Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Hum Mol Genet ; 29(3): 369-381, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31816043

ABSTRACT

Huntington's disease (HD) is a dominantly inherited neurodegenerative disease. There is accumulating evidence that HD patients have increased prevalence of conduction abnormalities and compromised sinoatrial node function which could lead to increased risk for arrhythmia. We used mutant Huntingtin (mHTT) expressing bacterial artificial chromosome Huntington's disease mice to determine if they exhibit electrocardiogram (ECG) abnormalities involving cardiac conduction that are known to increase risk of sudden arrhythmic death in humans. We obtained surface ECGs and analyzed arrhythmia susceptibility; we observed prolonged QRS duration, increases in PVCs as well as PACs. Abnormal histological and structural changes that could lead to cardiac conduction system dysfunction were seen. Finally, we observed decreases in desmosomal proteins, plakophilin-2 and desmoglein-2, which have been reported to cause cardiac arrhythmias and reduced conduction. Our study indicates that mHTT could cause progressive cardiac conduction system pathology that could increase the susceptibility to arrhythmias and sudden cardiac death in HD patients.


Subject(s)
Arrhythmias, Cardiac/pathology , Disease Models, Animal , Electrocardiography/methods , Heart Conduction System/pathology , Huntingtin Protein/genetics , Huntington Disease/complications , Neurons/pathology , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Heart Conduction System/metabolism , Humans , Mice , Mice, Transgenic , Neurons/metabolism
2.
Arch Biochem Biophys ; 661: 125-131, 2019 01.
Article in English | MEDLINE | ID: mdl-30445044

ABSTRACT

Mutations in cardiac troponin T (TnT) associated with hypertrophic cardiomyopathy generally lead to an increase in the Ca2+ sensitivity of contraction and susceptibility to arrhythmias. In contrast, TnT mutations linked to dilated cardiomyopathy decrease the Ca2+ sensitivity of contraction. Here we tested the hypothesis that two TnT disease mutations with opposite effects on myofilament Ca2+ sensitivity can attenuate each other's phenotype. We crossed transgenic mice expressing the HCM TnT-I79N mutation (I79N) with a DCM knock-in mouse model carrying the heterozygous TnT-R141W mutation (HET). The results of the Ca2+ sensitivity in skinned cardiac muscle preparations ranked from highest to lowest were as follow: I79N > I79N/HET > NTg > HET. Echocardiographic measurements revealed an improvement in hemodynamic parameters in I79N/HET compared to I79N and normalization of left ventricular dimensions and volumes compared to both I79N and HET. Ex vivo testing showed that the I79N/HET mouse hearts had reduced arrhythmia susceptibility compared to I79N mice. These results suggest that two disease mutations in TnT that have opposite effects on the myofilament Ca2+ sensitivity can paradoxically ameliorate each other's disease phenotype. Normalizing myofilament Ca2+ sensitivity may be a promising new treatment approach for a variety of diseases.


Subject(s)
Cardiomyopathy, Dilated , Echocardiography , Electrocardiography , Mutation, Missense , Myofibrils/metabolism , Troponin T , Amino Acid Substitution , Animals , Arrhythmias, Cardiac/diagnostic imaging , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cardiomyopathy, Dilated/diagnostic imaging , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Mice , Mice, Transgenic , Myofibrils/pathology , Troponin T/genetics , Troponin T/metabolism
3.
Proc Natl Acad Sci U S A ; 112(6): 1880-5, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25583515

ABSTRACT

BDNF and its associated tropomyosin-related kinase receptor B (TrkB) nurture vessels and nerves serving the heart. However, the direct effect of BDNF/TrkB signaling on the myocardium is poorly understood. Here we report that cardiac-specific TrkB knockout mice (TrkB(-/-)) display impaired cardiac contraction and relaxation, showing that BDNF/TrkB signaling acts constitutively to sustain in vivo myocardial performance. BDNF enhances normal cardiomyocyte Ca(2+) cycling, contractility, and relaxation via Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). Conversely, failing myocytes, which have increased truncated TrkB lacking tyrosine kinase activity and chronically activated CaMKII, are insensitive to BDNF. Thus, BDNF/TrkB signaling represents a previously unidentified pathway by which the peripheral nervous system directly and tonically influences myocardial function in parallel with ß-adrenergic control. Deficits in this system are likely additional contributors to acute and chronic cardiac dysfunction.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Calcium Signaling/physiology , Diastole/physiology , Myocardial Contraction/physiology , Receptor, trkB/metabolism , Analysis of Variance , Animals , Calcium/metabolism , Hemodynamics , Immunohistochemistry , Mice , Mice, Knockout , Patch-Clamp Techniques
4.
J Mol Cell Cardiol ; 104: 9-16, 2017 03.
Article in English | MEDLINE | ID: mdl-28131630

ABSTRACT

Functional impact of cardiac ryanodine receptor (type 2 RyR or RyR2) phosphorylation by protein kinase A (PKA) remains highly controversial. In this study, we characterized a functional link between PKA-mediated RyR2 phosphorylation level and sarcoplasmic reticulum (SR) Ca2+ release and leak in permeabilized rabbit ventricular myocytes. Changes in cytosolic [Ca2+] and intra-SR [Ca2+]SR were measured with Fluo-4 and Fluo-5N, respectively. Changes in RyR2 phosphorylation at two PKA sites, serine-2031 and -2809, were measured with phospho-specific antibodies. cAMP (10µM) increased Ca2+ spark frequency approximately two-fold. This effect was associated with an increase in SR Ca2+ load from 0.84 to 1.24mM. PKA inhibitory peptide (PKI; 10µM) abolished the cAMP-dependent increase of SR Ca2+ load and spark frequency. When SERCA was completely blocked by thapsigargin, cAMP did not affect RyR2-mediated Ca2+ leak. The lack of a cAMP effect on RyR2 function can be explained by almost maximal phosphorylation of RyR2 at serine-2809 after sarcolemma permeabilization. This high RyR2 phosphorylation level is likely the consequence of a balance shift between protein kinase and phosphatase activity after permeabilization. When RyR2 phosphorylation at serine-2809 was reduced to its "basal" level (i.e. RyR2 phosphorylation level in intact myocytes) using kinase inhibitor staurosporine, SR Ca2+ leak was significantly reduced. Surprisingly, further dephosphorylation of RyR2 with protein phosphatase 1 (PP1) markedly increased SR Ca2+ leak. At the same time, phosphorylation of RyR2 at serine 2031 did not significantly change under identical experimental conditions. These results suggest that RyR2 phosphorylation by PKA has a complex effect on SR Ca2+ leak in ventricular myocytes. At an intermediate level of RyR2 phosphorylation SR Ca2+ leak is minimal. However, complete dephosphorylation and maximal phosphorylation of RyR2 increases SR Ca2+ leak.


Subject(s)
Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Ventricles/metabolism , Ion Channel Gating , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Calcium Signaling , Cyclic AMP/metabolism , Myocardium/metabolism , Phosphorylation , Rabbits
5.
J Physiol ; 595(12): 3939-3947, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28205229

ABSTRACT

The major goal of this focused review is to highlight some of the recent advances and remaining open questions about how a mutation in a myofilament protein leads to an increased risk for sudden cardiac death (SCD). The link between myofilaments and SCD has been known for over 25 years, but identifying mutation carriers at risk for SCD is still a challenge and currently the only effective prevention is implantation of a defibrillator (ICD). In addition to recognized risk factors, other contributing factors need to be considered and assessed, e.g. 'microvascular dysfunction', to calibrate individual risk more accurately. Similarly, improving our understanding about the underlying mechanisms of SCD in patients with sarcomeric mutations will also allow us to design new and less invasive treatment options that will minimize risk and hopefully make implantation of an ICD unnecessary.


Subject(s)
Death, Sudden, Cardiac/pathology , Myofibrils/metabolism , Animals , Defibrillators, Implantable , Humans , Risk Factors
7.
Circ Res ; 112(9): e88-97, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23513055

ABSTRACT

RATIONALE: The spatial distribution of blood flow in the hearts of genetically modified mice is a phenotype of interest because derangements in blood flow may precede detectable changes in organ function. However, quantifying the regional distribution of blood flow within organs of mice is challenging because of the small organ volume and the high resolution required to observe spatial differences in flow. Traditional microsphere methods in which the numbers of microspheres per region are indirectly estimated from radioactive counts or extracted fluorescence have been limited to larger organs for 2 reasons; to ensure statistical confidence in the measured flow per region and to be able to physically dissect the organ to acquire spatial information. OBJECTIVE: To develop methods to quantify and statistically compare the spatial distribution of blood flow within organs of mice. METHODS AND RESULTS: We developed and validated statistical methods to compare blood flow between regions and with the same regions over time using 15-µm fluorescent microspheres. We then tested this approach by injecting fluorescent microspheres into isolated perfused mice hearts, determining the spatial location of every microsphere in the hearts, and then visualizing regional flow patterns. We demonstrated application of these statistical and visualizing methods in a coronary artery ligation model in mice. CONCLUSIONS: These new methods provide tools to investigate the spatial and temporal changes in blood flow within organs of mice at a much higher spatial resolution than currently available by other methods.


Subject(s)
Coronary Artery Disease/physiopathology , Coronary Circulation , Coronary Vessels/physiopathology , Endocardium/physiopathology , Fluorescent Dyes , Myocardial Perfusion Imaging/methods , Optical Imaging , Pericardium/physiopathology , Animals , Blood Flow Velocity , Computer Simulation , Coronary Vessels/surgery , Disease Models, Animal , Image Interpretation, Computer-Assisted , Ligation , Mice , Microspheres , Models, Statistical , Numerical Analysis, Computer-Assisted , Regional Blood Flow , Time Factors
8.
Circ Res ; 112(10): 1334-44, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23532597

ABSTRACT

RATIONALE: The Ca(2+) sensitivity of the myofilaments is increased in hypertrophic cardiomyopathy and other heart diseases and may contribute to a higher risk for sudden cardiac death. Ca(2+) sensitization increases susceptibility to reentrant ventricular tachycardia in animal models, but the underlying mechanism is unknown. OBJECTIVE: To investigate how myofilament Ca(2+) sensitization creates reentrant arrhythmia susceptibility. METHODS AND RESULTS: Using hypertrophic cardiomyopathy mouse models (troponinT-I79N) and a Ca(2+) sensitizing drug (EMD57033), here we identify focal energy deprivation as a direct consequence of myofilament Ca(2+) sensitization. To detect ATP depletion and thus energy deprivation, we measured accumulation of dephosphorylated Connexin 43 (Cx43) isoform P0 and AMP kinase activation by Western blotting and immunostaining. No differences were detected between groups at baseline, but regional accumulation of Connexin 43 isoform P0 occurred within minutes in all Ca(2+)-sensitized hearts, in vivo after isoproterenol challenge and in isolated hearts after rapid pacing. Lucifer yellow dye spread demonstrated reduced gap junctional coupling in areas with Connexin 43 isoform P0 accumulation. Optical mapping revealed that selectively the transverse conduction velocity was slowed and anisotropy increased. Myofilament Ca(2+) desensitization with blebbistatin prevented focal energy deprivation, transverse conduction velocity slowing, and the reentrant ventricular arrhythmias. CONCLUSIONS: Myofilament Ca(2+) sensitization rapidly leads to focal energy deprivation and reduced intercellular coupling during conditions that raise arrhythmia susceptibility. This is a novel proarrhythmic mechanism that can increase arrhythmia susceptibility in structurally normal hearts within minutes and may, therefore, contribute to sudden cardiac death in diseases with increased myofilament Ca(2+) sensitivity.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Calcium/physiology , Cardiomyopathy, Hypertrophic/physiopathology , Disease Susceptibility/physiopathology , Energy Metabolism/physiology , Myofibrils/physiology , Adenosine Triphosphate/metabolism , Adenylate Kinase/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Cardiotonic Agents/pharmacology , Connexin 43/metabolism , Disease Models, Animal , Disease Susceptibility/metabolism , Electrocardiography , Energy Metabolism/drug effects , Female , Gap Junctions/drug effects , Gap Junctions/physiology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Myofibrils/drug effects , Quinolines/pharmacology , Thiadiazines/pharmacology
9.
Circ Res ; 111(2): 170-9, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22647877

ABSTRACT

RATIONALE: Ca binding to the troponin complex represents a major portion of cytosolic Ca buffering. Troponin mutations that increase myofilament Ca sensitivity are associated with familial hypertrophic cardiomyopathy and confer a high risk for sudden death. In mice, Ca sensitization causes ventricular arrhythmias, but the underlying mechanisms remain unclear. OBJECTIVE: To test the hypothesis that myofilament Ca sensitization increases cytosolic Ca buffering and to determine the resulting arrhythmogenic changes in Ca homeostasis in the intact mouse heart. METHODS AND RESULTS: Using cardiomyocytes isolated from mice expressing troponin T (TnT) mutants (TnT-I79N, TnT-F110I, TnT-R278C), we found that increasing myofilament Ca sensitivity produced a proportional increase in cytosolic Ca binding. The underlying cause was an increase in the cytosolic Ca binding affinity, whereas maximal Ca binding capacity was unchanged. The effect was sufficiently large to alter Ca handling in intact mouse hearts at physiological heart rates, resulting in increased end-diastolic [Ca] at fast pacing rates, and enhanced sarcoplasmic reticulum Ca content and release after pauses. Accordingly, action potential (AP) regulation was altered, with postpause action potential prolongation, afterdepolarizations, and triggered activity. Acute Ca sensitization with EMD 57033 mimicked the effects of Ca-sensitizing TnT mutants and produced pause-dependent ventricular ectopy and sustained ventricular tachycardia after acute myocardial infarction. CONCLUSIONS: Myofilament Ca sensitization increases cytosolic Ca binding affinity. A major proarrhythmic consequence is a pause-dependent potentiation of Ca release, action potential prolongation, and triggered activity. Increased cytosolic Ca binding represents a novel mechanism of pause-dependent arrhythmia that may be relevant for inherited and acquired cardiomyopathies.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium/physiology , Cytosol/metabolism , Homeostasis/genetics , Intracellular Fluid/metabolism , Myofibrils/metabolism , Action Potentials/genetics , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Calcium/metabolism , Calcium Signaling/genetics , Cytosol/pathology , Humans , Mice , Mice, Transgenic , Mutation , Myofibrils/genetics , Protein Binding/genetics , Up-Regulation/genetics
10.
Circ Res ; 106(11): 1743-52, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20431056

ABSTRACT

RATIONALE: FK506-binding proteins FKBP12.6 and FKBP12 are associated with cardiac ryanodine receptors (RyR2), and cAMP-dependent protein kinase A (PKA)-dependent phosphorylation of RyR2 was proposed to interrupt FKBP12.6-RyR2 association and activate RyR2. However, the function of FKBP12.6/12 and role of PKA phosphorylation in cardiac myocytes are controversial. OBJECTIVE: To directly measure in situ binding of FKBP12.6/12 to RyR2 in ventricular myocytes, with simultaneous Ca sparks measurements as a RyR2 functional index. METHODS AND RESULTS: We used permeabilized rat and mouse ventricular myocytes, and fluorescently-labeled FKBP12.6/12. Both FKBP12.6 and FKBP12 concentrate at Z-lines, consistent with RyR2 and Ca spark initiation sites. However, only FKBP12.6 inhibits resting RyR2 activity. Assessment of fluorescent FKBP binding in myocyte revealed a high FKBP12.6-RyR2 affinity (K(d)=0.7+/-0.1 nmol/L) and much lower FKBP12-RyR2 affinity (K(d)=206+/-70 nmol/L). Fluorescence recovery after photobleach confirmed this K(d) difference and showed that it is mediated by k(off). RyR2 phosphorylation by PKA did not alter binding kinetics or affinity of FKBP12.6/12 for RyR2. Using quantitative immunoblots, we determined endogenous [FKBP12] in intact myocytes is approximately 1 micromol/L (similar to [RyR]), whereas [FKBP12.6] is

Subject(s)
Calcium Signaling , Cell Membrane Permeability , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Blotting, Western , Calcium Signaling/drug effects , Circular Dichroism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Fluorescence Recovery After Photobleaching , Heart Ventricles/metabolism , Humans , Kinetics , Mice , Mice, Knockout , Microscopy, Confocal , Mutagenesis, Site-Directed , Mutation , Myocytes, Cardiac/drug effects , Phosphorylation , Protein Binding , Rats , Ryanodine Receptor Calcium Release Channel/drug effects , Sarcoplasmic Reticulum/metabolism , Sirolimus/pharmacology , Swine , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Proteins/deficiency , Tacrolimus Binding Proteins/genetics
11.
Proc Natl Acad Sci U S A ; 106(18): 7636-41, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19383796

ABSTRACT

Heart muscle excitation-contraction (E-C) coupling is governed by Ca(2+) release units (CRUs) whereby Ca(2+) influx via L-type Ca(2+) channels (Cav1.2) triggers Ca(2+) release from juxtaposed Ca(2+) release channels (RyR2) located in junctional sarcoplasmic reticulum (jSR). Although studies suggest that the jSR protein triadin anchors cardiac calsequestrin (Casq2) to RyR2, its contribution to E-C coupling remains unclear. Here, we identify the role of triadin using mice with ablation of the Trdn gene (Trdn(-/-)). The structure and protein composition of the cardiac CRU is significantly altered in Trdn(-/-) hearts. jSR proteins (RyR2, Casq2, junctin, and junctophilin 1 and 2) are significantly reduced in Trdn(-/-) hearts, whereas Cav1.2 and SERCA2a remain unchanged. Electron microscopy shows fragmentation and an overall 50% reduction in the contacts between jSR and T-tubules. Immunolabeling experiments show reduced colocalization of Cav1.2 with RyR2 and substantial Casq2 labeling outside of the jSR in Trdn(-/-) myocytes. CRU function is impaired in Trdn(-/-) myocytes, with reduced SR Ca(2+) release and impaired negative feedback of SR Ca(2+) release on Cav1.2 Ca(2+) currents (I(Ca)). Uninhibited Ca(2+) influx via I(Ca) likely contributes to Ca(2+) overload and results in spontaneous SR Ca(2+) releases upon beta-adrenergic receptor stimulation with isoproterenol in Trdn(-/-) myocytes, and ventricular arrhythmias in Trdn(-/-) mice. We conclude that triadin is critically important for maintaining the structural and functional integrity of the cardiac CRU; triadin loss and the resulting alterations in CRU structure and protein composition impairs E-C coupling and renders hearts susceptible to ventricular arrhythmias.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Calcium/metabolism , Carrier Proteins/physiology , Heart/physiopathology , Muscle Proteins/physiology , Myocardial Contraction , Sarcoplasmic Reticulum/metabolism , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Calcium Channels, L-Type/metabolism , Carrier Proteins/genetics , Heart/physiology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains , Muscle Proteins/genetics , Myocardial Contraction/genetics , Myocardium/metabolism , Myocardium/ultrastructure , Sarcoplasmic Reticulum/ultrastructure , Sequence Deletion
12.
J Mol Cell Cardiol ; 50(1): 230-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20971119

ABSTRACT

Cardiac myocyte overexpression of CaMKIIδ(C) leads to cardiac hypertrophy and heart failure (HF) possibly caused by altered myocyte Ca(2+) handling. A central defect might be the marked CaMKII-induced increase in diastolic sarcoplasmic reticulum (SR) Ca(2+) leak which decreases SR Ca(2+) load and Ca(2+) transient amplitude. We hypothesized that inhibition of CaMKII near the SR membrane would decrease the leak, improve Ca(2+) handling and prevent the development of contractile dysfunction and HF. To test this hypothesis we crossbred CaMKIIδ(C) overexpressing mice (CaMK) with mice expressing the CaMKII-inhibitor AIP targeted to the SR via a modified phospholamban (PLB)-transmembrane-domain (SR-AIP). There was a selective decrease in the amount of activated CaMKII in the microsomal (SR/membrane) fraction prepared from these double-transgenic mice (CaMK/SR-AIP) mice. In ventricular cardiomyocytes from CaMK/SR-AIP mice, SR Ca(2+) leak, assessed both as diastolic Ca(2+) shift into SR upon tetracaine in intact myocytes or integrated Ca(2+) spark release in permeabilized myocytes, was significantly reduced. The reduced leak was accompanied by enhanced SR Ca(2+) load and twitch amplitude in double-transgenic mice (vs. CaMK), without changes in SERCA expression or NCX function. However, despite the improved myocyte Ca(2+) handling, cardiac hypertrophy and remodeling was accelerated in CaMK/SR-AIP and cardiac function worsened. We conclude that while inhibition of SR localized CaMKII in CaMK mice improves Ca(2+) handling, it does not necessarily rescue the HF phenotype. This implies that a non-SR CaMKIIδ(C) exerts SR-independent effects that contribute to hypertrophy and HF, and this CaMKII pathway may be exacerbated by the global enhancement of Ca transients.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Blotting, Western , Echocardiography , Heart Ventricles/cytology , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Phosphorylation
13.
J Mol Cell Cardiol ; 48(5): 824-33, 2010 May.
Article in English | MEDLINE | ID: mdl-20097204

ABSTRACT

Increased myofilament Ca(2+) sensitivity is a common attribute of many inherited and acquired cardiomyopathies that are associated with cardiac arrhythmias. Accumulating evidence supports the concept that increased myofilament Ca(2+) sensitivity is an independent risk factor for arrhythmias. This review describes and discusses potential underlying molecular and cellular mechanisms how myofilament Ca(2+) sensitivity affects cardiac excitation and leads to the generation of arrhythmias. Emphasized are downstream effects of increased myofilament Ca(2+) sensitivity: altered Ca(2+) buffering/handling, impaired energy metabolism and increased mechanical stretch, and how they may contribute to arrhythmogenesis.


Subject(s)
Actin Cytoskeleton/metabolism , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Disease Susceptibility/metabolism , Animals , Humans , Myocardium/metabolism , Myocardium/pathology , Signal Transduction/physiology , Stress, Mechanical
14.
J Mol Cell Cardiol ; 48(2): 293-301, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19835880

ABSTRACT

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is linked to mutations in the cardiac ryanodine receptor (RyR2) or calsequestrin. We recently found that the drug flecainide inhibits RyR2 channels and prevents CPVT in mice and humans. Here we compared the effects of flecainide and tetracaine, a known RyR2 inhibitor ineffective in CPVT myocytes, on arrhythmogenic Ca(2+) waves and elementary sarcoplasmic reticulum (SR) Ca(2+) release events, Ca(2+) sparks. In ventricular myocytes isolated from a CPVT mouse model, flecainide significantly reduced spark amplitude and spark width, resulting in a 40% reduction in spark mass. Surprisingly, flecainide significantly increased spark frequency. As a result, flecainide had no significant effect on spark-mediated SR Ca(2+) leak or SR Ca(2+) content. In contrast, tetracaine decreased spark frequency and spark-mediated SR Ca(2+) leak, resulting in a significantly increased SR Ca(2+) content. Measurements in permeabilized rat ventricular myocytes confirmed the different effects of flecainide and tetracaine on spark frequency and Ca(2+) waves. In lipid bilayers, flecainide inhibited RyR2 channels by open state block, whereas tetracaine primarily prolonged RyR2 closed times. The differential effects of flecainide and tetracaine on sparks and RyR2 gating can explain why flecainide, unlike tetracaine, does not change the balance of SR Ca(2+) fluxes. We suggest that the smaller spark mass contributes to flecainide's antiarrhythmic action by reducing the probability of saltatory wave propagation between adjacent Ca(2+) release units. Our results indicate that inhibition of the RyR2 open state provides a new therapeutic strategy to prevent diastolic Ca(2+) waves resulting in triggered arrhythmias, such as CPVT.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Flecainide/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/metabolism , Cell Membrane Permeability/drug effects , Humans , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Tetracaine/pharmacology
16.
J Am Soc Echocardiogr ; 33(8): 1023-1031.e2, 2020 08.
Article in English | MEDLINE | ID: mdl-32532642

ABSTRACT

BACKGROUND: Ultrasound-mediated cavitation of microbubble contrast agents produces high intravascular shear. We hypothesized that microbubble cavitation increases myocardial microvascular perfusion through shear-dependent purinergic pathways downstream from ATP release that is immediate and sustained through cellular ATP channels such as Pannexin-1. METHODS: Quantitative myocardial contrast echocardiography perfusion imaging and in vivo optical imaging of ATP was performed in wild-type and Pannexin-1-deficient (Panx1-/-) mice before and 5 and 30 minutes after 10 minutes of ultrasound-mediated (1.3 MHz, mechanical index 1.3) myocardial microbubble cavitation. Flow augmentation in a preclinical model closer to humans was evaluated in rhesus macaques undergoing myocardial contrast echocardiography perfusion imaging after high-power cavitation in the apical four-chamber plane for 10 minutes. RESULTS: Microbubble cavitation in wild-type mice (n = 7) increased myocardial perfusion by 64% ± 25% at 5 minutes and 95% ± 55% at 30 minutes compared with baseline (P < .05). In Panx1-/- mice (n = 5), perfusion increased by 28% ± 26% at 5 minutes (P = .04) but returned to baseline at 30 minutes. Myocardial ATP signal in wild-type (n = 7) mice undergoing cavitation compared with sham-treated controls (n = 3) was 450-fold higher at 5 minutes and 90-fold higher at 30 minutes after cavitation (P < .001). The ATP signal in Panx1-/- mice (n = 4) was consistently 10-fold lower than that in wild-type mice and was similar to sham controls at 30 minutes. In macaques (n = 8), myocardial perfusion increased twofold in the cavitation-exposed four-chamber plane, similar in degree to that produced by adenosine, but did not increase in the control two-chamber plane. CONCLUSIONS: Cavitation of microbubbles in the myocardial microcirculation produces an immediate release of ATP, likely from cell microporation, as well as sustained release, which is channel dependent and responsible for persistent flow augmentation. These findings provide mechanistic insight by which cavitation improves perfusion and reduces infarct size in patients with myocardial infarction.


Subject(s)
Contrast Media , Microbubbles , Animals , Connexins , Macaca mulatta , Mice , Mice, Inbred C57BL , Myocardium , Nerve Tissue Proteins , Ultrasonography
17.
Circ Res ; 100(1): 96-104, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17138943

ABSTRACT

Heart failure remains a leading cause of morbidity and mortality worldwide. Although depressed pump function is common, development of effective therapies to stimulate contraction has proven difficult. This is thought to be attributable to their frequent reliance on cAMP stimulation to increase activator Ca(2+). A potential alternative is nitroxyl (HNO), the 1-electron reduction product of nitric oxide (NO) that improves contraction and relaxation in normal and failing hearts in vivo. The mechanism for myocyte effects remains unknown. Here, we show that this activity results from a direct interaction of HNO with the sarcoplasmic reticulum Ca(2+) pump and the ryanodine receptor 2, leading to increased Ca(2+) uptake and release from the sarcoplasmic reticulum. HNO increases the open probability of isolated ryanodine-sensitive Ca(2+)-release channels and accelerates Ca(2+) reuptake into isolated sarcoplasmic reticulum by stimulating ATP-dependent Ca(2+) transport. Contraction improves with no net rise in diastolic calcium. These changes are not induced by NO, are fully reversible by addition of reducing agents (redox sensitive), and independent of both cAMP/protein kinase A and cGMP/protein kinase G signaling. Rather, the data support HNO/thiolate interactions that enhance the activity of intracellular Ca(2+) cycling proteins. These findings suggest HNO donors are attractive candidates for the pharmacological treatment of heart failure.


Subject(s)
Calcium/metabolism , Myocardium/metabolism , Myocytes, Cardiac/physiology , Nitrogen Oxides/metabolism , Sarcoplasmic Reticulum/metabolism , Adenosine Triphosphate/physiology , Animals , Biological Transport/physiology , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Mice , Mice, Inbred C57BL , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nitrites/pharmacology , Nitrogen Oxides/antagonists & inhibitors , Ryanodine Receptor Calcium Release Channel/metabolism , Sulfhydryl Compounds/metabolism
18.
Biochem Biophys Res Commun ; 376(1): 80-5, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18755143

ABSTRACT

The cardiac ryanodine receptor (RyR) controls Ca2+ release from the sarcoplasmic reticulum (SR) during excitation-contraction coupling. Three phosphorylation sites have been identified: Serine-(S)2808, S2814 and recently S2030. We measured phosphorylation with at least two different antibodies per site and demonstrate that for S2808 results were highly antibody-dependent and two out of three S2808 antibodies did not accurately report phosphorylation level. The RyR was substantially phosphorylated in quiescent rat cardiomyocytes at S2808 and less so at S2814, but appeared to be unphosphorylated at S2030. Basal phosphorylation at S2808/S2814 was maintained by a Ca2+ dependent kinase other than Ca2+/Calmodulin-dependent kinase (CaMKII). During stimulation with Isoproterenol S2808 was phosphorylated by protein kinase A (PKA) and S2814 was phosphorylated by CaMKII. Phosphatase 1 appears to be the main phosphatase dephosphorylating S2808/S2814, but phosphatase 2a may also dephosphorylate S2814. RyR phosphorylation is complex, but important in understanding RyR functional modulation.


Subject(s)
Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Serine/metabolism , Animals , Antibodies, Phospho-Specific/immunology , Base Sequence , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Isoproterenol/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation/drug effects , Phosphoserine/immunology , Rats
19.
J Am Heart Assoc ; 7(15): e008939, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30371234

ABSTRACT

Background Oxidative stress-mediated Ca2+/calmodulin-dependent protein kinase II (Ca MKII) phosphorylation of cardiac ion channels has emerged as a critical contributor to arrhythmogenesis in cardiac pathology. However, the link between mitochondrial-derived reactive oxygen species (md ROS ) and increased Ca MKII activity in the context of cardiac arrhythmias has not been fully elucidated and is difficult to establish experimentally. Methods and Results We hypothesize that pathological md ROS can cause erratic action potentials through the oxidation-dependent Ca MKII activation pathway. We further propose that Ca MKII -dependent phosphorylation of sarcolemmal slow Na+ channels alone is sufficient to elicit early afterdepolarizations. To test the hypotheses, we expanded our well-established guinea pig cardiomyocyte excitation- contraction coupling, mitochondrial energetics, and ROS - induced- ROS - release model by incorporating oxidative Ca MKII activation and Ca MKII -dependent Na+ channel phosphorylation in silico. Simulations show that md ROS mediated-Ca MKII activation elicits early afterdepolarizations by augmenting the late Na+ currents, which can be suppressed by blocking L-type Ca2+ channels or Na+/Ca2+ exchangers. Interestingly, we found that oxidative Ca MKII activation-induced early afterdepolarizations are sustained even after md ROS has returned to its physiological levels. Moreover, mitochondrial-targeting antioxidant treatment can suppress the early afterdepolarizations, but only if given in an appropriate time window. Incorporating concurrent md ROS -induced ryanodine receptors activation further exacerbates the proarrhythmogenic effect of oxidative Ca MKII activation. Conclusions We conclude that oxidative Ca MKII activation-dependent Na channel phosphorylation is a critical pathway in mitochondria-mediated cardiac arrhythmogenesis.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Sodium Channels/metabolism , Animals , Computer Simulation , Excitation Contraction Coupling , Guinea Pigs , Phosphorylation , Reactive Oxygen Species/metabolism , Sarcolemma
20.
Cardiovasc Res ; 113(5): 508-518, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28158679

ABSTRACT

AIMS: Fluid shear stress (SS) is known to regulate endothelial cell (EC) function. Most of the studies, however, focused on the effects of cell-free fluid-generated wall SS on ECs. The objective of this study was to investigate how changes in blood flow altered EC signalling and endothelial function directly through wall SS and indirectly through SS effects on red blood cells (RBCs). METHODS AND RESULTS: Experiments were conducted in individually perfused rat venules. We experimentally induced changes in SS that were quantified by measured flow velocity and fluid viscosity. The concomitant changes in EC [Ca2+]i and nitric oxide (NO) were measured with fluorescent markers, and EC barrier function was assessed by fluorescent microsphere accumulation at EC junctions using confocal imaging. EC eNOS activation was evaluated by immunostaining. In response to changes in SS, increases in EC [Ca2+]i and gap formation occurred only in blood or RBC solution perfused vessels, whereas SS-dependent NO production and eNOS-Ser1177 phosphorylation occurred in both plasma and blood perfused vessels. A bioluminescent assay detected SS-dependent ATP release from RBCs. Pharmacological inhibition and genetic modification of pannexin-1 channels on RBCs abolished SS-dependent ATP release and SS-induced increases in EC [Ca2+]i and gap formation. CONCLUSIONS: SS-induced EC NO production occurs in both cell free fluid and blood perfused vessels, whereas SS-induced increases in EC [Ca2+]i and EC gap formation require the presence of RBCs, attributing to SS-induced pannexin-1 channel dependent release of ATP from RBCs. Thus, changes in blood flow alter vascular EC function through both wall SS and SS exerted on RBCs, and RBC released ATP contributes to SS-induced changes in EC barrier function.


Subject(s)
Capillary Permeability , Endothelial Cells/metabolism , Erythrocytes/metabolism , Mechanotransduction, Cellular , Venules/metabolism , Adenosine Triphosphate/metabolism , Animals , Blood Flow Velocity , Blood Viscosity , Calcium/metabolism , Enzyme Activation , Female , Gap Junctions/metabolism , In Vitro Techniques , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Perfusion , Phosphorylation , Rats, Sprague-Dawley , Regional Blood Flow , Stress, Mechanical , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL