism , Diastole , Heart Failure/etiology , Male , Mice , Myocytes, Cardiac/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology , Up-Regulation , Ventricular Remodeling
8.
J Appl Physiol (1985) ; 113(11): 1677-83, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23042908

ABSTRACT

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a lethal ventricular arrhythmia evoked by physical or emotional stress. Recessively inherited CPVT is caused by either missense or null-allele mutations in the cardiac calsequestrin (CASQ2) gene. It was suggested that defects in CASQ2 cause protein deficiency and impair Ca(2+) uptake to the sarcoplasmic reticulum and Ca(2+)-dependent inhibition of ryanodine channels, leading to diastolic Ca(2+) leak, after-depolarizations, and arrhythmia. To examine the effect of exercise training on left ventricular remodeling and arrhythmia, CASQ2 knockout (KO) mice and wild-type controls underwent echocardiography and heart rhythm telemetry before and after 6 wk of training by treadmill exercise. qRT-PCR and Western blotting were used to measure gene and protein expression. Left ventricular fractional shortening was impaired in KO (33 ± 5 vs. 51 ± 7% in controls, P < 0.05) and improved after training (43 ± 12 and 51 ± 9% in KO and control mice, respectively, P = nonsignificant). The exercise tolerance was low in KO mice (16 ± 1 vs. 29 ± 2 min in controls, P < 0.01), but improved in trained animals (26 ± 2 vs. 30 ± 3 min, P = nonsignificant). The hearts of KO mice had a higher basal expression of the brain natriuretic peptide gene. After training, the expression of natriuretic peptide genes markedly decreased, with no difference between KO and controls. Exercise training was not associated with a change in ventricular tachycardia prevalence, but appeared to reduce arrhythmia load, as manifested by a decrease in ventricular beats during stress. We conclude that, in KO mice, which recapitulate the phenotype of human CPVT2, exercise training is well tolerated and could offer a strategy for heart conditioning against stress-induced arrhythmia.


Subject(s)
Exercise Therapy , Tachycardia, Ventricular/therapy , Ventricular Function, Left , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Blotting, Western , Calsequestrin/deficiency , Calsequestrin/genetics , Disease Models, Animal , Exercise Tolerance , Gene Expression Regulation , Genetic Predisposition to Disease , Male , Mice , Mice, Knockout , Myocardial Contraction , Myocardium/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Phenotype , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Recovery of Function , Reverse Transcriptase Polymerase Chain Reaction , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Time Factors , Ventricular Remodeling
9.
J Biomed Biotechnol ; 2011: 634075, 2011.
Article in English | MEDLINE | ID: mdl-21941434

ABSTRACT

We compared structure and function of EDL and Soleus muscles in adult (4-6 m) mice lacking both Calsequestrin (CASQ) isoforms, the main SR Ca²âº-binding proteins. Lack of CASQ induced ultrastructural alterations in ~30% of Soleus fibers, but not in EDL. Twitch time parameters were prolonged in both muscles, although tension was not reduced. However, when stimulated for 2 sec at 100 hz, Soleus was able to sustain contraction, while in EDL active tension declined by 70-80%. The results presented in this paper unmask a differential effect of CASQ1&2 ablation in fast versus slow fibers. CASQ is essential in EDL to provide large amount of Ca²âº released from the SR during tetanic stimulation. In contrast, Soleus deals much better with lack of CASQ because slow fibers require lower Ca²âº amounts and slower cycling to function properly. Nevertheless, Soleus suffers more severe structural damage, possibly because SR Ca²âº leak is more pronounced.


Subject(s)
Calcium-Binding Proteins/deficiency , Calsequestrin/deficiency , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Analysis of Variance , Animals , Body Weight , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calsequestrin/genetics , Calsequestrin/metabolism , Female , Histocytochemistry , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction , Muscle Fibers, Fast-Twitch/chemistry , Muscle Fibers, Slow-Twitch/chemistry , Muscle Strength , Muscle Tonus , Protein Isoforms
10.
J Mol Cell Cardiol ; 51(5): 760-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21798265

ABSTRACT

Ca(2+) waves can trigger ventricular arrhythmias such as catecholaminergic-polymorphic ventricular tachycardia (CPVT). Drugs that prevent Ca(2+) waves may have antiarrhythmic properties. Here, we use permeabilized ventricular myocytes from a CPVT mouse model lacking calsequestrin (casq2) to screen all clinically available class I antiarrhythmic drugs and selected other antiarrhythmic agents for activity against Ca(2+) waves. Casq2-/- myocytes were imaged in line-scan mode and the following Ca(2+) wave parameters analyzed: wave incidence, amplitude, frequency, and propagation speed. IC(50) (potency) and maximum inhibition (efficacy) were calculated for each drug. Drugs fell into 3 distinct categories. Category 1 drugs (flecainide and R-propafenone) suppressed wave parameters with the highest potency (IC(50)<10 µM) and efficacy (>50% maximum wave inhibition). Category 2 drugs (encainide, quinidine, lidocaine, and verapamil) had intermediate potency (IC(50) 20-40 µM) and efficacy (20-40% maximum wave inhibition). Category 3 drugs (procainamide, disopyramide, mexiletine, cibenzoline, and ranolazine) had no significant effects on Ca(2+) waves at the highest concentration tested (100 µM). Propafenone was stereoselective, with R-propafenone suppressing waves more potently than S-propafenone (IC(50): R-propafenone 2 ± 0.2 µM vs. S-propafenone 54 ± 18 µM). Both flecainide and R-propafenone decreased Ca(2+) spark mass and converted propagated Ca(2+) waves into non-propagated wavelets and frequent sparks, suggesting that reduction in spark mass, not spark frequency, was responsible for wave suppression. Among all class I antiarrhythmic drugs, flecainide and R-propafenone inhibit Ca(2+) waves with the highest potency and efficacy. Permeabilized casq2-/- myocytes are a simple in-vitro assay for finding drugs with activity against Ca(2+) waves. This article is part of a Special Issue entitled 'Possible Editorial'.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Calcium Signaling/physiology , Calcium/metabolism , Calsequestrin/deficiency , Flecainide/pharmacology , Myocytes, Cardiac , Tachycardia, Ventricular/metabolism , Animals , Calcium Signaling/drug effects , Calsequestrin/genetics , Cell Membrane Permeability/drug effects , Cells, Cultured , Disease , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Mice , Mice, Knockout , Microscopy, Confocal , Molecular Imaging , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Propafenone/pharmacology , Saponins/pharmacology , Stereoisomerism , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/pathology , Tachycardia, Ventricular/physiopathology
11.
Circ Arrhythm Electrophysiol ; 4(2): 128-35, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21270101

ABSTRACT

BACKGROUND: Catecholaminergic polymorphic ventricular tachycardia (CPVT) is caused by mutations in the cardiac ryanodine receptor (RyR2) or calsequestrin (Casq2) and can be difficult to treat. The class Ic antiarrhythmic drug flecainide blocks RyR2 channels and prevents CPVT in mice and humans. It is not known whether other class I antiarrhythmic drugs also block RyR2 channels and to what extent RyR2 channel inhibition contributes to antiarrhythmic efficacy in CPVT. METHODS AND RESULTS: We first measured the effect of all class I antiarrhythmic drugs marketed in the United States (quinidine, procainamide, disopyramide, lidocaine, mexiletine, flecainide, and propafenone) on single RyR2 channels incorporated into lipid bilayers. Only flecainide and propafenone inhibited RyR2 channels, with the S-enantiomer of propafenone having a significantly lower potency than R-propafenone or flecainide. In Casq2(-/-) myocytes, the propafenone enantiomers and flecainide significantly reduced arrhythmogenic Ca(2+) waves at clinically relevant concentrations, whereas Na(+) channel inhibitors without RyR2 blocking properties did not. In Casq2(-/-) mice, 5 mg/kg R-propafenone or 20 mg/kg S-propafenone prevented exercise-induced CPVT, whereas procainamide (20 mg/kg) or lidocaine (20 mg/kg) were ineffective (n=5 to 9 mice, P<0.05). QRS duration was not significantly different, indicating a similar degree of Na(+) channel inhibition. Clinically, propafenone (900 mg/d) prevented ICD shocks in a 22-year-old CPVT patient who had been refractory to maximal standard drug therapy and bilateral stellate ganglionectomy. CONCLUSIONS: RyR2 cardiac Ca(2+) release channel inhibition appears to determine efficacy of class I drugs for the prevention of CPVT in Casq2(-/-) mice. Propafenone may be an alternative to flecainide for CPVT patients symptomatic on ß-blockers.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Calcium Channel Blockers/pharmacology , Flecainide/pharmacology , Myocytes, Cardiac/drug effects , Propafenone/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Analysis of Variance , Animals , Calsequestrin/deficiency , Calsequestrin/genetics , Defibrillators, Implantable , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Countershock/instrumentation , Electrocardiography , Humans , Ion Channel Gating/drug effects , Lidocaine/pharmacology , Male , Mice , Mice, Knockout , Mutation, Missense , Myocytes, Cardiac/metabolism , Procainamide/pharmacology , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sodium Channel Blockers/pharmacology , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Time Factors , Young Adult
12.
J Cardiovasc Electrophysiol ; 22(3): 316-24, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20807279

ABSTRACT

BACKGROUND: Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a familial arrhythmic syndrome caused by mutations in genes encoding the calcium-regulation proteins cardiac ryanodine receptor (RyR2) or calsequestrin-2 (CASQ2). Mechanistic studies indicate that CPVT is mediated by diastolic Ca(2+) overload and increased Ca(2+) leak through the RyR2 channel, implying that treatment targeting these defects might be efficacious in CPVT. METHOD AND RESULTS: CPVT mouse models that lack CASQ2 were treated with Ca(2+) -channel inhibitors, ß-adrenergic inhibitors, or Mg(2+) . Treatment effects on ventricular arrhythmia, sarcoplasmic reticulum (SR) protein expression and Ca(2+) transients of isolated myocytes were assessed. Each study agent reduced the frequency of stress-induced ventricular arrhythmia in mutant mice. The Ca(2+) channel blocker verapamil was most efficacious and completely prevented arrhythmia in 85% of mice. Verapamil significantly increased the SR Ca(2+) content in mutant myocytes, diminished diastolic Ca(2+) overload, increased systolic Ca(2+) amplitude, and prevented Ca(2+) oscillations in stressed mutant myocytes. CONCLUSIONS: Ca(2+) channel inhibition by verapamil rectified abnormal calcium handling in CPVT myocytes and prevented ventricular arrhythmias. Verapamil-induced partial normalization of SR Ca(2+) content in mutant myocytes implicates CASQ2 as modulator of RyR2 activity, rather than or in addition to, Ca(2+) buffer protein. Agents such as verapamil that attenuate cardiomyocyte calcium overload are appropriate for assessing clinical efficacy in human CPVT.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Calsequestrin/metabolism , Mutation , Myocytes, Cardiac/drug effects , Verapamil/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Calsequestrin/deficiency , Calsequestrin/genetics , Diltiazem/pharmacology , Disease Models, Animal , Electrocardiography , Gene Knock-In Techniques , Magnesium/pharmacology , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Propranolol/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/prevention & control , Time Factors
13.
J Gen Physiol ; 136(3): 325-38, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20713548

ABSTRACT

Contractile activation in striated muscles requires a Ca(2+) reservoir of large capacity inside the sarcoplasmic reticulum (SR), presumably the protein calsequestrin. The buffering power of calsequestrin in vitro has a paradoxical dependence on [Ca(2+)] that should be valuable for function. Here, we demonstrate that this dependence is present in living cells. Ca(2+) signals elicited by membrane depolarization under voltage clamp were compared in single skeletal fibers of wild-type (WT) and double (d) Casq-null mice, which lack both calsequestrin isoforms. In nulls, Ca(2+) release started normally, but the store depleted much more rapidly than in the WT. This deficit was reflected in the evolution of SR evacuability, E, which is directly proportional to SR Ca(2+) permeability and inversely to its Ca(2+) buffering power, B. In WT mice E starts low and increases progressively as the SR is depleted. In dCasq-nulls, E started high and decreased upon Ca(2+) depletion. An elevated E in nulls is consistent with the decrease in B expected upon deletion of calsequestrin. The different value and time course of E in cells without calsequestrin indicate that the normal evolution of E reflects loss of B upon SR Ca(2+) depletion. Decrement of B upon SR depletion was supported further. When SR calcium was reduced by exposure to low extracellular [Ca(2+)], release kinetics in the WT became similar to that in the dCasq-null. E became much higher, similar to that of null cells. These results indicate that calsequestrin not only stores Ca(2+), but also varies its affinity in ways that progressively increase the ability of the store to deliver Ca(2+) as it becomes depleted, a novel feedback mechanism of potentially valuable functional implications. The study revealed a surprisingly modest loss of Ca(2+) storage capacity in null cells, which may reflect concurrent changes, rather than detract from the physiological importance of calsequestrin.


Subject(s)
Calcium Signaling , Calcium-Binding Proteins/metabolism , Calcium/metabolism , Calsequestrin/metabolism , Muscle, Skeletal/metabolism , Animals , Buffers , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Calsequestrin/deficiency , Calsequestrin/genetics , Kinetics , Membrane Potentials , Mice , Mice, Knockout , Microscopy, Confocal , Muscle Fibers, Fast-Twitch/metabolism , Patch-Clamp Techniques , Sarcoplasmic Reticulum/metabolism
15.
J Physiol ; 587(Pt 21): 5197-209, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19736296

ABSTRACT

Many physiological processes and pharmacological agents modulate the ryanodine receptor (RyR), the primary sarcoplasmic reticulum (SR) Ca(2+) release channel in the heart. However, how such modulations translate into functional effects during cardiac excitation-contraction coupling (ECC) is much less clear. Using a low dose (250 microM) of caffeine we sensitized the RyR and examined SR Ca(2+) release using dynamic measurements of cytosolic Ca(2+) ([Ca(2+)](i)) and free Ca(2+) within the SR ([Ca(2+)](SR)). In field stimulated (1 Hz) rabbit ventricular myocytes, application of 250 microM caffeine caused an initial 33% increase in SR Ca(2+) release, which was followed by a decrease in SR Ca(2+) load (28%) and steady-state SR Ca(2+) release (23%). To investigate the effects of caffeine on local SR Ca(2+) release, we measured [Ca(2+)](SR) from individual release junctions during ECC as well as during spontaneous Ca(2+) sparks. In intact myocytes during ECC, caffeine increased global fractional SR Ca(2+) release by decreasing the [Ca(2+)](SR) level at which local release terminated by 21%. Similarly, in permeabilized myocytes during spontaneous Ca(2+) sparks, caffeine decreased the [Ca(2+)](SR) level for release termination by 12%. Finally, we examined if Ca(2+) release termination was changed in myocytes from failing hearts, where remodelling processes lead to altered RyR function. In myocytes from failing rabbit hearts, the [Ca(2+)](SR) termination level for Ca(2+) sparks was 13% lower than that of non-failing myocytes. Collectively, these data suggest that altering the termination level for local Ca(2+) release may represent a novel mechanism to increase SR Ca(2+) release and contractility during ECC.


Subject(s)
Calcium Signaling/drug effects , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Caffeine/pharmacology , Calcium/metabolism , Calpain/metabolism , Calsequestrin/deficiency , Cells, Cultured , Central Nervous System Stimulants/pharmacology , Myocytes, Cardiac/drug effects , Rabbits , Sarcoplasmic Reticulum/drug effects
16.
J Physiol ; 587(Pt 20): 4863-72, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19703963

ABSTRACT

During the cardiac cycle, the release of Ca(2+) from the sarcoplasmic reticulum (SR) through the ryanodine receptor (RyR2) channel complex is controlled by the levels of cytosolic and luminal Ca(2+) and alterations in these regulatory processes have been implicated in cardiac disease including arrhythmia. To better understand the mechanisms of regulation of SR Ca(2+) release by Ca(2+) on both sides of the SR membrane, we investigated SR Ca(2+) release in a wide range of cytosolic Ca(2+) concentrations ([Ca(2+)](cyt); 1-100 microm) in permeabilized canine ventricular myocytes by monitoring [Ca(2+)] inside the SR ([Ca(2+)](SR)). Exposing myocytes to activating [Ca(2+)](cyt) resulted in spontaneous oscillations of [Ca(2+)](SR) due to periodic opening and closing of the RyR2s. Elevating [Ca(2+)](cyt) (up to 10 microm) increased the frequency of [Ca(2+)](SR) oscillations; however at higher [Ca(2+)](cyt) (>50 microm) the oscillations diminished due to RyR2s staying perpetually open, resulting in depleted SR. Ablation of cardiac calsequestrin (CASQ2) altered the [Ca(2+)](cyt) dependence of Ca(2+) release oscillations such that oscillations were highly frequent at low [Ca(2+)](cyt) (100 nm) but became diminished at moderate [Ca(2+)](cyt) (10 microm), as determined in myocytes from calsequestrin-null versus wild-type mice. Our results suggest that under conditions of continuous activation by cytosolic Ca(2+), RyR2s can periodically cycle between open and deactivated states due to effects of luminal Ca(2+). Deactivation at reduced [Ca(2+)]SR appears to involve reduction of sensitivity to cytosolic Ca(2+) and might be mediated by CASQ2. Inactivation by cytosolic Ca(2+) plays no detectable role in controlling SR Ca(2+) release.


Subject(s)
Calcium Signaling/drug effects , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Caffeine/pharmacology , Calcium/metabolism , Calpain/metabolism , Calsequestrin/deficiency , Central Nervous System Stimulants/pharmacology , Dogs , Humans , Male , Mice , Myocytes, Cardiac/drug effects , Sarcoplasmic Reticulum/drug effects
18.
J Physiol ; 587(Pt 13): 3101-11, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19403601

ABSTRACT

Since its discovery in 1971, calsequestrin has been recognized as the main Ca(2+) binding protein inside the sarcoplasmic reticulum (SR), the organelle that stores and upon demand mobilizes Ca(2+) for contractile activation of muscle. This article reviews the potential roles of calsequestrin in excitation-contraction coupling of skeletal muscle. It first considers the quantitative demands for a structure that binds Ca(2+) inside the SR in view of the amounts of the ion that must be mobilized to elicit muscle contraction. It briefly discusses existing evidence, largely gathered in cardiac muscle, of two roles for calsequestrin: as Ca(2+) reservoir and as modulator of the activity of Ca(2+) release channels, and then considers the results of an incipient body of work that manipulates the cellular endowment of calsequestrin. The observations include evidence that both the Ca(2+) buffering capacity of calsequestrin in solution and that of the SR in intact cells decay as the free Ca(2+) concentration is lowered. Together with puzzling observations of increase of Ca(2+) inside the SR, in cells or vesicular fractions, upon activation of Ca(2+) release, this is interpreted as evidence that the Ca(2+) buffering in the SR is non-linear, and is optimized for support of Ca(2+) release at the physiological levels of SR Ca(2+) concentration. Such non-linearity of buffering is qualitatively explained by a speculation that puts together ideas first proposed by others. The speculation pictures calsequestrin polymers as 'wires' that both bind Ca(2+) and efficiently deliver it near the release channels. In spite of the kinetic changes, the functional studies reveal that cells devoid of calsequestrin are still capable of releasing large amounts of Ca(2+) into the myoplasm, consistent with the long term viability and apparent good health of mice engineered for calsequestrin ablation. The experiments therefore suggest that other molecules are capable of providing sites for reversible binding of large amounts of Ca(2+) inside the sarcoplasmic reticulum.


Subject(s)
Calcium/metabolism , Calsequestrin/metabolism , Muscle, Skeletal/metabolism , Animals , Binding Sites , Calsequestrin/deficiency , Calsequestrin/genetics , Mice , Mice, Knockout , Models, Biological , Sarcoplasmic Reticulum/metabolism
19.
Eur Biophys J ; 39(1): 27-36, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19434403

ABSTRACT

This review focuses on molecular interactions between calsequestrin, triadin, junctin and the ryanodine receptor in the lumen of the sarcoplasmic reticulum. These interactions modulate changes in Ca(2+) release in response to changes in the Ca(2+) load within the sarcoplasmic reticulum store in striated muscle and are of fundamental importance to Ca(2+) homeostasis, since massive adaptive changes occur when expression of the proteins is manipulated, while mutations in calsequestrin lead to functional changes which can be fatal. We find that calsequestrin plays a different role in the heart and skeletal muscle, enhancing Ca(2+) release in the heart, but depressing Ca(2+) release in skeletal muscle. We also find that triadin and junctin exert independent influences on the ryanodine receptor in skeletal muscle where triadin alone modifies excitation-contraction coupling, while junctin alone supports functional interactions between calsequestrin and the ryanodine receptor.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Calsequestrin/metabolism , Carrier Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Striated/cytology , Muscle, Striated/metabolism , Animals , Calsequestrin/deficiency , Calsequestrin/genetics , Humans , Ryanodine Receptor Calcium Release Channel/metabolism
20.
J Physiol ; 587(Pt 13): 3081-7, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19451205

ABSTRACT

Cardiac calsequestrin (Casq2) and triadin are proteins located in specialized areas of the sarcoplasmic reticulum (SR) where the SR forms junctions with the sarcolemma (junctional SR). Casq2, triadin and junctin form a protein complex that is associated with cardiac ryanodine receptor 2 (RyR2) SR Ca(2+) release channels. This review highlights new insights of the roles of triadin and Casq2 derived from gene-targeted knock-out and knock-in mouse models that have recently become available. Characterization of the mouse models suggests that Casq2's contribution to SR Ca(2+) storage and release during excitation-contraction coupling is largely dispensable. Casq2's primary role appears to be in protecting the heart against premature Ca(2+) release and triggered arrhythmias. Furthermore, both cardiac Casq2 and triadin are important for the structural organization of the SR, which had previously not been recognized. In particular, ablation of triadin causes a 50% reduction in the extent of the junctional SR, which results in impaired excitation-contraction coupling at the level of the myocyte. While catecholamines could normalize contractile function by increasing I(Ca) and SR Ca(2+) content, it comes at the price of an increased risk for spontaneous Ca(2+) releases in triadin knock-out myocytes and catecholamine-induced ventricular arrhythmias in triadin knock-out mice.


Subject(s)
Calsequestrin/physiology , Carrier Proteins/physiology , Heart/physiology , Muscle Proteins/physiology , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/physiopathology , Calcium Signaling , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Calsequestrin/deficiency , Calsequestrin/genetics , Carrier Proteins/genetics , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Knockout , Mice, Transgenic , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/physiology , Models, Cardiovascular , Muscle Proteins/deficiency , Muscle Proteins/genetics , Myocardial Contraction/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...