Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Cardiovasc Res ; 96(3): 433-43, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22893680

ABSTRACT

AIMS: The channel function of the cardiac ryanodine receptor (RyR2) is modulated by calmodulin (CaM). However, the involvement of CaM in aberrant Ca(2+) release in diseased hearts remains unclear. Here, we investigated the pathogenic role of defective CaM binding to the RyR2 in the channel dysfunction associated with heart failure. METHODS AND RESULTS: The involvement of CaM in aberrant Ca(2+) release was assessed in normal and pacing-induced failing canine hearts. The apparent affinity of CaM for RyR2 was considerably lower in failing sarcoplasmic reticulum (SR) compared with normal SR. Thus, the amount of CaM bound to RyR2 was markedly decreased in failing myocytes. Expression of the CaM isoform Gly-Ser-His-CaM (GSH-CaM), which has much higher binding affinity than wild-type CaM for RyR1, restored normal CaM binding to RyR2 in both SR and myocytes of failing hearts. The Ca(2+) spark frequency (SpF) was markedly higher and the SR Ca(2+) content was lower in failing myocytes compared with normal myocytes. The incorporation of GSH-CaM into the failing myocytes corrected the aberrant SpF and SR Ca(2+) content to normal levels. CONCLUSION: Reduced CaM binding to RyR2 seems to play a critical role in the pathogenesis of aberrant Ca(2+) release in failing hearts. Correction of the reduced CaM binding to RyR2 stabilizes the RyR2 channel function and thereby restores normal Ca(2+) handling and contractile function to failing hearts.


Subject(s)
Calmodulin/metabolism , Heart Failure/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiac Pacing, Artificial , Disease Models, Animal , Dogs , Enzyme Activation , Heart Failure/physiopathology , Sarcoplasmic Reticulum/metabolism , Time Factors
2.
Circulation ; 124(6): 682-94, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21768539

ABSTRACT

BACKGROUND: The molecular mechanism by which catecholaminergic polymorphic ventricular tachycardia is induced by single amino acid mutations within the cardiac ryanodine receptor (RyR2) remains elusive. In the present study, we investigated mutation-induced conformational defects of RyR2 using a knockin mouse model expressing the human catecholaminergic polymorphic ventricular tachycardia-associated RyR2 mutant (S2246L; serine to leucine mutation at the residue 2246). METHODS AND RESULTS: All knockin mice we examined produced ventricular tachycardia after exercise on a treadmill. cAMP-dependent increase in the frequency of Ca²âº sparks was more pronounced in saponin-permeabilized knockin cardiomyocytes than in wild-type cardiomyocytes. Site-directed fluorescent labeling and quartz microbalance assays of the specific binding of DP2246 (a peptide corresponding to the 2232 to 2266 region: the 2246 domain) showed that DP2246 binds with the K201-binding sequence of RyR2 (1741 to 2270). Introduction of S2246L mutation into the DP2246 increased the affinity of peptide binding. Fluorescence quench assays of interdomain interactions within RyR2 showed that tight interaction of the 2246 domain/K201-binding domain is coupled with domain unzipping of the N-terminal (1 to 600)/central (2000 to 2500) domain pair in an allosteric manner. Dantrolene corrected the mutation-caused domain unzipping of the domain switch and stopped the exercise-induced ventricular tachycardia. CONCLUSIONS: The catecholaminergic polymorphic ventricular tachycardia-linked mutation of RyR2, S2246L, causes an abnormally tight local subdomain-subdomain interaction within the central domain involving the mutation site, which induces defective interaction between the N-terminal and central domains. This results in an erroneous activation of Ca²âº channel in a diastolic state reflecting on the increased Ca²âº spark frequency, which then leads to lethal arrhythmia.


Subject(s)
Calcium/metabolism , Ion Channel Gating/genetics , Mutation, Missense , Point Mutation , Ryanodine Receptor Calcium Release Channel/genetics , Tachycardia, Ventricular/etiology , Allosteric Site , Amino Acid Sequence , Amino Acid Substitution , Animals , Dantrolene/therapeutic use , Diastole , Epinephrine/toxicity , Gene Knock-In Techniques , Humans , Ion Channel Gating/physiology , Mice , Mice, Transgenic , Models, Cardiovascular , Molecular Sequence Data , Muscle Relaxants, Central/therapeutic use , Myocytes, Cardiac/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology , Running , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/physiology , Tachycardia, Ventricular/chemically induced , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/physiopathology , Tachycardia, Ventricular/prevention & control
3.
Biochem J ; 438(2): 379-87, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21649588

ABSTRACT

We have shown previously that the inter-domain interaction between the two domains of RyR (ryanodine receptor), CaMBD [CaM (calmodulin)-binding domain] and CaMLD (CaM-like domain), activates the Ca(2+) channel, and this process is called activation-link formation [Gangopadhyay and Ikemoto (2008) Biochem. J. 411, 415-423]. Thus CaM that is bound to CaMBD is expected to interfere the activation-link formation, thereby stabilizing the closed state of the channel under normal conditions. In the present paper, we report that, upon stimulation of neonatal cardiomyocytes with the pro-hypertrophy agonist ET-1 (endothelin-1), CaM dissociates from the RyR, which induces a series of intracellular events: increased frequency of Ca(2+) transients, translocation of the signalling molecules CaM, CaMKII (CaM kinase II) and the transcription factor NFAT (nuclear factor of activated T-cells) to the nucleus. These events then lead to the development of hypertrophy. Importantly, an anti-CaMBD antibody that interferes with activation-link formation prevented all of these intracellular events triggered by ET-1 and prevented the development of hypertrophy. These results indicate that the aberrant formation of the activation link between CaMBD and CaMLD of RyR is a key step in the development of hypertrophy in cultured cardiomyocytes.


Subject(s)
Calmodulin/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Animals, Newborn , Antibodies/immunology , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Size/drug effects , Endothelin-1/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Myocytes, Cardiac/drug effects , NFATC Transcription Factors/metabolism , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley
4.
Biophys J ; 100(10): 2367-77, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21575570

ABSTRACT

In skeletal muscle, excitation-contraction coupling involves the activation of dihydropyridine receptors (DHPR) and type-1 ryanodine receptors (RyR1) to produce depolarization-dependent sarcoplasmic reticulum Ca²âº release via orthograde signaling. Another form of DHPR-RyR1 communication is retrograde signaling, in which RyRs modulate the gating of DHPR. DP4 (domain peptide 4), is a peptide corresponding to residues Leu²44²-Pro²477 of the central domain of the RyR1 that produces RyR1 channel destabilization. Here we explore the effects of DP4 on orthograde excitation-contraction coupling and retrograde RyR1-DHPR signaling in isolated murine muscle fibers. Intracellular dialysis of DP4 increased the peak amplitude of Ca²âº release during step depolarizations by 64% without affecting its voltage-dependence or kinetics, and also caused a similar increase in Ca²âº release during an action potential waveform. DP4 did not modify either the amplitude or the voltage-dependence of the intramembrane charge movement. However, DP4 augmented DHPR Ca²âº current density without affecting its voltage-dependence. Our results demonstrate that the conformational changes induced by DP4 regulate both orthograde E-C coupling and retrograde RyR1-DHPR signaling.


Subject(s)
Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Peptides/chemistry , Peptides/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction/drug effects , Action Potentials/drug effects , Aniline Compounds/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Dialysis , Fluorescence , In Vitro Techniques , Intracellular Space/drug effects , Intracellular Space/metabolism , Ion Channel Gating/drug effects , Mice , Nonlinear Dynamics , Patch-Clamp Techniques , Protein Conformation , Time Factors , Xanthenes/metabolism
5.
Biochem Biophys Res Commun ; 396(2): 515-21, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20433809

ABSTRACT

We have recently shown that stimulation of cultured neonatal cardiomyocytes with endothelin-1 (ET-1) first produces conformational disorder within the ryanodine receptor (RyR2) and diastolic Ca(2+) leak from the sarcoplasmic reticulum (SR), then develops hypertrophy (HT) in the cardiomyocytes (Hamada et al., 2009 [3]). The present paper addresses the following question. By what mechanism does crosstalk between defective operation of RyR2 and activation of the HT gene program occur? Here we show that the immuno-stain of calmodulin (CaM) is localized chiefly in the cytoplasmic area in the control cells; whereas, in the ET-1-treated/hypertrophied cells, major immuno-staining is localized in the nuclear region. In addition, fluorescently labeled CaM that has been introduced into the cardiomyocytes using the BioPORTER system moves from the cytoplasm to the nucleus with the development of HT. The immuno-confocal imaging of Ca(2+)/CaM-dependent protein kinase II (CaMKII) also shows cytoplasm-to-nucleus shift of the immuno-staining pattern in the hypertrophied cells. In an early phase of hypertrophic growth, the frequency of spontaneous Ca(2+) transients increases, which accompanies with cytoplasm-to-nucleus translocation of CaM. In a later phase of hypertrophic growth, further increase in the frequency of spontaneous Ca(2+) transients results in the appearance of trains of Ca(2+) spikes, which accompanies with nuclear translocation of CaMKII. The cardio-protective reagent dantrolene (the reagent that corrects the de-stabilized inter-domain interaction within the RyR2 to a normal mode) ameliorates aberrant intracellular Ca(2+) events and prevents nuclear translocation of both CaM and CaMKII, then prevents the development of HT. These results suggest that translocation of CaM and CaMKII from the cytoplasm to the nucleus serves as messengers to transmit the pathogenic signal elicited in the surface membrane and in the RyR2 to the nuclear transcriptional sites to activate HT program.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/metabolism , Cardiomegaly/metabolism , Cell Nucleus/metabolism , Myocytes, Cardiac/metabolism , Active Transport, Cell Nucleus , Animals , Cardiomegaly/chemically induced , Cells, Cultured , Cytoplasm/metabolism , Endothelin-1/pharmacology , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/metabolism
6.
Cardiovasc Res ; 87(4): 609-17, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20388639

ABSTRACT

AIMS: Calmodulin (CaM) is well known to modulate the channel function of the cardiac ryanodine receptor (RyR2). However, the possible role of CaM on the aberrant Ca(2+) release in diseased hearts remains unclear. In this study, we investigated the state of RyR2-bound CaM and channel dysfunctions in pacing-induced failing hearts. METHODS AND RESULTS: The characteristics of CaM binding to RyR2 and the role of CaM on the aberrant Ca(2+) release were assessed in normal and failing canine hearts. The affinity of CaM binding to RyR2 was lower in failing sarcoplasmic reticulum (SR) than in normal SR. Addition of FK506, which dissociates FKBP12.6 from RyR2, to normal SR reduced the CaM-binding affinity. Dantrolene restored a normal level of the CaM-binding affinity in either FK506-treated (normal) SR or failing SR, suggesting that the defective inter-domain interaction between the N-terminal domain and the central domain of RyR2 (the therapeutic target of dantrolene) is involved in the reduction of the CaM-binding affinity in failing hearts. In saponin-permeabilized cardiomyocytes, the frequency of spontaneous Ca(2+) sparks was much more increased in failing cardiomyocytes than in normal cardiomyocytes, whereas the addition of a high concentration of CaM attenuated the aberrant increase of Ca(2+) sparks. CONCLUSION: The defective inter-domain interaction between N-terminal and central domains within RyR2 reduces the binding affinity of CaM to RyR2, thereby causing the spontaneous Ca(2+) release events in failing hearts. Correction of the defective CaM binding may be a new strategy to protect against the aberrant Ca(2+) release in heart failure.


Subject(s)
Calcium Signaling , Calmodulin/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Calcium Signaling/drug effects , Cardiac Pacing, Artificial , Dantrolene/pharmacology , Disease Models, Animal , Dogs , Excitation Contraction Coupling , Heart Failure/etiology , Microscopy, Confocal , Peptide Fragments/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Structure, Tertiary , Sarcoplasmic Reticulum/drug effects , Tacrolimus/pharmacology , Tacrolimus Binding Proteins/metabolism
7.
J Cell Sci ; 123(Pt 10): 1775-84, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20427316

ABSTRACT

Naturally occurring mutations in the cardiac ryanodine receptor (RyR2) have been linked to certain types of cardiac arrhythmias and sudden death. Two mutation hotspots that lie in the N-terminal and central regions of RyR2 are predicted to interact with one another and to form an important channel regulator switch. To monitor the conformational dynamics involving these regions, we generated a fluorescence resonance energy transfer (FRET) pair. A yellow fluorescent protein (YFP) was inserted into RyR2 after residue Ser437 in the N-terminal region, and a cyan fluorescent protein (CFP) was inserted after residue Ser2367 in the central region, to form a dual YFP- and CFP-labeled RyR2 (RyR2(S437-YFP/S2367-CFP)). We transfected HEK293 cells with RyR2(S437-YFP/S2367-CFP) cDNAs, and then examined them by using confocal microscopy and by measuring the FRET signal in live cells. The FRET signals are influenced by modulators of RyR2, by domain peptides that mimic the effects of disease causing RyR2 mutations, and by various drugs. Importantly, FRET signals were also readily detected in cells co-transfected with single CFP (RyR2(S437-YFP)) and single YFP (RyR2(S2367-CFP)) labeled RyR2, indicating that the interaction between the N-terminal and central mutation regions is an inter-subunit interaction. Our studies demonstrate that FRET analyses of this CFP- and YFP-labeled RyR2 can be used not only for investigating the conformational dynamics associated with RyR2 channel gating, but potentially, also for identifying drugs that are capable of stabilizing the conformations of RyR2.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium Channels/metabolism , Myocardium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/genetics , Cell Line , Cloning, Molecular , Death, Sudden, Cardiac/etiology , Fluorescence Resonance Energy Transfer , Genetic Predisposition to Disease , Humans , Microscopy, Confocal , Models, Biological , Mutation/genetics , Polymorphism, Genetic , Protein Conformation , Protein Interaction Domains and Motifs/genetics , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/genetics , Transgenes/genetics
8.
Biochem Biophys Res Commun ; 394(3): 660-6, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20226167

ABSTRACT

Calmodulin (CaM), one of the accessory proteins of the cardiac ryanodine receptor (RyR2), is known to play a significant role in the channel regulation of the RyR2. However, the possible involvement of calmodulin in the pathogenic process of catecholaminergic polymorphic ventricular tachycardia (CPVT) has not been investigated. In this study, we investigated the state of RyR2-bound CaM and channel dysfunctions using a knock-in (KI) mouse model with CPVT-linked RyR2 mutation (R2474S). Without added effectors, the affinity of CaM binding to the RyR2 was indistinguishable between KI and WT hearts. In response to cAMP (1 micromol/L), the RyR2 phosphorylation at Ser2808 increased in both WT and KI hearts to the same extent. However, cAMP caused a significant decrease of the CaM-binding affinity in KI hearts, but the affinity was unchanged in WT. Dantrolene restored a normal level of CaM-binding affinity in the cAMP-treated KI hearts, suggesting that defective inter-domain interaction between the N-terminal domain and the central domain of the RyR2 (the target of therapeutic effect of dantrolene) is involved in the cAMP-induced reduction of the CaM-binding affinity. In saponin-permeabilized cardiomyocytes, the addition of cAMP increased the frequency of spontaneous Ca(2+) sparks to a significantly larger extent in KI cardiomyocytes than in WT cardiomyocytes, whereas the addition of a high concentration of CaM attenuated the aberrant increase of Ca(2+) sparks. In conclusion, CPVT mutation causes defective inter-domain interaction, significant reduction in the ability of CaM binding to the RyR2, spontaneous Ca(2+) leak, and then lethal arrhythmia.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Knock-In Techniques , Mice , Mice, Mutant Strains , Mutation , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Tachycardia, Ventricular/genetics
9.
Circ Res ; 106(8): 1413-24, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20224043

ABSTRACT

RATIONALE: Catecholaminergic polymorphic ventricular tachycardia (CPVT) is caused by a single point mutation in a well-defined region of the cardiac type 2 ryanodine receptor (RyR)2. However, the underlying mechanism by which a single mutation in such a large molecule produces drastic effects on channel function remains unresolved. OBJECTIVE: Using a knock-in (KI) mouse model with a human CPVT-associated RyR2 mutation (R2474S), we investigated the molecular mechanism by which CPVT is induced by a single point mutation within the RyR2. METHODS AND RESULTS: The R2474S/+ KI mice showed no apparent structural or histological abnormalities in the heart, but they showed clear indications of other abnormalities. Bidirectional or polymorphic ventricular tachycardia was induced after exercise on a treadmill. The interaction between the N-terminal (amino acids 1 to 600) and central (amino acids 2000 to 2500) domains of the RyR2 (an intrinsic mechanism to close Ca(2+) channels) was weakened (domain unzipping). On protein kinase A-mediated phosphorylation of the RyR2, this domain unzipping further increased, resulting in a significant increase in the frequency of spontaneous Ca(2+) transients. cAMP-induced aberrant Ca(2+) release events (Ca(2+) sparks/waves) occurred at much lower sarcoplasmic reticulum Ca(2+) content as compared to the wild type. Addition of a domain-unzipping peptide, DPc10 (amino acids 2460 to 2495), to the wild type reproduced the aforementioned abnormalities that are characteristic of the R2474S/+ KI mice. Addition of DPc10 to the (cAMP-treated) KI cardiomyocytes produced no further effect. CONCLUSIONS: A single point mutation within the RyR2 sensitizes the channel to agonists and reduces the threshold of luminal [Ca(2+)] for activation, primarily mediated by defective interdomain interaction within the RyR2.


Subject(s)
Calcium Signaling , Myocytes, Cardiac/metabolism , Point Mutation , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/metabolism , Animals , Caffeine , Calcium Signaling/genetics , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Epinephrine , Excitation Contraction Coupling , Genotype , Isoproterenol , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Contraction , Peptide Fragments/metabolism , Phenotype , Phosphorylation , Physical Exertion , Protein Conformation , Protein Structure, Tertiary , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/metabolism , Structure-Activity Relationship , Tachycardia, Ventricular/chemically induced , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/physiopathology , Time Factors
10.
Neurobiol Aging ; 31(12): 2080-90, 2010 Dec.
Article in English | MEDLINE | ID: mdl-19108934

ABSTRACT

Inclusion body myositis (IBM), the most common muscle disorder in the elderly, is partly characterized by dysregulation of ß-amyloid precursor protein (ßAPP) expression and abnormal, intracellular accumulation of full-length ßAPP and ß-amyloid epitopes. The present study examined the effects of ß-amyloid accumulation on force generation and Ca(2+) release in skeletal muscle from transgenic mice harboring human ßAPP and assessed the consequence of Aß(1-42) modulation of the ryanodine receptor Ca(2+) release channels (RyRs). ß-Amyloid laden muscle produced less peak force and exhibited Ca(2+) transients with smaller amplitude. To determine whether modification of RyRs by ß-amyloid underlie the effects observed in muscle, in vitro Ca(2+) release assays and RyR reconstituted in planar lipid bilayer experiments were conducted in the presence of Aß(1-42). Application of Aß(1-42) to RyRs in bilayers resulted in an increased channel open probability and changes in gating kinetics, while addition of Aß(1-42) to the rabbit SR vesicles resulted in RyR-mediated Ca(2+) release. These data may relate altered ßAPP metabolism in IBM to reductions in RyR-mediated Ca(2+) release and muscle contractility.


Subject(s)
Amyloid beta-Peptides/toxicity , Calcium/antagonists & inhibitors , Calcium/metabolism , Muscle Contraction/drug effects , Muscle, Skeletal/metabolism , Myositis, Inclusion Body/metabolism , Peptide Fragments/toxicity , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/physiology , Animals , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cations, Divalent/antagonists & inhibitors , Cations, Divalent/metabolism , Mice , Mice, Transgenic , Muscle Contraction/genetics , Muscle, Skeletal/physiopathology , Myositis, Inclusion Body/physiopathology , Peptide Fragments/genetics , Peptide Fragments/physiology , Rabbits
11.
J Am Coll Cardiol ; 53(21): 1993-2005, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19460614

ABSTRACT

OBJECTIVES: We sought to investigate the effect of dantrolene, a drug generally used to treat malignant hyperthermia, on the Ca2+ release and cardiomyocyte function in failing hearts. BACKGROUND: The N-terminal (N: 1-600) and central (C: 2000-2500) domains of the ryanodine receptor (RyR) harbor many mutations associated with malignant hyperthermia in skeletal muscle RyR (RyR1) and polymorphic ventricular tachycardia in cardiac RyR (RyR2). There is strong evidence that interdomain interaction between these regions plays an important role in the mechanism of channel regulation. METHODS: Sarcoplasmic reticulum vesicles and cardiomyocytes were isolated from the left ventricular muscles of dogs (normal or rapid ventricular pacing for 4 weeks), for Ca2+ leak, transient, and spark assays. To assess the zipped or unzipped state of the interacting domains, the RyR was labeled fluorescently with methylcoumarin acetate in a site-directed manner. We used a quartz-crystal microbalance technique to identify the dantrolene binding site within the RyR2. RESULTS: Dantrolene specifically bound to domain 601-620 in RyR2. In the sarcoplasmic reticulum isolated from pacing-induced failing dog hearts, the defective interdomain interaction (domain unzipping) had already occurred, causing spontaneous Ca2+ leak. Dantrolene suppressed both domain unzipping and the Ca2+ leak, demonstrating identical drug concentration-dependence (IC50 = 0.3 micromol/l). In failing cardiomyocytes, both diastolic Ca2+ sparks and delayed afterdepolarization were observed frequently, but 1 micromol/l dantrolene inhibited both events. CONCLUSIONS: Dantrolene corrects defective interdomain interactions within RyR2 in failing hearts, inhibits spontaneous Ca2+ leak, and in turn improves cardiomyocyte function in failing hearts. Thus, dantrolene may have a potential to treat heart failure, specifically targeting the RyR2.


Subject(s)
Dantrolene/therapeutic use , Heart Failure/drug therapy , Malignant Hyperthermia/drug therapy , Muscle Relaxants, Central/therapeutic use , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Ventricular Function, Right/drug effects , Animals , Calcium/metabolism , Disease Models, Animal , Dogs , Heart Failure/etiology , Heart Failure/physiopathology , Malignant Hyperthermia/complications , Malignant Hyperthermia/physiopathology , Membrane Potentials/drug effects , Microscopy, Confocal , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Ryanodine Receptor Calcium Release Channel/drug effects , Treatment Outcome , Ventricular Function, Right/physiology
12.
Biochem Biophys Res Commun ; 380(3): 493-7, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19284993

ABSTRACT

Recent studies on cardiac hypertrophy animal model suggest that inter-domain interactions within the ryanodine receptor (RyR2) become defective concomitant with the development of hypertrophy (e.g. de-stabilization of the interaction between N-terminal and central domains of RyR2; T. Oda, M. Yano, T. Yamamoto, T. Tokuhisa, S. Okuda, M. Doi, T. Ohkusa, Y. Ikeda, S. Kobayashi, N. Ikemoto, M. Matsuzaki, Defective regulation of inter-domain interactions within the ryanodine receptor plays a key role in the pathogenesis of heart failure, Circulation 111 (2005) 3400-3410). To determine if de-stabilization of the inter-domain interaction in fact causes hypertrophy, we introduced DPc10 (a peptide corresponding to the G(2460)-P(2495) region of RyR2, which is known to de-stabilize the N-terminal/central domain interaction) into rat neonatal cardiomyocytes by mediation of peptide carrier BioPORTER. After incubation for 24h the peptide induced hypertrophy, as evidenced by significant increase in cell size and [(3)H]leucine uptake. K201 or dantrolene, the reagents known to correct the de-stabilized inter-domain interaction to a normal mode, prevented the DPc10-induced hypertrophy. These results suggest that disruption of the normal N-terminal/central inter-domain interaction within the RyR2 is a causative mechanism of cardiomyocyte hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/pathology , Cells, Cultured , Endothelin-1/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Peptide Fragments/pharmacology , Rats , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/pharmacology
13.
Cell Calcium ; 45(3): 264-74, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19131108

ABSTRACT

Alternative splicing of ASI residues (Ala(3481)-Gln(3485)) in the skeletal muscle ryanodine receptor (RyR1) is developmentally regulated: the residues are present in adult ASI(+)RyR1, but absent in the juvenile ASI(-)RyR1 which is over-expressed in adult myotonic dystrophy type 1 (DM1). Although this splicing switch may influence RyR1 function in developing muscle and DM1, little is known about the properties of the splice variants. We examined excitation-contraction (EC) coupling and the structure and interactions of the ASI domain (Thr(3471)-Gly(3500)) in the splice variants. Depolarisation-dependent Ca(2+) release was enhanced by >50% in myotubes expressing ASI(-)RyR1 compared with ASI(+)RyR1, although DHPR L-type currents and SR Ca(2+) content were unaltered, while ASI(-)RyR1 channel function was actually depressed. The effect on EC coupling did not depend on changes in ASI domain secondary structure. Probing RyR1 function with peptides possessing the ASI domain sequence indicated that the domain contributes to an inhibitory module in RyR1. The action of the peptide depended on a sequence of basic residues and their alignment in an alpha-helix adjacent to the ASI splice site. This is the first evidence that the ASI residues contribute to an inhibitory module in RyR1 that influences EC coupling. Implications for development and DM1 are discussed.


Subject(s)
Alternative Splicing/genetics , Bone and Bones/metabolism , Muscle Contraction , Ryanodine Receptor Calcium Release Channel/genetics , Amino Acid Sequence , Amino Acids, Basic , Animals , Calcium/metabolism , Ion Channel Gating , Kinetics , Magnetic Resonance Spectroscopy , Mice , Molecular Sequence Data , Muscle Fibers, Skeletal/metabolism , Peptides/chemistry , Sarcoplasmic Reticulum/metabolism , Sequence Analysis, Protein
14.
Cardiovasc Res ; 81(3): 536-45, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18996969

ABSTRACT

AIMS: A domain peptide (DP) matching the Gly(2460)-Pro(2495) region of the cardiac type-2 ryanodine receptor (RyR2), DPc10, is known to mimic channel dysfunction associated with catecholaminergic polymorphic ventricular tachycardia (CPVT), owing to its interference in a normal interaction of the N-terminal (1-600) and central (2000-2500) domains (viz. domain unzipping). Using DPc10 and two other DPs harboring different mutation sites, we investigated the underlying mechanism of abnormal Ca(2+) cycling in failing hearts. METHODS AND RESULTS: Sarcoplasmic reticulum (SR) vesicles and cardiomyocytes were isolated from dog left ventricular muscles for Ca(2+) leak and spark assays. The RyR2 moiety of the SR was fluorescently labelled with methylcoumarin acetate (MCA) using DPs corresponding to the 163-195 and 4090-4123 regions of RyR2 (DP163-195 and DP4090-4123, respectively) as site-directed carriers. Both DPs mediated a specific MCA fluorescence labelling of RyR2. Addition of either DP to the MCA-labelled SR induced domain unzipping, as evidenced by an increased accessibility of the bound MCA to a large-size fluorescence quencher. Both SR Ca(2+) leak and Ca(2+) spark frequency (SpF) were markedly increased in failing cardiomyocytes. Upon introduction of DP163-195 or DP4090-4123 into normal SR or cardiomyocytes, both Ca(2+) leak and SpF increased to the levels comparable with those of failing myocytes. K201 (JTV519) suppressed all of the effects induced by DP163-195 (domain unzipping and increased Ca(2+) leak and SpF) or those in failing cardiomyocytes, but did not suppress the effects induced by DP4090-4123. CONCLUSION: Defective inter-domain interaction between N-terminal and central domains induces diastolic Ca(2+) leak, leading to heart failure and lethal arrhythmia. Mutation at the C-terminal region seen in CPVT does not seem to communicate with the aforementioned N-terminal and central inter-domain interaction, although spontaneous Ca(2+) leak is similarly induced.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium Signaling , Calcium/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Arrhythmias, Cardiac/etiology , Binding Sites , Cardiac Pacing, Artificial/adverse effects , Disease Models, Animal , Dogs , Fluorescent Dyes , Heart Failure/etiology , Heart Ventricles/metabolism , Microscopy, Confocal , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Ryanodine Receptor Calcium Release Channel/genetics , Spectrometry, Fluorescence , Tacrolimus Binding Proteins/metabolism , Time Factors
15.
Biochem J ; 411(2): 415-23, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18171325

ABSTRACT

In the present study we show that the interaction of the CaM (calmodulin)-binding domain (Lys(3614)-Asn(3643)) with the Cys(4114)-Asn(4142) region (a region included in the CaM-like domain) serves as an intrinsic regulator of the RyR1 (type-1 ryanodine receptor). We tested the effects of antibodies raised against the two putative key regions of RyR1 [anti-(Lys(3614)-Asn(3643)) and anti-(Cys(4114)-Asn(4142)) antibodies]. Both antibodies produced significant inhibition of [3H]ryanodine-binding activity of RyR1. This suggests that the inter-domain interaction between the two domains, Lys(3614)-Asn(3643) and Cys(4114)-Asn(4142), activates the channel, and that the binding of antibody to either side of the interacting domain pair interfered with the formation of a 'channel-activation link' between the two regions. In order to spectroscopically monitor the mode of interaction of these domains, the site of inter-domain interaction was fluorescently labelled with MCA [(7-methoxycoumarin-4-yl)acetyl] in a site-directed manner. The accessibility of the bound MCA to a large molecular mass fluorescence quencher, BSA-QSY (namely, the size of a gap between the interacting domains) decreased with an increase of [Ca2+] in a range of 0.03-2.0 microM, as determined by Stern-Volmer fluorescence quenching analysis. The Ca2+-dependent decrease in the quencher accessibility was more pronounced in the presence of 150 microM 4-CmC (4-chlorometacresol), and was reversed by 1 mM Mg2+ (a well-known inhibitor of Ca2+/agonist-induced channel activation). These results suggest that the Lys(3614)-Asn(3643) and Cys(4114)-Asn(4142) regions of RyR1 interact with each other in a Ca2+- and agonist-dependent manner, and this serves as a mechanism of Ca2+- and agonist-dependent activation of the RyR1 Ca2+ channel.


Subject(s)
Calcium Channels/metabolism , Calcium/agonists , Calcium/metabolism , Calmodulin/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Asparagine/genetics , Asparagine/metabolism , Cysteine/genetics , Cysteine/metabolism , Lysine/genetics , Lysine/metabolism , Molecular Weight , Protein Binding , Rabbits , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism
16.
Circulation ; 117(6): 762-72, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18227387

ABSTRACT

BACKGROUND: We previously demonstrated that defective interdomain interaction between N-terminal (0 to 600) and central regions (2000 to 2500) of ryanodine receptor 2 (RyR2) induces Ca2+ leak in failing hearts and that K201 (JTV519) inhibits the Ca2+ leak by correcting the defective interdomain interaction. In the present report, we identified the K201-binding domain and characterized the role of this novel domain in the regulation of the RyR2 channel. METHODS AND RESULTS: An assay using a quartz-crystal microbalance technique (a very sensitive mass-measuring technique) revealed that K201 specifically bound to recombinant RyR2 fragments 1741 to 2270 and 1981 to 2520 but not to other RyR2 fragments from the 1 to 2750 region (1 to 610, 494 to 1000, 741 to 1260, 985 to 1503, 1245 to 1768, 2234 to 2750). By further analysis of the fragment(1741-2270), K201 was found to specifically bind to its subfragment(2114-2149). With the use of the peptide matching this subfragment (DP(2114-2149)) as a carrier, the RyR2 was fluorescently labeled with methylcoumarin acetate (MCA) in a site-directed manner. After tryptic digestion, the major MCA-labeled fragment of RyR2 (155 kDa) was detected by an antibody raised against the central region (Ab(2132)). Moreover, of several recombinant RyR2 fragments, only fragment(2234-2750) was specifically MCA labeled; this suggests that the K201-binding domain(2114-2149) binds with domain(2234-2750). Addition of DP(2114-2149) to the MCA-labeled sarcoplasmic reticulum interfered with the interaction between domain(2114-2149) and domain(2234-2750), causing domain unzipping, as evidenced by an increased accessibility of the bound MCA to a large-size fluorescence quencher. In failing cardiomyocytes, the frequency of spontaneous Ca2+ spark was markedly increased compared with normal cardiomyocytes, whereas incorporation of DP(2114-2149) markedly decreased the frequency of spontaneous Ca2+ spark. CONCLUSIONS: We first identified the K201-binding site as domain(2114-2149) of RyR2. Interruption of the interdomain interaction between the domain(2114-2149) and central domain(2234-2750) seems to mediate stabilization of RyR2 in failing hearts, which may lead to a novel therapeutic strategy against heart failure and perhaps lethal arrhythmia.


Subject(s)
Calcium/metabolism , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Sequence , Animals , Annexin A5/chemistry , Annexin A5/metabolism , Binding Sites , Disease Models, Animal , Dogs , Heart Failure/metabolism , Linear Models , Molecular Sequence Data , Ryanodine Receptor Calcium Release Channel/chemistry , Sarcoplasmic Reticulum , Sequence Homology, Amino Acid , Thiazepines/metabolism
17.
Eur Biophys J ; 37(4): 455-67, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18038129

ABSTRACT

The clustering of cardiac RyR mutations, linked to sudden cardiac death (SCD), into several regions in the amino acid sequence underlies the hypothesis that these mutations interfere with stabilising interactions between different domains of the RyR2. SCD mutations cause increased channel sensitivity to cytoplasmic and luminal Ca(2+). A synthetic peptide corresponding to part of the central domain (DPc10:(2460)G-P(2495)) was designed to destabilise the interaction of the N-terminal and central domains of wild-type RyR2 and mimic the effects of SCD mutations. With Ca(2+) as the sole regulating ion, DPc10 caused increased channel activity which could be reversed by removal of the peptide whereas in the presence of ATP DPc10 caused no activation. In support of the domain destablising hypothesis, the corresponding peptide (DPc10-mut) containing the CPVT mutation R2474S did not affect channel activity under any circumstances. DPc10-induced activation was due to a small increase in RyR2 sensitivity to cytoplasmic Ca(2+) and a large increase in the magnitude of luminal Ca(2+) activation. The increase in the luminal Ca(2+) response appeared reliant on the luminal-to-cytoplasmic Ca(2+) flux in the channel, indicating that luminal Ca(2+) was activating the RyR2 via its cytoplasmic Ca(2+) sites. DPc10 had no significant effect on the RyR2 gating associated with luminal Ca(2+) sensing sites. The results were fitted by the luminal-triggered Ca(2+) feed-through model and the effects of DPc10 were explained entirely by perturbations in cytoplasmic Ca(2+)-activation mechanism.


Subject(s)
Calcium/chemistry , Calcium/metabolism , Cytoplasm/metabolism , Lipid Bilayers/chemistry , Peptides/chemistry , Ryanodine Receptor Calcium Release Channel/chemistry , Animals , Dose-Response Relationship, Drug , Ions , Models, Biological , Models, Chemical , Mutation , Myocardium/metabolism , Protein Structure, Tertiary , Sarcoplasmic Reticulum/metabolism , Sheep
18.
J Membr Biol ; 220(1-3): 11-20, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17879109

ABSTRACT

In this study, we present evidence for the mechanism of neomycin inhibition of skeletal ryanodine receptors (RyRs). In single-channel recordings, neomycin produced monophasic inhibition of RyR open probability and biphasic inhibition of [(3)H]ryanodine binding. The half-maximal inhibitory concentration (IC(50)) for channel blockade by neomycin was dependent on membrane potential and cytoplasmic [Ca(2+)], suggesting that neomycin acts both as a pore plug and as a competitive antagonist at a cytoplasmic Ca(2+) binding site that causes allosteric inhibition. This novel Ca(2+)/neomycin binding site had a neomycin affinity of 100 nM: and a Ca(2+) affinity of 35 nM,: which is 30-fold higher than that of the well-described cytoplasmic Ca(2+) activation site. Therefore, a new high-affinity class of Ca(2+) binding site(s) on the RyR exists that mediates neomycin inhibition. Neomycin plugging of the channel pore induced brief (1-2 ms) conductance substates at 30% of the fully open conductance, whereas allosteric inhibition caused complete channel closure with durations that depended on the neomycin concentration. We quantitatively account for these results using a dual inhibition model for neomycin that incorporates voltage-dependent pore plugging and Ca(2+)-dependent allosteric inhibition.


Subject(s)
Calcium/metabolism , Ion Channel Gating/drug effects , Neomycin/pharmacology , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Binding Sites , Calcium/pharmacology , Dose-Response Relationship, Drug , Ion Channel Gating/physiology , Lipid Bilayers/metabolism , Markov Chains , Muscle, Skeletal/metabolism , Protein Binding/drug effects , Rabbits , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
19.
J Am Coll Cardiol ; 49(16): 1722-32, 2007 Apr 24.
Article in English | MEDLINE | ID: mdl-17448375

ABSTRACT

OBJECTIVES: We investigated whether defective intracellular Ca2+ handling is corrected by carvedilol in heart failure. BACKGROUND: In heart failure, the interaction between the N-terminal and central domains of the ryanodine receptor (RyR), the domains where many mutations have been found in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT), is defective, as shown in our recent report. METHODS: Sarcoplasmic reticulum vesicles were isolated from canine left ventricular muscle (normal or 4-weeks rapid ventricular pacing). The RyR was labeled with the fluorescent conformational probe methylcoumarin acetate (MCA) with DPc10 (a synthetic peptide corresponding to Gly2460-Pro2495 of RyR, one of the mutable domains in CPVT) as a site-direction carrier. RESULTS: Normal cardiac function was well preserved in carvedilol-treated/paced dogs (CV+) but not in the untreated/paced dogs (CV-). In CV-, the interdomain interaction within RyR was defective (i.e., in an unzipped state), as determined by the fluorescence quenching technique. However, in CV+, the domain interaction remained normal (i.e., in a zipped state). In CV-, oxidative stress of RyR (reduction in the number of free thiols) was severe, but it was negligible in CV+. In (CV-) failing cardiomyocytes, incubation with low-dose CV (30 nmol/l), which eliminated intracellular reactive oxygen species with no acute effect on cell shortening, markedly improved the contractile function and Ca2+ transient. However, after domain unzipping by DPc10, CV was without effect. CONCLUSIONS: Carvedilol, at a concentration that is sufficient to produce antioxidant effect, improves the intracellular Ca2+ handling and contractile dysfunction by correcting defective interdomain interaction within the RyR in the failing heart.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Calcium/metabolism , Carbazoles/pharmacology , Cardiac Output, Low/physiopathology , Free Radicals/metabolism , Propanolamines/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Animals , Cardiac Output, Low/drug therapy , Carvedilol , Dogs , In Vitro Techniques , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/physiology
20.
Biochemistry ; 46(14): 4272-9, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17361990

ABSTRACT

Ryanodine receptor (RyR) mutations linked with some congenital skeletal and cardiac diseases are localized to three easily definable regions: region 1 (N-terminal domain), region 2 (central domain), and a rather broad region 3 containing the channel pore. As shown in our recent studies, the interdomain interaction between regions 1 and 2 plays a critical role in channel regulation and pathogenesis. Here we present evidence that within region 3 there is a similar channel regulation mechanism mediated by an interdomain interaction. DP15, a peptide corresponding to RyR1 residues 4820-4841, produced significant activation of [3H]ryanodine binding above threshold Ca2+ concentrations (>or=0.3 microM), but MH mutations (L4823P or L4837V) made in DP15 almost completely abolished its channel activating function. To identify the DP15 binding site(s) within RyR1, DP15 (labeled with a fluorescent probe Alexa Fluor 680 and a photoaffinity cross-linker APG) was cross-linked to RyR1, and the site of cross-linking was identified by gel analysis of fluorescently labeled proteolytic fragments with the aid of Western blotting with site-specific antibodies. The shortest fluorescently labeled band was a 96 kDa fragment which was stained with an antibody directed to the region of residues 4114-4142 of RyR1, indicating that the interaction between the region of residues 4820-4841 adjacent to the channel pore and the 96 kDa segment containing the region of residues 4114-4142 is involved in the mechanism of Ca2+-dependent channel regulation. In further support of this concept, anti-DP15 antibody and cardiac counterpart of DP15 produced channel activation similar to that of DP15.


Subject(s)
Calcium Channels/chemistry , Peptide Fragments/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/metabolism , Antibodies/pharmacology , Binding Sites , Blotting, Western , Calcium/metabolism , Calcium/pharmacology , Cross-Linking Reagents/metabolism , Cytoplasm/chemistry , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Models, Biological , Molecular Sequence Data , Molecular Weight , Muscle, Skeletal/chemistry , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/immunology , Peptide Fragments/physiology , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Rabbits , Ryanodine Receptor Calcium Release Channel/analysis , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum
SELECTION OF CITATIONS
SEARCH DETAIL
...