Next Article in Journal
Inhibition of KIF23 Alleviates IPAH by Targeting Pyroptosis and Proliferation of PASMCs
Next Article in Special Issue
New Panx-1 Blockers: Synthesis, Biological Evaluation and Molecular Dynamic Studies
Previous Article in Journal
Transcription Factors in Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Approaches of Ryanodine Receptor-Associated Heart Diseases

by
Norbert Szentandrássy
1,2,
Zsuzsanna É. Magyar
1,
Judit Hevesi
3,
Tamás Bányász
1,
Péter P. Nánási
1,4 and
János Almássy
1,5,*
1
Department of Physiology, Faculty of Medicine, University of Debrecen, 98 Nagyerdei krt, 4032 Debrecen, Hungary
2
Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 98 Nagyerdei krt, 4032 Debrecen, Hungary
3
Department of Orthodontics, Faculty of Dentistry, University of Debrecen, 98 Nagyerdei krt, 4032 Debrecen, Hungary
4
Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, 98 Nagyerdei krt, 4032 Debrecen, Hungary
5
Department of Physiology, Semmelweis University, P.O. Box 2, 1428 Budapest, Hungary
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(8), 4435; https://doi.org/10.3390/ijms23084435
Submission received: 13 February 2022 / Revised: 11 April 2022 / Accepted: 14 April 2022 / Published: 18 April 2022

Abstract

:
Cardiac diseases are the leading causes of death, with a growing number of cases worldwide, posing a challenge for both healthcare and research. Therefore, the most relevant aim of cardiac research is to unravel the molecular pathomechanisms and identify new therapeutic targets. Cardiac ryanodine receptor (RyR2), the Ca2+ release channel of the sarcoplasmic reticulum, is believed to be a good therapeutic target in a group of certain heart diseases, collectively called cardiac ryanopathies. Ryanopathies are associated with the impaired function of the RyR, leading to heart diseases such as congestive heart failure (CHF), catecholaminergic polymorphic ventricular tachycardia (CPVT), arrhythmogenic right ventricular dysplasia type 2 (ARVD2), and calcium release deficiency syndrome (CRDS). The aim of the current review is to provide a short insight into the pathological mechanisms of ryanopathies and discuss the pharmacological approaches targeting RyR2.

1. Introduction

Ca2+ Release in the Heart in Health and Disease

RyR2 is a ligand-gated Ca2+ release channel of the sarcoplasmic reticulum (SR), activated by Ca2+ carried by the L-type Ca2+ channel (LTCC) during systole. This mechanism is called Ca2+-induced Ca2+ release (CICR), which is the crucial step of cardiac excitation–contraction coupling (ECC). The Ca2+ current serves as an essential trigger for Ca2+ release, but most of the Ca2+ required for contraction comes from the SR. In the beginning of diastole, Ca2+ release is terminated and RyR2s remain closed during the whole course of diastole. [Ca2+]i is restored by the sarcoplasmic reticulum Ca2+ ATP-ase (SERCA), the plasma membrane Ca2+ pump (PMCA), and the Na+-Ca2+ exchanger (NCX) [1].
Cardiac contractility depends on the peak systolic [Ca2+]i; therefore, it is determined by the SR Ca2+ content, the magnitude of the LTCC current, and the sensitivity of RyR2 to Ca2+ (ECC gain). Ca2+ fluxes through the sarcolemma, and SR are linked through a dynamic balance, which protects the cardiomyocyte against cytoplasmic Ca2+ overload while maintaining Ca2+ release amplitudes. The basis of this autoregulation is that (1) RyR2 is activated by increasing SR [Ca2+], (2) LTCC inactivation is Ca2+-dependent, and (3) NCX-mediated Ca2+ clearance rate is enhanced by elevated [Ca2+]i [2,3]. The function of this mechanism was demonstrated using the RyR2 agonist caffeine [4]. The authors showed that the caffeine-induced increase of Ca2+ release amplitude was only transient, as the Ca2+ release amplitude returned to control values during treatment. In the new steady state, Ca2+ release operated at lower SR Ca2+ content but higher fractional Ca2+ release. The underlaying mechanism of this autoregulation was explained by the NCX activity: higher [Ca2+]i transients drive larger Ca2+ efflux through the NCX, which eventually decreased the SR Ca2+ content to a lower steady state level. This experimental setup was designed to simulate the situation in certain heart diseases such as congestive heart failure (CHF) or catecholaminergic polymorphic ventricular tachycardia (CPVT) when RyR2 is substantially active during diastole; however, the results suggested that the autoregulation of Ca2+ release is broken in these conditions. Ca2+ leaked out of the SR and the resulting Ca2+ overload, which induced an inward current through the NCX, causing two problems: (1) it generated premature depolarization (delayed afterdepolarizations, DAD), associated with triggered arrythmias, and (2) depleted the SR Ca2+ content to severely low levels (below the capacity of autoregulation) that depressed Ca2+ release amplitude [5,6]. Apparently, these two points are logically inconsistent, because although low SR Ca2+ level explains the diminished Ca2+ transients and contractility in CHF, it should be associated with low arrhythmogenic propensity (since SR depletion negatively feeds back to diastolic Ca2+ leak). However, in contrast, Ca2+ leak remains paradoxically sustained in CHF, which is linked to high arrhythmogenic activity. This discrepancy might be resolved by the impaired function of Ca2+ handling proteins, including the RyR2 [4,7]. Next, we discuss the functional alterations of RyR that may account for the broken function of Ca2+ release autoregulation in heart diseases.

2. RyR2 Associated Heart Diseases

2.1. Congestive Heart Failure (CHF)

The primary causes of CHF include myocardial infarction and chronic hypertension, which initiate the morphological and functional remodeling of the cardiac tissue, resulting in cardiac hypertrophy. Cardiac remodeling is an adaptive response to increased wall stress and altered neurohormonal stimuli after myocardial infarction or hypertension, which compensates for the impaired pump function. However, in the long term, ongoing maladaptive remodeling gradually demolishes the structural integrity of the tissue, leading to decompensation and heart failure with reduced ejection fraction (HFrEF) [8,9,10,11,12]. This condition is associated with electrical instability, ventricular tachyarrhythmias, and sudden cardiac death. Although death can be prevented in many patients using implanted cardioverter-defibrillators (ICD), a large proportion of patients produce ICD-irresponsive arrhythmias. In these cases, the failure of ICDs to rescue the patient indicate that the primary cause of sudden death was acute mechanical failure, and tachyarrhythmias (if develop) are only a result of a secondary, ischemic process [13].
So far, several signaling pathways have been identified to induce hypertrophy, including the calcineurin–NFAT pathway, which is stimulated by pathological intracellular Ca2+ concentrations ([Ca2+]i), generated by enhanced Ca2+ release [14,15,16]. A critical point of this pathological signal is the impaired function of Ca2+ release channels, including the ryanodine receptor (RyR2-cardiac isoform), which was shown to be essential in developing hypertrophy [17,18,19,20,21,22,23,24,25]. In addition to its role in hypertrophic signaling, RyR2 is also responsible for triggered arrythmias [26,27].

Impaired Function of RyR2 in CHF

There is a general agreement that RyR2 is hyperactive (i.e., leaky) in CHF, and it contributes (along with reduced SERCA and enhanced NCX function) significantly to cardiac dysfunction [7,28,29]. Although the mechanism by which RyR2 becomes leaky is controversial, there is an agreement that the hyperadrenergic state in HF is an important factor [30]. The first proposal for the pathomechanism was made by Marks’ group, who showed that CHF is associated with enhanced PKA activity and increased phosphorylation state of RyR2 at the amino acid S2808 [31]. This modification would cause the dissociation of an accessory protein called FKBP12.6 (calstabin2). As FKBP12.6 stabilizes the closed state of the channel by preventing subconductive open states, RyR2 devoid of FKBP12.6 spontaneously opens during diastole, resulting in SR Ca2+ depletion and systolic dysfunction [31,32,33]. In line with these data, a non-phosphorylatable mutation (S2808A) prevented the progression of CHF in experimental post-myocardial infarction (MI) in mice [34]. However, other research groups failed to show high phosphorylation rates of S2808 and lower FKBP12.6 association with RyR2 and debating the role of S2808 in the regulation of RyR2 [35,36,37,38,39,40]. Moreover, Valdivia and Houser’s group showed that the S2808A mutation had no effect on the post-MI progression of CHF [41]. Furthermore, S2808A mice displayed unaltered β-adrenergic response and myocyte function, and they were not significantly protected against maladaptive cardiac remodeling [42,43]. Other studies even question the direct role of RyR2 in the regulation of cardiac contractility by PKA. For example, Bers’ group showed that PKA activation dramatically enhanced elementary Ca2+ release and Ca2+ content of the SR in wild type, but not in phospholamban-knockout cardiomyocytes, indicating that the effects of PKA on Ca2+ release were completely due to the phosphorylation of phospholamban, which resulted in enhanced SR Ca2+ load and RyR2 gating [44]. This mechanism, when luminal [Ca2+] reaches a threshold and triggers Ca2+ spillover by opening RyR2s, is called store overload-induced Ca2+ release (SOICR), manifested as propagating Ca2+ waves [45,46].
Nevertheless, PKA is not the only effector of the β-adrenergic pathway. There is solid evidence that the Ca2+–calmodulin-dependent protein kinase II (CaMKII) is also activated during β-adrenergic stimulation [47,48]. RyR was shown to be a CaMKII target, as CaMKII increased Ca2+ release in both wild-type and phospholamban-knockout myocytes [49,50]. The role of CaMKII phosphorylation on RyR2 function and RyR2-related cardiac pathology was mainly investigated using S2814A RyR2 transgenic mice in which one of the CaMKII phosphorylation sites was ablated [51]. The RyR2-S2814A mice were less prone to produce premature beats than wild-type animals [52]. Increased CaMKII activity was shown to promote HF progression by the phosphorylation of RyR2 and increased SR Ca2+ leak [53,54,55]. This is also supported by the result that CaMKII overexpression reduced SR Ca2+ load and enhanced SR Ca2+ release [56]. More importantly, CaMKII was found to be activated in cardiac hypertrophy and it induced dilated cardiomyopathy and HF, whereas CaMKII inhibition protected against these alterations [57,58]. In conclusion, hyperphosphorylation of RyR2 leads to increased diastolic Ca2+ leak in heart failure, but the effect of CaMKII is more specific on RyR2, and CaMKII phosphorylation is more important than PKA.
Another accessory protein (beside FKBP12.6), critical for the stable closed state of the channel, is calmodulin (CaM) [59]. CaM binding was shown to be low both in heart failure and some CPVT mutants, which were associated with a leaky channel phenotype [60,61]. Supporting these results, a more recent study demonstrated that enhancing the binding affinity of RyR2 to CaM by genetic modification of RyR2 reduced the expression of hypertrophy-related genes, thus suppressing the development of hypertrophy, improving intracellular Ca2+ signaling, and rescuing mice suffering from pressure-overload induced hypertrophy [62].

2.2. Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT)

CPVT is an inherited disorder, linked to missense mutations of RyR2 (CPVT-1) (or other ECC proteins, including the SR Ca2+ buffer- and RyR-accessory protein calsequestrin (CPVT-2), CaM, Triadin, and the trans-2,3-enoyl-CoA reductase-like gene (TECRL)) [63,64]. This review discusses the most common forms of CPVT: CPVT-1 and CPVT-2, representing ≈70% and <5% of all cases, respectively. To date, more than 170 CPVT-1 linked mutations have been identified [63].
CPVT is a recognized cause of sudden cardiac death. Patients exhibit ventricular tachycardia only during catecholaminergic stimulation (e.g., during exercise or emotional stress), otherwise they are asymptomatic and present physiological ECGs at rest. Their heart is structurally normal, indicating that the excitation-contraction coupling (ECC) is intact under baseline conditions [65,66,67,68]. Moreover, normally functioning ECC implies CICR, and thus the sensitivity of RyR2 to cytoplasmic Ca2+ is unaltered in CPVT-1. While under catecholaminergic stimulation, when the SR Ca2+ content increases (due to phospholamban phosphorylation), CPVT develops. Chen’s group showed that it is the high SOICR propensity owing to low opening threshold of RyR2 to luminal [Ca2+] that accounts for the manifestation of CPVT (gain-of-function) [69,70]. Oversensitive RyR2s are repeatedly gated in diastole by increased SR Ca2+ load, which results in premature Ca2+ release, DADs, and tachycardia. An alternative pathomechanism is suggested by the evidence that mutant RyR2s display enhanced activity when phosphorylated by PKA. Similarly to that of CHF’s pathomechanism, Marks’s group proposed that mutant channels have lower FKBP12.6 affinity to the channels, and RyR2 phosphorylation by PKA dissociates the protein from RyR2, creating leaky channels [71]. The role of FKBP12.6 in the process, however, remains highly controversial. Many other groups reported either increased or decreased FKBP12.6 affinity for various mutants. Supporting the FKBP12.6 dissociation model, Marks’ group showed that the FKBP12.6 knockout mice were afflicted by exercise-induced sudden cardiac death, and their cardiomyocytes displayed an increased rate of DADs [71]. In contrast, Chen and colleagues did not observe CPVT phenotype (or any kind of abnormality) in mice with the same genetic background [72].
CPVT-2 is linked to the mutations of calsequestrin (CSQ). CSQ is the Ca2+ buffer of the SR and also a regulatory protein of RyR. CSQ was shown to inhibit RyR activity, while mutant CSQ loses control over RyR. Thus, in CPVT-2, mutant CSQ fails to suppress RyR activity, which is associated with high luminal Ca2+ sensitivity (i.e., low SOICR-threshold) and spontaneous diastolic Ca2+ release [73,74,75]. To date, 15 CPVT-2 mutations have been identified [76].

2.3. Calcium Release Deficiency Syndrome (CRDS)

Sudden cardiac death or aborted cardiac arrest also appear in patients carrying certain RyR2 mutations but have not shown prior symptoms or history of recurrent ventricular tachycardia. CRDS is characterized by ventricular arrhythmias, but negative CPVT. Therefore, CRDS patients produce negative results when tested with standard exercise test. However, on extended monitoring, epinephrine challenge or programmed electrical stimulation protocol with a pattern of long-burst, long-pause, and short-coupled ventricular extra-stimulus (LBLPS) provoked single ectopic events or non-sustained ventricular tachycardia (NSVT). These cardiac events could be suppressed by flecainide. Occasionally, NSVT transformed to ventricular fibrillation. Apparently, arrhythmia can be barely provoked in these patients, but when it happens, it is poorly tolerated [77].
CRDS is caused by loss-of-function mutations (or exon duplication) of RyR2, which raises the threshold of the channel for SOICR and decreases its sensitivity to both cytosolic Ca2+ and caffeine [77,78,79,80]. At a single cell level, early afterdepolarization (EAD) has been recorded [80]. The mechanism by which a CRDS-RyR2 could lead to ventricular tachycardia is explained by Chen’s group: when the SR load rarely reaches the higher threshold of the CRDS-RyR2, it causes EADs (known sources of re-entry), manifesting in ventricular tachycardia [78,81,82]. However, this model cannot explain why SOICR would be triggered during the terminal repolarization (just at the end of systole, in phase 2 and 3), when the SR load is the lowest during the whole heart cycle, rather than in phase 4 (diastole), when the SR is being loaded up. Therefore, we propose an alternative hypothesis for the generation of EADs, which needs to be experimentally tested. We assume that the suppressed activity of RyR would cause weaker inactivation of LTCC, resulting in prolonged Ca2+ influx and repolarization, with consequent activation of NCX (or reactivation of LTCC), resulting in regenerative cation current activation and EAD.

2.4. Arrhythmogenic Right Ventricular Dysplasia Type 2 (ARVD2)

ARVD2 (or arrhythmogenic right ventricular cardiomyopathy (ARVC)) is a cardiomyopathy linked to missense mutations of the RyR2 gene. It is characterized by thinning of the right ventricular wall, fibrofatty substitution of the myocardium, and electrical instability. ARVD2 is often responsible for the sudden cardiac deaths of juveniles and athletes. ARVD2 is clinically different from the other forms of ARVD because ARVD2 displays stress/exercise-induced ventricular arrhythmias [83,84].
These mutations are believed to destabilize the closed state of the channel, resulting in hyperactivation or hypersensitization to physiologically relevant agonists (gain-of-function). In accordance with this hypothesis, HEK293 cells overexpressing RyR2s with distinct ARVD-linked mutations showed long-lasting elevations of [Ca2+]i following caffeine activation [85,86]. However, one of the examined mutants (L433P) exhibited lower sensitivity to activation [85,86]. Some investigators showed reduced FKBP12.6 binding to ARVD RyR2, but it was not confirmed by other groups. ARVD2 RyR2 has a lower threshold for SOICR and shows abnormal termination of Ca2+ release, which abnormalities are expected to exacerbate during exercise/stress and is associated with electrical instability, explaining why certain ARVD2 patients positively respond to β-blockers (including carvedilol) and flecainide [70,87,88,89,90,91].
In addition, the significant SR leak observed in the hyperactive ARVD2 phenotype should cause EC uncoupling and impaired intracellular Ca2+ homeostasis, which is expected to induce apoptosis/necrosis. In accordance with this, earlier reports observed apoptosis in ARVD2 [83].

2.5. The Role of Oxidative Stress in Cardiac Pathology

As mitochondria constitute the significant volume of cardiomyocytes, by absorbing Ca2+, they could act as a significant Ca2+ buffer to modulate Ca2+ signaling. However, when they get overloaded with Ca2+, they generate excess amounts of reactive oxygen species (ROS), which would lead to the oxidation and CaMKII phosphorylation of RyR2 and enhanced diastolic Ca2+ release. This pathological process is a positive feedback cycle in which leak generates further leak. Experiments using ROS probes showed that enhanced RyR2 activity (induced by caffeine or intrinsic hyperactivity in CPVT myocytes) resulted in increased mitochondrial ROS emission, oxidized RyR2, and enhanced SR leak under β-adrenergic stimulation. Importantly, the SR leak was reduced by mitochondrial ROS scavenging. Furthermore, genetic inhibition of mitochondrial Ca2+ uptake was associated with the reduced signal of a ROS biosensor, indicating that increased mitochondrial ROS emission depends on Ca2+ influx into the mitochondria [92,93].
A recent study has shown that Mito-TEMPO, a mitochondrium-targeted antioxidant with a superoxide dismutase mimetic property (but not other, non-targeted antioxidants), prevented the onset of HF or reversed established HF, preserved the contractile function and abrogated sudden cardiac death in a mouse model. These results suggest that cardiac decompensation is a mitochondrial ROS-mediated process. Therefore, recently, mitochondrial targeted antioxidants, although they have not been used clinically for the treatment of heart diseases, are considered as promising new compounds with significant therapeutic potential [94].
Earlier, redox modification of RyR2 due to the production of ROS in CHF was suggested to contribute to the enhanced RyR2 sensitivity to luminal [Ca2+] and also account for the development of SOICR [45,95,96,97]. SOICR is linked to increased incidence of diastolic Ca2+ waves, which activate an inward NCX current during diastole, resulting in DADs, the electrical events known to be the primary causes of arrythmias in failing hearts. Finally, amplified diastolic activity of RyR2 leads to attenuated systolic Ca2+ transients in CHF.

3. RyR2 as a Therapeutic Target in RyR-Related Heart Diseases

Patients hospitalized with HF have a 5-year mortality rate of 43% on average [98]. Unfortunately, the currently used antiarrhythmic drug therapy has been shown to have only little benefit on survival [99]. Additional commonly used therapeutics aim at inhibiting neurohormonal stimuli activated during the disease, such as angiotensin II, endothelin-1, TNF-α, and catecholamines. These stimuli induce hypertrophy as drivers of downstream signaling pathways. Inhibiting these hypertrophic signals using angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers, aldosterone antagonists, and β-adrenergic receptor blockers are efficient drugs, as they significantly reduce the morbidity and mortality of patients. However, their therapeutic success is also limited, since the disease also continues to progress during these therapies [8,9,11].
The conventional drug therapy for CPVT combines β-blockers and Ca2+ channel blockers, which unfortunately only incompletely prevent sudden cardiac death [45].
Thus, there is an obvious need for new therapeutic approaches that inhibit hypertrophic signalization and prevent hypertrophy and arrhythmias. As reviewed above, both cardiac remodeling and triggered arrhythmias are Ca2+-dependent processes governed by diastolic SR Ca2+ leak, and thus therapies that prevent the functional remodeling of RyR2 are believed to be promising new strategies. Certainly, SOICR suppressors would also make a good anti-CPVT therapy. However, currently, there is not any clinically available RyR2-specific inhibitors, and the aim of this paper is to review RyR2-targeted pharmacology that has significant therapeutic potential. The summarized information about these drugs is found in Table 1.

3.1. Flecainide

Flecainide is a class Ic antiarrhythmic drug, a Na+ channel blocker. Because of its negative inotropic and proarrhythmic effects (in patients with structural heart diseases), it is contraindicated in CHF but applied in various non-structural heart diseases [100]. Flecainide showed significant efficiency in preventing ventricular arrhythmias during an exercise test in CPVT patients. The patients were recruited in the study because despite their ongoing conventional therapy (β-blocker + Ca2+ channel blocker), they still developed exercise-induced ventricular arrhythmias. When receiving additional flecainide therapy, 76% of them experienced partial or complete elimination of exercise-induced arrhythmias [101]. Flecainide was shown to inhibit spontaneous Ca2+ release events in cardiomyocytes [102,103]. The drug also inhibited RyR2 activity in single-channel current measurements, but only at positive membrane potentials that drive non-physiological ionic flux (from the cytoplasmic to the luminal side of RyR2), which questioned the direct role of RyR2 inhibition in the antiarrhythmic effect of flecainide and attributed the antiarrhythmic action entirely to Na+ current inhibition [104,105,106,107]. According to this alternative hypothesis, Na+ channel inhibition would lower the intracellular [Na+], resulting in higher NCX-mediated Ca2+ efflux, which would decrease SR Ca2+ content and therefore the probability of SOICR as well [103]. Very recently, Knollmann’s group resolved this debate using N-methylated flecainides [108]. They showed that the modified flecainide retained Na+ channel blocking ability but had reduced RyR2 inhibitory potency. The authors separated the effects attributed to Na+ channel or RyR2 inhibition by using the new compound as a research tool. They recorded spontaneous Ca2+ release events in calsequestrin-knockout cardiomyocytes (they exhibit CPVT phenotype) under experimental conditions when Na+ channel function was completely abolished, finding that flecainide, but not N-methylated flecainide, reduced the frequency of spontaneous Ca2+ release events. In addition, when the Na+ channels were functional, flecainide was still much more effective compared to the N-methylated version. Furthermore, flecainide, but not N-methylated flecainide, prevented ventricular tachycardia in calsequestrin-knockout mice. In these in vivo experiments, both drugs caused a similar degree of QRS prolongation, indicating that they caused equivalent Na+ channel block. These data suggest that RyR2 inhibition is an important component of flecainide’s antiarrhythmic action [108].

3.2. Carvedilol

Mortality rate in HF correlates with plasma catecholamine levels, and the therapeutic benefit of β-adrenergic receptor blockers support this finding [8,109,110]. Carvedilol is one of the most effective β-blockers that inhibit ventricular tachyarrhythmias and reduce mortality in HF patients. In addition to its β-receptor blocking effect, it also suppresses SOICR by inhibiting RyR2. There is a large difference in the carvedilol concentration required to suppress SOICR (0.3 µM) and it is necessary for the β-blocking effect (1 nM) [45]. Still, carvedilol’s SOICR-inhibiting activity may contribute to its therapeutic benefit because the drug reaches a much higher concentration in the cardiac tissue. Nevertheless, effective SOICR inhibition would probably require such higher plasma concentrations that would cause bradycardia as an adverse effect. Therefore, in order to separate these two effects, Chen’s group modified the structure of carvedilol so that it lost its β-receptor blocking effect but retained its RyR2 inhibiting action. These new compounds (VK-II-86, CS-I-34, and CS-I-59) prevented CPVT in mice but did not cause bradycardia. They also showed that VK-II-86, combined with metoprolol or bisoprolol, was more effective in suppressing ventricular tachyarrhythmias than the new drugs alone. The authors proposed that a combination therapy using selective SOICR inhibitors and β-blockers offer a promising new approach in the treatment of ventricular tachyarrhythmias [45,111].

3.3. Dantrolene

Dantrolene is a hydantoin derivative, indicated for use as a muscle relaxant to treat malignant hyperthermia (MH) crisis. MH is a rare idiosyncratic reaction of susceptible individuals to volatile anesthetics (e.g., halothane, isoflurane) and succinylcholine and characterized by muscle rigidity when the patient is exposed to these drugs during surgery. General skeletal muscle contracture due to high muscle work leads to the rapid increase of the body temperature, lactic acidosis, and hyperkalemia. These symptoms are fatal unless the patient is treated with the muscle relaxant dantrolene, and the body is cooled down. It turned out that MH susceptibility is linked to point mutations in the skeletal muscle-type RyR (RyR1), from which more than 200 have been discovered to date [82]. These mutations render RyR1 leaky and more sensitive to its ligands. Thus, volatile anesthetics trigger abnormal Ca2+ release in the resting muscle, which can be inhibited by dantrolene [112,113]. The similarity between the molecular pathomechanism of MH and cardiac ryanopathies raised the possibility that dantrolene might be an effective antiarrhythmic drug. Importantly, dantrolene did not exert adverse cardiac effects when applied in MH, and more recent studies demonstrated that the drug suppressed ventricular tachycardia [114,115]. Dantrolene (applied in 1 µM, while the therapeutic plasma concentration in MH is ≈10 µM) had antiarrhythmic effects and preserved inotropy in failing rabbit cardiomyocytes probably by inhibiting diastolic Ca2+ leak, increasing the threshold for SOICR and decreasing Ca2+ depletion from the SR. Interestingly, dantrolene remained ineffective in healthy myocytes [116]. Many other studies provided evidence that dantrolene inhibits CPVT in knock-in mice, inhibiting resting Ca2+ leak and spontaneous Ca2+ transients in myocytes derived from induced pluripotent stem cells carrying different CPVT mutations or isolated from failing human hearts. Dantrolene was also shown to suppress ventricular tachycardia in certain animal models [117,118,119,120,121]. These results raise the question as to how dantrolene acts on the two different RyR isoforms. The dantrolene binding site was identified first in RyR1. The same primary sequence in the equivalent region was found also in RyR2. Experimental data suggest that this putative binding site in RyR2 is unavailable in the healthy channel but becomes accessible for dantrolene after certain pathological modifications of the protein. It looks as though mutations and posttranslational modifications in HF disrupt certain interdomain interactions, important in stabilizing the closed state of the channel, which are believed to be re-stabilized by dantrolene [122,123]. An alternative explanation for dantrolene’s mechanism of action is that it restores CaM binding to RyR2, which provides a possible answer to the question why dantrolene affects Ca2+ release only in diseased hearts [60,124].
As the hepatotoxicity of dantrolene precludes its chronic therapeutic use, a safer dantrolene derivative would be favorable in therapy. The information reviewed here suggests that the development of safer and more RyR2-selective dantrolene derivatives would result in a useful, new class of antiarrhythmic drugs. Following these ideas, Laver and his colleagues discovered the effects of another hydantoin derivative, phenytoin (diphenylhydantoin), on the function of RyR2 [125]. Phenytoin is an approved drug prescribed in epilepsy. Its therapeutic benefit is mainly attributed to neuronal Na+ current inhibition. Moreover, it was previously shown to be antiarrhythmic [126,127]. Laver’s group found that phenytoin inhibited sheep RyR2 activity in a much lower concentration than the drug’s therapeutic plasma concentration. These effects were independent of the holding potential and CaM. However, the effect was [Ca2+]-dependent, as the drug was not potent at ≥10 µM cytoplasmic [Ca2+], indicating that it would not inhibit the amplitude of Ca2+ transients. Most importantly, phenytoin selectively inhibited RyR2s isolated from failing human hearts, as it did not affect RyR2s from healthy individuals [125].

3.4. Tetracaine Derivatives

The therapeutic benefit of RyR2 blockers observed in preclinical studies raises the question as to whether every RyR2 channel blocker would make a potentially good antiarrhythmic drug. The example of tetracaine suggests that the answer is “no”. Tetracaine was shown to abolish spontaneous Ca2+ release—its prolonged application caused a rebound activation of diastolic Ca2+ release because of the inhibition of diastolic Ca2+ leak resulting in elevated SR Ca2+ content and consequently higher SOICR propensity [102,128,129,130]. However, a similar undesirable phenomenon was not observed using flecainide, carvedilol, or dantrolene. The explanation is that while tetracaine causes long-lasting closed events (which allows for a longer time for SR overloading), flecainide and carvedilol employ a fast blocking mode of action, which only reduces the intra-burst event duration, and thus it reduces the open probability [45,102]. This mechanism will not allow SR Ca2+ overload because it has weaker impact on the Ca2+ flux through the channel. This is the reason why flecainide does not affect SR Ca2+ content either. In addition, flecainide was reported to lose inhibitory effects at systolic Ca2+ concentrations, which explains why flecainide does not decrease the amplitude of systolic Ca2+ release.
Recently, Abramson and his colleagues synthetized a novel tetracaine derivative, called EL20 (2-(diethylamino)ethyl 4-(butylamino)-2-methoxybenzoate), which lacks the proarrhythmic activity—at least partially—because EL20 reduces the open probability of RyR2 without inducing long-lasting closed events [131]. Interestingly, the inhibitory effect of the drug was “antagonized” by CaM. As RyR2 was demonstrated to be depleted of CaM in certain CPVTs and heart failure, EL20 may offer a selective therapeutic mechanism in CPVT and heart failure when CaM binding to RyR2 is low. Importantly, no ECG alteration was observed during EL20 treatment, indicating that Na+ current inhibition was not significant in the applied drug concentration during the time course of the experiment [131].

3.5. 1,4-Benzothiazepines

The benzothiazepine derivative K201 (4-[3(1-(4-benzyl)piperidinyl)propionyl]-7-methoxy-2,2,4,5-tetrahydro-1,4-benzothiazepine, also known as JTV519) was developed by Kirin Pharmaceutical Laboratory and first tested by Kaneko in 1994, who reported that it suppressed myocardial injury in a myofibrillar overcontraction model [132]. The author concluded that K201 acted as an “intracellular Ca2+ blocker”, which stimulated further experiments on the RyR2. In these studies, K201 was shown to prevent FKBP12.6 depletion from RyR2 and to restore FKBP12.6 binding to RyR2 in a canine heart failure model [133]. In another study, the drug stabilized the impaired function of the channel in vitro [134,135]. An elegant investigation by the Marks’ group used FKBP12.6 knockout (FKBP−/−) and haploinsufficient (FKBP+/−) mice to test the role of FKBP12.6 in the drug’s antiarrhythmic action. Both FKBP−/− and +/− mice exhibited exercise-induced ventricular tachycardia, but only the FKBP+/− mice were rescued from sudden cardiac death by K201 treatment. They also provided single-channel current data to show that the open probability of RyR2 isolated from exercised FKBP+/− mice was significantly lower in the case of K201-treated animals, but this inhibitory effect was not observed on RyR2s isolated from the FKBP−/− mice. In further experiments, cardiac SR vesicles of wild-type mice were subject to PKA treatment, which was supposed to strip FKBP off RyR2, and these channels were incorporated into planar lipid bilayers. FKBP12.6 was added to the channels, but this addition remained without effect, unless K201 was also included into the medium. These data strongly support the idea that the pharmacological effect of the drug is linked to FKBP12.6 binding to RyR2. Furthermore, a binding assay demonstrated that K201 increased the affinity of RyR2 for FKBP12.6 [136]. In line with these data, K201 was also shown to stabilize CPVT RyR2s in a closed state by enhancing FKBP12.6 binding [137,138].
In order to determine whether restoring FKBP12.6 binding to RyR2 improves cardiac function in CHF, MI was induced in wild-type and FKBP−/− mice. K201-treated wild-type mice showed a significant increase in ejection fraction, while K201 failed to cause improvement in FKBP−/− mice compared to placebo controls. Coimmunoprecipitation experiments demonstrated an increased amount of FKBP12.6 bound to RyR2 in K201-treated animals. These data suggest that FKBP12.6 is essential for the beneficial effects of K201 in CHF, and K201 acts by enhancing FKBP12.6 affinity to RyR2 [139].
K201 has non-specific Na+, Ca2+, and K+ current inhibiting actions that motivated further research to develop newer derivatives (collectively called rycals) that lack these off-target effects. One compound from this group, S107, was reported to have K201-like effects [33,140,141]. S107 inhibited the SR Ca2+ leak and reduced ventricular arrhythmias, infarct size, and left ventricular remodeling in a rat ischemia–reperfusion model [142]. Clinical trials using rycals were scheduled to begin in 2009 and have shown promising results in pilot clinical trials for the treatment of heart failure and cardiac arrhythmias.

4. Conclusions

Fixing dysregulated Ca2+ release offers an effective therapy in both CHF and CPVT; however, currently, there is not any clinically available RyR2-specific inhibitor. There is a substantial effort to find RyR2-selective inhibitors, but not all RyR2 blockers would theoretically make an appropriate antiarrhythmic medicine. Drug repurposing looks to be the primary strategy in RyR2-targeted drug development and modification of these lead molecules have promising prospects. These efforts probably will lead to many newer RyR2-selective drugs with strong therapeutic potential soon.

Author Contributions

N.S., writing—original draft preparation, writing—review and editing; Z.É.M., writing—original draft preparation, writing—review and editing; J.H., writing—review and editing; T.B., writing—review and editing; P.P.N., writing—review and editing, funding acquisition; J.A., writing—original draft preparation, writing—review and editing, funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Research Development and Innovation Fund of Hungary (NKFIH K138090, NKFIH FK135130, and TKP2020-NKA-04).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bers, D.M. Cardiac excitation-contraction coupling. Nature 2002, 415, 198–205. [Google Scholar] [CrossRef] [PubMed]
  2. Eisner, D.A.; Kashimura, T.; O’Neill, S.C.; Venetucci, L.A.; Trafford, A.W. What role does modulation of the ryanodine receptor play in cardiac inotropy and arrhythmogenesis? J. Mol. Cell. Cardiol. 2009, 46, 474–481. [Google Scholar] [CrossRef] [PubMed]
  3. Díaz, M.E.; Graham, H.K.; Trafford, A.W. Enhanced sarcolemmal Ca2+ efflux reduces sarcoplasmic reticulum Ca2+ content and systolic Ca2+ in cardiac hypertrophy. Cardiovasc. Res. 2004, 62, 538–547. [Google Scholar] [CrossRef]
  4. Venetucci, L.A.; Trafford, A.W.; Eisner, D.A. Increasing ryanodine receptor open probability alone does not produce arrhythmogenic calcium waves: Threshold sarcoplasmic reticulum calcium content is required. Circ. Res. 2007, 100, 105–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Venetucci, L.A.; Trafford, A.W.; O’Neill, S.C.; Eisner, D.A. Na/Ca exchange: Regulator of intracellular calcium and source of arrhythmias in the heart. Ann. N. Y. Acad. Sci. 2007, 1099, 315–325. [Google Scholar] [CrossRef]
  6. Venetucci, L.A.; Trafford, A.W.; O’Neill, S.C.; Eisner, D.A. The sarcoplasmic reticulum and arrhythmogenic calcium release. Cardiovasc. Res. 2008, 77, 285–292. [Google Scholar] [CrossRef] [Green Version]
  7. Bers, D.M. Ryanodine receptor S2808 phosphorylation in heart failure: Smoking gun or red herring. Circ. Res. 2012, 110, 796–799. [Google Scholar] [CrossRef] [Green Version]
  8. McMurray, J.J.; Pfeffer, M.A. Heart failure. Lancet 2005, 365, 1877–1889. [Google Scholar] [CrossRef]
  9. Burchfield, J.S.; Xie, M.; Hill, J.A. Pathological ventricular remodeling: Mechanisms: Part 1 of 2. Circulation 2013, 128, 388–400. [Google Scholar] [CrossRef] [Green Version]
  10. Xie, M.; Burchfield, J.S.; Hill, J.A. Pathological ventricular remodeling: Therapies: Part 2 of 2. Circulation 2013, 128, 1021–1030. [Google Scholar] [CrossRef]
  11. Sutton, M.G.; Sharpe, N. Left ventricular remodeling after myocardial infarction: Pathophysiology and therapy. Circulation 2000, 101, 2981–2988. [Google Scholar] [CrossRef] [PubMed]
  12. Diwan, A.; Dorn, G.W. Decompensation of cardiac hypertrophy: Cellular mechanisms and novel therapeutic targets. Physiology 2007, 22, 56–64. [Google Scholar] [CrossRef] [PubMed]
  13. Packer, M. What causes sudden death in patients with chronic heart failure and a reduced ejection fraction? Eur. Heart J. 2020, 41, 1757–1763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Heineke, J.; Molkentin, J.D. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat. Rev. Mol. Cell Biol. 2006, 7, 589–600. [Google Scholar] [CrossRef]
  15. Molkentin, J.D. Calcineurin-NFAT signaling regulates the cardiac hypertrophic response in coordination with the MAPKs. Cardiovasc. Res. 2004, 63, 467–475. [Google Scholar] [CrossRef]
  16. Colella, M.; Grisan, F.; Robert, V.; Turner, J.D.; Thomas, A.P.; Pozzan, T. Ca2+ oscillation frequency decoding in cardiac cell hypertrophy: Role of calcineurin/NFAT as Ca2+ signal integrators. Proc. Natl. Acad. Sci. USA 2008, 105, 2859–2864. [Google Scholar] [CrossRef] [Green Version]
  17. Ling, H.; Zhang, T.; Pereira, L.; Means, C.K.; Cheng, H.; Gu, Y.; Dalton, N.D.; Peterson, K.L.; Chen, J.; Bers, D.; et al. Requirement for Ca2+/calmodulin-dependent kinase II in the transition from pressure overload-induced cardiac hypertrophy to heart failure in mice. J. Clin. Investig. 2009, 119, 1230–1240. [Google Scholar] [CrossRef] [Green Version]
  18. Zou, Y.; Liang, Y.; Gong, H.; Zhou, N.; Ma, H.; Guan, A.; Sun, A.; Wang, P.; Niu, Y.; Jiang, H.; et al. Ryanodine receptor type 2 is required for the development of pressure overload-induced cardiac hypertrophy. Hypertension 2011, 58, 1099–1110. [Google Scholar] [CrossRef] [Green Version]
  19. Sedej, S.; Schmidt, A.; Denegri, M.; Walther, S.; Matovina, M.; Arnstein, G.; Gutschi, E.-M.; Windhager, I.; Ljubojević, S.; Negri, S.; et al. Subclinical abnormalities in sarcoplasmic reticulum Ca(2+) release promote eccentric myocardial remodeling and pump failure death in response to pressure overload. J. Am. Coll. Cardiol. 2014, 63, 1569–1579. [Google Scholar] [CrossRef] [Green Version]
  20. Duan, D.D. A leakage leads to failure: Roles of sarcoplasmic reticulum Ca2+ leak via RyR2 in heart failure progression. Hypertension 2010, 55, 849–851. [Google Scholar] [CrossRef] [Green Version]
  21. van Oort, R.J.; Respress, J.L.; Li, N.; Reynolds, C.; De Almeida, A.C.; Skapura, D.G.; De Windt, L.J.; Wehrens, X.H.T. Accelerated development of pressure overload-induced cardiac hypertrophy and dysfunction in an RyR2-R176Q knockin mouse model. Hypertension 2010, 55, 932–938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Houser, S.R.; Molkentin, J.D. Does contractile Ca2+ control calcineurin-NFAT signaling and pathological hypertrophy in cardiac myocytes? Sci. Signal. 2008, 1, pe31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Houser, S.R. Ca(2+) signaling domains responsible for cardiac hypertrophy and arrhythmias. Circ. Res. 2009, 104, 413–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Berridge, M.J. Remodelling Ca2+ signalling systems and cardiac hypertrophy. Biochem. Soc. Trans. 2006, 34, 228–231. [Google Scholar] [CrossRef] [PubMed]
  25. Alvarado, F.J.; Bos, J.M.; Yuchi, Z.; Valdivia, C.R.; Hernández, J.J.; Zhao, Y.-T.; Henderlong, D.S.; Chen, Y.; Booher, T.R.; Marcou, C.A.; et al. Cardiac hypertrophy and arrhythmia in mice induced by a mutation in ryanodine receptor 2. JCI Insight 2019, 5, e126544. [Google Scholar] [CrossRef] [Green Version]
  26. Bers, D.M.; Eisner, D.A.; Valdivia, H.H. Sarcoplasmic reticulum Ca2+ and heart failure: Roles of diastolic leak and Ca2+ transport. Circ. Res. 2003, 93, 487–490. [Google Scholar] [CrossRef] [Green Version]
  27. Bers, D.M. Calcium cycling and signaling in cardiac myocytes. Annu. Rev. Physiol. 2008, 70, 23–49. [Google Scholar] [CrossRef] [Green Version]
  28. Shannon, T.R.; Pogwizd, S.M.; Bers, D.M. Elevated sarcoplasmic reticulum Ca2+ leak in intact ventricular myocytes from rabbits in heart failure. Circ. Res. 2003, 93, 592–594. [Google Scholar] [CrossRef] [Green Version]
  29. George, C.H. Sarcoplasmic reticulum Ca2+ leak in heart failure: Mere observation or functional relevance? Cardiovasc. Res. 2008, 77, 302–314. [Google Scholar] [CrossRef] [Green Version]
  30. Engelhardt, S.; Hein, L.; Dyachenkow, V.; Kranias, E.G.; Isenberg, G.; Lohse, M.J. Altered calcium handling is critically involved in the cardiotoxic effects of chronic beta-adrenergic stimulation. Circulation 2004, 109, 1154–1160. [Google Scholar] [CrossRef] [Green Version]
  31. Marx, S.O.; Reiken, S.; Hisamatsu, Y.; Jayaraman, T.; Burkhoff, D.; Rosemblit, N.; Marks, A.R. PKA phosphorylation dissociates FKBP12.6 from the calcium release channel (ryanodine receptor): Defective regulation in failing hearts. Cell 2000, 101, 365–376. [Google Scholar] [CrossRef] [Green Version]
  32. Shan, J.; Kushnir, A.; Betzenhauser, M.J.; Reiken, S.; Li, J.; Lehnart, S.E.; Lindegger, N.; Mongillo, M.; Mohler, P.J.; Marks, A.R. Phosphorylation of the ryanodine receptor mediates the cardiac fight or flight response in mice. J. Clin. Investig. 2010, 120, 4388–4398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Shan, J.; Betzenhauser, M.J.; Kushnir, A.; Reiken, S.; Meli, A.C.; Wronska, A.; Dura, M.; Chen, B.-X.; Marks, A.R. Role of chronic ryanodine receptor phosphorylation in heart failure and β-adrenergic receptor blockade in mice. J. Clin. Investig. 2010, 120, 4375–4387. [Google Scholar] [CrossRef] [Green Version]
  34. Wehrens, X.H.T.; Lehnart, S.E.; Reiken, S.; Vest, J.A.; Wronska, A.; Marks, A.R. Ryanodine receptor/calcium release channel PKA phosphorylation: A critical mediator of heart failure progression. Proc. Natl. Acad. Sci. USA 2006, 103, 511–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Xiao, B.; Sutherland, C.; Walsh, M.P.; Chen, S.R.W. Protein kinase A phosphorylation at serine-2808 of the cardiac Ca2+-release channel (ryanodine receptor) does not dissociate 12.6-kDa FK506-binding protein (FKBP12.6). Circ. Res. 2004, 94, 487–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Xiao, B.; Jiang, M.T.; Zhao, M.; Yang, D.; Sutherland, C.; Lai, F.A.; Walsh, M.P.; Warltier, D.C.; Cheng, H.; Chen, S.R.W. Characterization of a novel PKA phosphorylation site, serine-2030, reveals no PKA hyperphosphorylation of the cardiac ryanodine receptor in canine heart failure. Circ. Res. 2005, 96, 847–855. [Google Scholar] [CrossRef] [Green Version]
  37. Jiang, M.T.; Lokuta, A.J.; Farrell, E.F.; Wolff, M.R.; Haworth, R.A.; Valdivia, H.H. Abnormal Ca2+ release, but normal ryanodine receptors, in canine and human heart failure. Circ. Res. 2002, 91, 1015–1022. [Google Scholar] [CrossRef] [Green Version]
  38. Stange, M.; Xu, L.; Balshaw, D.; Yamaguchi, N.; Meissner, G. Characterization of recombinant skeletal muscle (Ser-2843) and cardiac muscle (Ser-2809) ryanodine receptor phosphorylation mutants. J. Biol. Chem. 2003, 278, 51693–51702. [Google Scholar] [CrossRef] [Green Version]
  39. Guo, T.; Cornea, R.L.; Huke, S.; Camors, E.; Yang, Y.; Picht, E.; Fruen, B.R.; Bers, D.M. Kinetics of FKBP12.6 binding to ryanodine receptors in permeabilized cardiac myocytes and effects on Ca sparks. Circ. Res. 2010, 106, 1743–1752. [Google Scholar] [CrossRef]
  40. MacDonnell, S.M.; García-Rivas, G.; Scherman, J.A.; Kubo, H.; Chen, X.; Valdivia, H.; Houser, S.R. Adrenergic regulation of cardiac contractility does not involve phosphorylation of the cardiac ryanodine receptor at serine 2808. Circ. Res. 2008, 102, e65–e72. [Google Scholar] [CrossRef]
  41. Zhang, H.; Makarewich, C.A.; Kubo, H.; Wang, W.; Duran, J.M.; Li, Y.; Berretta, R.M.; Koch, W.J.; Chen, X.; Gao, E.; et al. Hyperphosphorylation of the cardiac ryanodine receptor at serine 2808 is not involved in cardiac dysfunction after myocardial infarction. Circ. Res. 2012, 110, 831–840. [Google Scholar] [CrossRef] [Green Version]
  42. Benkusky, N.A.; Weber, C.S.; Scherman, J.A.; Farrell, E.F.; Hacker, T.A.; John, M.C.; Powers, P.A.; Valdivia, H.H. Intact beta-adrenergic response and unmodified progression toward heart failure in mice with genetic ablation of a major protein kinase A phosphorylation site in the cardiac ryanodine receptor. Circ. Res. 2007, 101, 819–829. [Google Scholar] [CrossRef] [PubMed]
  43. Alvarado, F.J.; Chen, X.; Valdivia, H.H. Ablation of the cardiac ryanodine receptor phospho-site Ser2808 does not alter the adrenergic response or the progression to heart failure in mice. Elimination of the genetic background as critical variable. J. Mol. Cell. Cardiol. 2017, 103, 40–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Li, Y.; Kranias, E.G.; Mignery, G.A.; Bers, D.M. Protein kinase A phosphorylation of the ryanodine receptor does not affect calcium sparks in mouse ventricular myocytes. Circ. Res. 2002, 90, 309–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Zhou, Q.; Xiao, J.; Jiang, D.; Wang, R.; Vembaiyan, K.; Wang, A.; Smith, C.D.; Xie, C.; Chen, W.; Zhang, J.; et al. Carvedilol and its new analogs suppress arrhythmogenic store overload-induced Ca2+ release. Nat. Med. 2011, 17, 1003–1009. [Google Scholar] [CrossRef] [Green Version]
  46. Györke, S.; Terentyev, D. Modulation of ryanodine receptor by luminal calcium and accessory proteins in health and cardiac disease. Cardiovasc. Res. 2008, 77, 245–255. [Google Scholar] [CrossRef]
  47. Grimm, M.; Brown, J.H. Beta-adrenergic receptor signaling in the heart: Role of CaMKII. J. Mol. Cell. Cardiol. 2010, 48, 322–330. [Google Scholar] [CrossRef] [Green Version]
  48. Curran, J.; Hinton, M.J.; Ríos, E.; Bers, D.M.; Shannon, T.R. Beta-adrenergic enhancement of sarcoplasmic reticulum calcium leak in cardiac myocytes is mediated by calcium/calmodulin-dependent protein kinase. Circ. Res. 2007, 100, 391–398. [Google Scholar] [CrossRef] [Green Version]
  49. Takasago, T.; Imagawa, T.; Shigekawa, M. Phosphorylation of the cardiac ryanodine receptor by cAMP-dependent protein kinase. J. Biochem. 1989, 106, 872–877. [Google Scholar] [CrossRef]
  50. Guo, T.; Zhang, T.; Mestril, R.; Bers, D.M. Ca2+/Calmodulin-dependent protein kinase II phosphorylation of ryanodine receptor does affect calcium sparks in mouse ventricular myocytes. Circ. Res. 2006, 99, 398–406. [Google Scholar] [CrossRef] [Green Version]
  51. Wehrens, X.H.T.; Lehnart, S.E.; Reiken, S.R.; Marks, A.R. Ca2+/calmodulin-dependent protein kinase II phosphorylation regulates the cardiac ryanodine receptor. Circ. Res. 2004, 94, e61–e70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Said, M.; Becerra, R.; Valverde, C.A.; Kaetzel, M.A.; Dedman, J.R.; Mundiña-Weilenmann, C.; Wehrens, X.H.; Vittone, L.; Mattiazzi, A. Calcium-calmodulin dependent protein kinase II (CaMKII): A main signal responsible for early reperfusion arrhythmias. J. Mol. Cell. Cardiol. 2011, 51, 936–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Respress, J.L.; van Oort, R.J.; Li, N.; Rolim, N.; Dixit, S.S.; deAlmeida, A.; Voigt, N.; Lawrence, W.S.; Skapura, D.G.; Skårdal, K.; et al. Role of RyR2 phosphorylation at S2814 during heart failure progression. Circ. Res. 2012, 110, 1474–1483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ai, X.; Curran, J.W.; Shannon, T.R.; Bers, D.M.; Pogwizd, S.M. Ca2+/calmodulin-dependent protein kinase modulates cardiac ryanodine receptor phosphorylation and sarcoplasmic reticulum Ca2+ leak in heart failure. Circ. Res. 2005, 97, 1314–1322. [Google Scholar] [CrossRef] [Green Version]
  55. Grimm, M.; Ling, H.; Willeford, A.; Pereira, L.; Gray, C.B.B.; Erickson, J.R.; Sarma, S.; Respress, J.L.; Wehrens, X.H.T.; Bers, D.M.; et al. CaMKIIδ mediates β-adrenergic effects on RyR2 phosphorylation and SR Ca(2+) leak and the pathophysiological response to chronic β-adrenergic stimulation. J. Mol. Cell. Cardiol. 2015, 85, 282–291. [Google Scholar] [CrossRef] [Green Version]
  56. Maier, L.S.; Zhang, T.; Chen, L.; DeSantiago, J.; Brown, J.H.; Bers, D.M. Transgenic CaMKIIdeltaC overexpression uniquely alters cardiac myocyte Ca2+ handling: Reduced SR Ca2+ load and activated SR Ca2+ release. Circ. Res. 2003, 92, 904–911. [Google Scholar] [CrossRef] [Green Version]
  57. Zhang, T.; Maier, L.S.; Dalton, N.D.; Miyamoto, S.; Ross, J.; Bers, D.M.; Brown, J.H. The deltaC isoform of CaMKII is activated in cardiac hypertrophy and induces dilated cardiomyopathy and heart failure. Circ. Res. 2003, 92, 912–919. [Google Scholar] [CrossRef] [Green Version]
  58. Zhang, R.; Khoo, M.S.C.; Wu, Y.; Yang, Y.; Grueter, C.E.; Ni, G.; Price, E.E.; Thiel, W.; Guatimosim, S.; Song, L.-S.; et al. Calmodulin kinase II inhibition protects against structural heart disease. Nat. Med. 2005, 11, 409–417. [Google Scholar] [CrossRef]
  59. Xu, L.; Meissner, G. Mechanism of calmodulin inhibition of cardiac sarcoplasmic reticulum Ca2+ release channel (ryanodine receptor). Biophys. J. 2004, 86, 797–804. [Google Scholar] [CrossRef] [Green Version]
  60. Ono, M.; Yano, M.; Hino, A.; Suetomi, T.; Xu, X.; Susa, T.; Uchinoumi, H.; Tateishi, H.; Oda, T.; Okuda, S.; et al. Dissociation of calmodulin from cardiac ryanodine receptor causes aberrant Ca2+ release in heart failure. Cardiovasc. Res. 2010, 87, 609–617. [Google Scholar] [CrossRef] [Green Version]
  61. Xu, X.; Yano, M.; Uchinoumi, H.; Hino, A.; Suetomi, T.; Ono, M.; Tateishi, H.; Oda, T.; Okuda, S.; Doi, M.; et al. Defective calmodulin binding to the cardiac ryanodine receptor plays a key role in CPVT-associated channel dysfunction. Biochem. Biophys. Res. Commun. 2010, 394, 660–666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kohno, M.; Kobayashi, S.; Yamamoto, T.; Yoshitomi, R.; Kajii, T.; Fujii, S.; Nakamura, Y.; Kato, T.; Uchinoumi, H.; Oda, T.; et al. Enhancing calmodulin binding to cardiac ryanodine receptor completely inhibits pressure-overload induced hypertrophic signaling. Commun. Biol. 2020, 3, 714. [Google Scholar] [CrossRef] [PubMed]
  63. Priori, S.G.; Mazzanti, A.; Santiago, D.J.; Kukavica, D.; Trancuccio, A.; Kovacic, J.C. Precision Medicine in Catecholaminergic Polymorphic Ventricular Tachycardia: JACC Focus Seminar 5/5. J. Am. Coll. Cardiol. 2021, 77, 2592–2612. [Google Scholar] [CrossRef] [PubMed]
  64. Dulhunty, A.F. Molecular Changes in the Cardiac RyR2 With Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). Front. Physiol. 2022, 13, 830367. [Google Scholar] [CrossRef] [PubMed]
  65. Priori, S.G.; Napolitano, C.; Tiso, N.; Memmi, M.; Vignati, G.; Bloise, R.; Sorrentino, V.; Danieli, G.A. Mutations in the cardiac ryanodine receptor gene (hRyR2) underlie catecholaminergic polymorphic ventricular tachycardia. Circulation 2001, 103, 196–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. George, C.H.; Higgs, G.V.; Lai, F.A. Ryanodine receptor mutations associated with stress-induced ventricular tachycardia mediate increased calcium release in stimulated cardiomyocytes. Circ. Res. 2003, 93, 531–540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Uchinoumi, H.; Yano, M.; Suetomi, T.; Ono, M.; Xu, X.; Tateishi, H.; Oda, T.; Okuda, S.; Doi, M.; Kobayashi, S.; et al. Catecholaminergic polymorphic ventricular tachycardia is caused by mutation-linked defective conformational regulation of the ryanodine receptor. Circ. Res. 2010, 106, 1413–1424. [Google Scholar] [CrossRef]
  68. Priori, S.G.; Chen, S.R.W. Inherited dysfunction of sarcoplasmic reticulum Ca2+ handling and arrhythmogenesis. Circ. Res. 2011, 108, 871–883. [Google Scholar] [CrossRef] [Green Version]
  69. Jiang, D.; Xiao, B.; Yang, D.; Wang, R.; Choi, P.; Zhang, L.; Cheng, H.; Chen, S.R.W. RyR2 mutations linked to ventricular tachycardia and sudden death reduce the threshold for store-overload-induced Ca2+ release (SOICR). Proc. Natl. Acad. Sci. USA 2004, 101, 13062–13067. [Google Scholar] [CrossRef] [Green Version]
  70. Jiang, D.; Wang, R.; Xiao, B.; Kong, H.; Hunt, D.J.; Choi, P.; Zhang, L.; Chen, S.R.W. Enhanced store overload-induced Ca2+ release and channel sensitivity to luminal Ca2+ activation are common defects of RyR2 mutations linked to ventricular tachycardia and sudden death. Circ. Res. 2005, 97, 1173–1181. [Google Scholar] [CrossRef] [Green Version]
  71. Wehrens, X.H.T.; Lehnart, S.E.; Huang, F.; Vest, J.A.; Reiken, S.R.; Mohler, P.J.; Sun, J.; Guatimosim, S.; Song, L.S.; Rosemblit, N.; et al. FKBP12.6 deficiency and defective calcium release channel (ryanodine receptor) function linked to exercise-induced sudden cardiac death. Cell 2003, 113, 829–840. [Google Scholar] [CrossRef] [Green Version]
  72. Xiao, J.; Tian, X.; Jones, P.P.; Bolstad, J.; Kong, H.; Wang, R.; Zhang, L.; Duff, H.J.; Gillis, A.M.; Fleischer, S.; et al. Removal of FKBP12.6 does not alter the conductance and activation of the cardiac ryanodine receptor or the susceptibility to stress-induced ventricular arrhythmias. J. Biol. Chem. 2007, 282, 34828–34838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Terentyev, D.; Nori, A.; Santoro, M.; Viatchenko-Karpinski, S.; Kubalova, Z.; Gyorke, I.; Terentyeva, R.; Vedamoorthyrao, S.; Blom, N.A.; Valle, G.; et al. Abnormal interactions of calsequestrin with the ryanodine receptor calcium release channel complex linked to exercise-induced sudden cardiac death. Circ. Res. 2006, 98, 1151–1158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Terentyev, D.; Kubalova, Z.; Valle, G.; Nori, A.; Vedamoorthyrao, S.; Terentyeva, R.; Viatchenko-Karpinski, S.; Bers, D.M.; Williams, S.C.; Volpe, P.; et al. Modulation of SR Ca release by luminal Ca and calsequestrin in cardiac myocytes: Effects of CASQ2 mutations linked to sudden cardiac death. Biophys. J. 2008, 95, 2037–2048. [Google Scholar] [CrossRef] [Green Version]
  75. Kirchhefer, U.; Wehrmeister, D.; Postma, A.V.; Pohlentz, G.; Mormann, M.; Kucerova, D.; Müller, F.U.; Schmitz, W.; Schulze-Bahr, E.; Wilde, A.A.; et al. The human CASQ2 mutation K206N is associated with hyperglycosylation and altered cellular calcium handling. J. Mol. Cell. Cardiol. 2010, 49, 95–105. [Google Scholar] [CrossRef]
  76. Faggioni, M.; Kryshtal, D.O.; Knollmann, B.C. Calsequestrin mutations and catecholaminergic polymorphic ventricular tachycardia. Pediatr. Cardiol. 2012, 33, 959–967. [Google Scholar] [CrossRef] [Green Version]
  77. Ormerod, J.O.M.; Ormondroyd, E.; Li, Y.; Taylor, J.; Wei, J.; Guo, W.; Wang, R.; Sarton, C.N.S.; McGuire, K.; Dreau, H.M.P.; et al. Provocation Testing and Therapeutic Response in a Newly Described Channelopathy: RyR2 Calcium Release Deficiency Syndrome. Circ. Genom. Precis. Med. 2022, 15, e003589. [Google Scholar] [CrossRef]
  78. Li, Y.; Wei, J.; Guo, W.; Sun, B.; Estillore, J.P.; Wang, R.; Yoruk, A.; Roston, T.M.; Sanatani, S.; Wilde, A.A.M.; et al. Human RyR2 (Ryanodine Receptor 2) Loss-of-Function Mutations: Clinical Phenotypes and In Vitro Characterization. Circ. Arrhythm. Electrophysiol. 2021, 14, e010013. [Google Scholar] [CrossRef]
  79. Zhong, X.; Guo, W.; Wei, J.; Tang, Y.; Liu, Y.; Zhang, J.Z.; Tan, V.H.; Zhang, L.; Wang, R.; Jones, P.P.; et al. Identification of loss-of-function RyR2 mutations associated with idiopathic ventricular fibrillation and sudden death. Biosci. Rep. 2021, 41, BSR20210209. [Google Scholar] [CrossRef]
  80. Sun, B.; Yao, J.; Ni, M.; Wei, J.; Zhong, X.; Guo, W.; Zhang, L.; Wang, R.; Belke, D.; Chen, Y.-X.; et al. Cardiac ryanodine receptor calcium release deficiency syndrome. Sci. Transl. Med. 2021, 13, eaba7287. [Google Scholar] [CrossRef]
  81. Markandeya, Y.S.; Kamp, T.J. Rational strategy to stop arrhythmias: Early afterdepolarizations and L-type Ca2+ current. J. Gen. Physiol. 2015, 145, 475–479. [Google Scholar] [CrossRef] [PubMed]
  82. Riazi, S.; Kraeva, N.; Hopkins, P.M. Malignant Hyperthermia in the Post-Genomics Era: New Perspectives on an Old Concept. Anesthesiology 2018, 128, 168–180. [Google Scholar] [CrossRef] [PubMed]
  83. Tiso, N.; Stephan, D.A.; Nava, A.; Bagattin, A.; Devaney, J.M.; Stanchi, F.; Larderet, G.; Brahmbhatt, B.; Brown, K.; Bauce, B.; et al. Identification of mutations in the cardiac ryanodine receptor gene in families affected with arrhythmogenic right ventricular cardiomyopathy type 2 (ARVD2). Hum. Mol. Genet. 2001, 10, 189–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Tester, D.J.; Bombei, H.M.; Fitzgerald, K.K.; Giudicessi, J.R.; Pitel, B.A.; Thorland, E.C.; Russell, B.G.; Hamrick, S.K.; Kim, C.S.J.; Haglund-Turnquist, C.M.; et al. Identification of a Novel Homozygous Multi-Exon Duplication in RYR2 Among Children With Exertion-Related Unexplained Sudden Deaths in the Amish Community. JAMA Cardiol. 2020, 5, 13–18. [Google Scholar] [CrossRef] [PubMed]
  85. Thomas, N.L.; George, C.H.; Lai, F.A. Functional heterogeneity of ryanodine receptor mutations associated with sudden cardiac death. Cardiovasc. Res. 2004, 64, 52–60. [Google Scholar] [CrossRef] [Green Version]
  86. Tang, Y.; Tian, X.; Wang, R.; Fill, M.; Chen, S.R.W. Abnormal termination of Ca2+ release is a common defect of RyR2 mutations associated with cardiomyopathies. Circ. Res. 2012, 110, 968–977. [Google Scholar] [CrossRef] [Green Version]
  87. Tiso, N.; Salamon, M.; Bagattin, A.; Danieli, G.A.; Argenton, F.; Bortolussi, M. The binding of the RyR2 calcium channel to its gating protein FKBP12.6 is oppositely affected by ARVD2 and VTSIP mutations. Biochem. Biophys. Res. Commun. 2002, 299, 594–598. [Google Scholar] [CrossRef]
  88. Chelu, M.G.; Danila, C.I.; Gilman, C.P.; Hamilton, S.L. Regulation of ryanodine receptors by FK506 binding proteins. Trends Cardiovasc. Med. 2004, 14, 227–234. [Google Scholar] [CrossRef]
  89. Zissimopoulos, S.; Thomas, N.L.; Jamaluddin, W.W.; Lai, F.A. FKBP12.6 binding of ryanodine receptors carrying mutations associated with arrhythmogenic cardiac disease. Biochem. J. 2009, 419, 273–278. [Google Scholar] [CrossRef] [Green Version]
  90. Ermakov, S.; Gerstenfeld, E.P.; Svetlichnaya, Y.; Scheinman, M.M. Use of flecainide in combination antiarrhythmic therapy in patients with arrhythmogenic right ventricular cardiomyopathy. Heart Rhythm 2017, 14, 564–569. [Google Scholar] [CrossRef]
  91. Hiroi, Y.; Fujiu, K.; Komatsu, S.; Sonoda, M.; Sakomura, Y.; Imai, Y.; Oishi, Y.; Nakamura, F.; Ajiki, K.; Hayami, N.; et al. Carvedilol therapy improved left ventricular function in a patient with arrhythmogenic right ventricular cardiomyopathy. Jpn. Heart J. 2004, 45, 169–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Tow, B.D.; Deb, A.; Neupane, S.; Patel, S.; Reed, M.; Loper, A.-B.; Eliseev, R.A.; Knollmann, B.C.; Györke, S.; Liu, B. SR-mitochondria crosstalk shapes Ca signaling to impact pathophenotype in disease models marked by dysregulated intracellular Ca release. Cardiovasc. Res. 2021, cvab324. [Google Scholar] [CrossRef] [PubMed]
  93. Hamilton, S.; Terentyeva, R.; Martin, B.; Perger, F.; Li, J.; Stepanov, A.; Bonilla, I.M.; Knollmann, B.C.; Radwański, P.B.; Györke, S.; et al. Increased RyR2 activity is exacerbated by calcium leak-induced mitochondrial ROS. Basic Res. Cardiol. 2020, 115, 38. [Google Scholar] [CrossRef]
  94. Dey, S.; DeMazumder, D.; Sidor, A.; Foster, D.B.; O’Rourke, B. Mitochondrial ROS Drive Sudden Cardiac Death and Chronic Proteome Remodeling in Heart Failure. Circ. Res. 2018, 123, 356–371. [Google Scholar] [CrossRef] [PubMed]
  95. Belevych, A.E.; Terentyev, D.; Viatchenko-Karpinski, S.; Terentyeva, R.; Sridhar, A.; Nishijima, Y.; Wilson, L.D.; Cardounel, A.J.; Laurita, K.R.; Carnes, C.A.; et al. Redox modification of ryanodine receptors underlies calcium alternans in a canine model of sudden cardiac death. Cardiovasc. Res. 2009, 84, 387–395. [Google Scholar] [CrossRef] [PubMed]
  96. Terentyev, D.; Györke, I.; Belevych, A.E.; Terentyeva, R.; Sridhar, A.; Nishijima, Y.; de Blanco, E.C.; Khanna, S.; Sen, C.K.; Cardounel, A.J.; et al. Redox modification of ryanodine receptors contributes to sarcoplasmic reticulum Ca2+ leak in chronic heart failure. Circ. Res. 2008, 103, 1466–1472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Kubalova, Z.; Terentyev, D.; Viatchenko-Karpinski, S.; Nishijima, Y.; Györke, I.; Terentyeva, R.; da Cuñha, D.N.Q.; Sridhar, A.; Feldman, D.S.; Hamlin, R.L.; et al. Abnormal intrastore calcium signaling in chronic heart failure. Proc. Natl. Acad. Sci. USA 2005, 102, 14104–14109. [Google Scholar] [CrossRef] [Green Version]
  98. Roger, V.L.; Go, A.S.; Lloyd-Jones, D.M.; Benjamin, E.J.; Berry, J.D.; Borden, W.B.; Bravata, D.M.; Dai, S.; Ford, E.S.; Fox, C.S.; et al. Heart disease and stroke statistics—2012 update: A report from the American Heart Association. Circulation 2012, 125, e2–e220. [Google Scholar] [CrossRef]
  99. Kamath, G.S.; Mittal, S. The role of antiarrhythmic drug therapy for the prevention of sudden cardiac death. Prog. Cardiovasc. Dis. 2008, 50, 439–448. [Google Scholar] [CrossRef]
  100. McCauley, M.D.; Wehrens, X.H.T. Targeting ryanodine receptors for anti-arrhythmic therapy. Acta Pharmacol. Sin. 2011, 32, 749–757. [Google Scholar] [CrossRef] [Green Version]
  101. van der Werf, C.; Kannankeril, P.J.; Sacher, F.; Krahn, A.D.; Viskin, S.; Leenhardt, A.; Shimizu, W.; Sumitomo, N.; Fish, F.A.; Bhuiyan, Z.A.; et al. Flecainide therapy reduces exercise-induced ventricular arrhythmias in patients with catecholaminergic polymorphic ventricular tachycardia. J. Am. Coll. Cardiol. 2011, 57, 2244–2254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Watanabe, H.; Chopra, N.; Laver, D.; Hwang, H.S.; Davies, S.S.; Roach, D.E.; Duff, H.J.; Roden, D.M.; Wilde, A.A.M.; Knollmann, B.C. Flecainide prevents catecholaminergic polymorphic ventricular tachycardia in mice and humans. Nat. Med. 2009, 15, 380–383. [Google Scholar] [CrossRef] [PubMed]
  103. Sikkel, M.B.; Collins, T.P.; Rowlands, C.; Shah, M.; O’Gara, P.; Williams, A.J.; Harding, S.E.; Lyon, A.R.; MacLeod, K.T. Flecainide reduces Ca2+ spark and wave frequency via inhibition of the sarcolemmal sodium current. Cardiovasc. Res. 2013, 98, 286–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Mehra, D.; Imtiaz, M.S.; van Helden, D.F.; Knollmann, B.C.; Laver, D.R. Multiple modes of ryanodine receptor 2 inhibition by flecainide. Mol. Pharmacol. 2014, 86, 696–706. [Google Scholar] [CrossRef] [Green Version]
  105. Hwang, H.S.; Hasdemir, C.; Laver, D.; Mehra, D.; Turhan, K.; Faggioni, M.; Yin, H.; Knollmann, B.C. Inhibition of cardiac Ca2+ release channels (RyR2) determines efficacy of class I antiarrhythmic drugs in catecholaminergic polymorphic ventricular tachycardia. Circ. Arrhythm. Electrophysiol. 2011, 4, 128–135. [Google Scholar] [CrossRef] [Green Version]
  106. Bannister, M.L.; Thomas, N.L.; Sikkel, M.B.; Mukherjee, S.; Maxwell, C.; MacLeod, K.T.; George, C.H.; Williams, A.J. The mechanism of flecainide action in CPVT does not involve a direct effect on RyR2. Circ. Res. 2015, 116, 1324–1335. [Google Scholar] [CrossRef] [Green Version]
  107. Liu, N.; Denegri, M.; Ruan, Y.; Avelino-Cruz, J.E.; Perissi, A.; Negri, S.; Napolitano, C.; Coetzee, W.A.; Boyden, P.A.; Priori, S.G. Short communication: Flecainide exerts an antiarrhythmic effect in a mouse model of catecholaminergic polymorphic ventricular tachycardia by increasing the threshold for triggered activity. Circ. Res. 2011, 109, 291–295. [Google Scholar] [CrossRef] [Green Version]
  108. Kryshtal, D.O.; Blackwell, D.J.; Egly, C.L.; Smith, A.N.; Batiste, S.M.; Johnston, J.N.; Laver, D.R.; Knollmann, B.C. RYR2 Channel Inhibition Is the Principal Mechanism of Flecainide Action in CPVT. Circ. Res. 2021, 128, 321–331. [Google Scholar] [CrossRef]
  109. Cohn, J.N.; Levine, T.B.; Olivari, M.T.; Garberg, V.; Lura, D.; Francis, G.S.; Simon, A.B.; Rector, T. Plasma norepinephrine as a guide to prognosis in patients with chronic congestive heart failure. N. Engl. J. Med. 1984, 311, 819–823. [Google Scholar] [CrossRef] [Green Version]
  110. Barry, W.H.; Gilbert, E.M. How do beta-blockers improve ventricular function in patients with congestive heart failure? Circulation 2003, 107, 2395–2397. [Google Scholar] [CrossRef] [Green Version]
  111. Zhang, J.; Zhou, Q.; Smith, C.D.; Chen, H.; Tan, Z.; Chen, B.; Nani, A.; Wu, G.; Song, L.-S.; Fill, M.; et al. Non-β-blocking R-carvedilol enantiomer suppresses Ca2+ waves and stress-induced ventricular tachyarrhythmia without lowering heart rate or blood pressure. Biochem. J. 2015, 470, 233–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. MacLennan, D.H.; Phillips, M.S. Malignant hyperthermia. Science 1992, 256, 789–794. [Google Scholar] [CrossRef] [PubMed]
  113. Yuchi, Z.; Lau, K.; Van Petegem, F. Disease mutations in the ryanodine receptor central region: Crystal structures of a phosphorylation hot spot domain. Structure 2012, 20, 1201–1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Fruen, B.R.; Mickelson, J.R.; Louis, C.F. Dantrolene inhibition of sarcoplasmic reticulum Ca2+ release by direct and specific action at skeletal muscle ryanodine receptors. J. Biol. Chem. 1997, 272, 26965–26971. [Google Scholar] [CrossRef] [Green Version]
  115. Roden, D.M.; Knollmann, B.C. Dantrolene: From better bacon to a treatment for ventricular fibrillation. Circulation 2014, 129, 834–836. [Google Scholar] [CrossRef] [Green Version]
  116. Maxwell, J.T.; Domeier, T.L.; Blatter, L.A. Dantrolene prevents arrhythmogenic Ca2+ release in heart failure. Am. J. Physiol. Heart Circ. Physiol. 2012, 302, H953–H963. [Google Scholar] [CrossRef] [Green Version]
  117. Kobayashi, S.; Yano, M.; Suetomi, T.; Ono, M.; Tateishi, H.; Mochizuki, M.; Xu, X.; Uchinoumi, H.; Okuda, S.; Yamamoto, T.; et al. Dantrolene, a therapeutic agent for malignant hyperthermia, markedly improves the function of failing cardiomyocytes by stabilizing interdomain interactions within the ryanodine receptor. J. Am. Coll. Cardiol. 2009, 53, 1993–2005. [Google Scholar] [CrossRef] [Green Version]
  118. Kobayashi, S.; Yano, M.; Uchinoumi, H.; Suetomi, T.; Susa, T.; Ono, M.; Xu, X.; Tateishi, H.; Oda, T.; Okuda, S.; et al. Dantrolene, a therapeutic agent for malignant hyperthermia, inhibits catecholaminergic polymorphic ventricular tachycardia in a RyR2(R2474S/+) knock-in mouse model. Circ. J. 2010, 74, 2579–2584. [Google Scholar] [CrossRef] [Green Version]
  119. Zamiri, N.; Massé, S.; Ramadeen, A.; Kusha, M.; Hu, X.; Azam, M.A.; Liu, J.; Lai, P.F.H.; Vigmond, E.J.; Boyle, P.M.; et al. Dantrolene improves survival after ventricular fibrillation by mitigating impaired calcium handling in animal models. Circulation 2014, 129, 875–885. [Google Scholar] [CrossRef] [Green Version]
  120. Jung, C.B.; Moretti, A.; Schnitzler, M.M.Y.; Iop, L.; Storch, U.; Bellin, M.; Dorn, T.; Ruppenthal, S.; Pfeiffer, S.; Goedel, A.; et al. Dantrolene rescues arrhythmogenic RYR2 defect in a patient-specific stem cell model of catecholaminergic polymorphic ventricular tachycardia. EMBO Mol. Med. 2012, 4, 180–191. [Google Scholar] [CrossRef]
  121. Hartmann, N.; Pabel, S.; Herting, J.; Schatter, F.; Renner, A.; Gummert, J.; Schotola, H.; Danner, B.C.; Maier, L.S.; Frey, N.; et al. Antiarrhythmic effects of dantrolene in human diseased cardiomyocytes. Heart Rhythm 2017, 14, 412–419. [Google Scholar] [CrossRef] [PubMed]
  122. Paul-Pletzer, K.; Yamamoto, T.; Ikemoto, N.; Jimenez, L.S.; Morimoto, H.; Williams, P.G.; Ma, J.; Parness, J. Probing a putative dantrolene-binding site on the cardiac ryanodine receptor. Biochem. J. 2005, 387, 905–909. [Google Scholar] [CrossRef] [PubMed]
  123. Kobayashi, S.; Bannister, M.L.; Gangopadhyay, J.P.; Hamada, T.; Parness, J.; Ikemoto, N. Dantrolene stabilizes domain interactions within the ryanodine receptor. J. Biol. Chem. 2005, 280, 6580–6587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kajii, T.; Kobayashi, S.; Shiba, S.; Fujii, S.; Tamitani, M.; Kohno, M.; Nakamura, Y.; Nanno, T.; Kato, T.; Okuda, S.; et al. Dantrolene prevents ventricular tachycardia by stabilizing the ryanodine receptor in pressure- overload induced failing hearts. Biochem. Biophys. Res. Commun. 2020, 521, 57–63. [Google Scholar] [CrossRef]
  125. Ashna, A.; van Helden, D.F.; Dos Remedios, C.; Molenaar, P.; Laver, D.R. Phenytoin Reduces Activity of Cardiac Ryanodine Receptor 2; A Potential Mechanism for Its Cardioprotective Action. Mol. Pharmacol. 2020, 97, 250–258. [Google Scholar] [CrossRef]
  126. Conn, R.D. Diphenylhydantoin sodium in cardiac arrhythmias. N. Engl. J. Med. 1965, 272, 277–282. [Google Scholar] [CrossRef]
  127. Rosen, M.; Lisak, R.; Rubin, I.L. Diphenylhydantoin in cardiac arrhythmias. Am. J. Cardiol. 1967, 20, 674–678. [Google Scholar] [CrossRef]
  128. Györke, S.; Lukyanenko, V.; Györke, I. Dual effects of tetracaine on spontaneous calcium release in rat ventricular myocytes. J. Physiol. 1997, 500 Pt 2, 297–309. [Google Scholar] [CrossRef]
  129. Venetucci, L.A.; Trafford, A.W.; Díaz, M.E.; O’Neill, S.C.; Eisner, D.A. Reducing ryanodine receptor open probability as a means to abolish spontaneous Ca2+ release and increase Ca2+ transient amplitude in adult ventricular myocytes. Circ. Res. 2006, 98, 1299–1305. [Google Scholar] [CrossRef] [Green Version]
  130. Valdivia, H.H.; Valdivia, C.R. Tetracaine derivatives for catecholaminergic polymorphic ventricular tachycardia: New drugs for correction of diastolic Ca2+ leak? Heart Rhythm 2018, 15, 587–588. [Google Scholar] [CrossRef]
  131. Klipp, R.C.; Li, N.; Wang, Q.; Word, T.A.; Sibrian-Vazquez, M.; Strongin, R.M.; Wehrens, X.H.T.; Abramson, J.J. EL20, a potent antiarrhythmic compound, selectively inhibits calmodulin-deficient ryanodine receptor type 2. Heart Rhythm 2018, 15, 578–586. [Google Scholar] [CrossRef] [PubMed]
  132. Kaneko, N. New 1,4-Benzothiazepine Derivative, K201, demonstrates cardioprotective effects against sudden cardiac cell death and in-tracellular calcium blocking action. Drug Dev. Res. 1994, 33, 429–438. [Google Scholar] [CrossRef]
  133. Yano, M.; Kobayashi, S.; Kohno, M.; Doi, M.; Tokuhisa, T.; Okuda, S.; Suetsugu, M.; Hisaoka, T.; Obayashi, M.; Ohkusa, T.; et al. FKBP12.6-mediated stabilization of calcium-release channel (ryanodine receptor) as a novel therapeutic strategy against heart failure. Circulation 2003, 107, 477–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Kohno, M.; Yano, M.; Kobayashi, S.; Doi, M.; Oda, T.; Tokuhisa, T.; Okuda, S.; Ohkusa, T.; Kohno, M.; Matsuzaki, M. A new cardioprotective agent, JTV519, improves defective channel gating of ryanodine receptor in heart failure. Am. J. Physiol. Heart Circ. Physiol. 2003, 284, H1035–H1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Sacherer, M.; Sedej, S.; Wakuła, P.; Wallner, M.; Vos, M.A.; Kockskämper, J.; Stiegler, P.; Sereinigg, M.; von Lewinski, D.; Antoons, G.; et al. JTV519 (K201) reduces sarcoplasmic reticulum Ca2+ leak and improves diastolic function in vitro in murine and human non-failing myocardium. Br. J. Pharmacol. 2012, 167, 493–504. [Google Scholar] [CrossRef] [Green Version]
  136. Wehrens, X.H.T.; Lehnart, S.E.; Reiken, S.R.; Deng, S.-X.; Vest, J.A.; Cervantes, D.; Coromilas, J.; Landry, D.W.; Marks, A.R. Protection from cardiac arrhythmia through ryanodine receptor-stabilizing protein calstabin2. Science 2004, 304, 292–296. [Google Scholar] [CrossRef]
  137. Lehnart, S.E.; Wehrens, X.H.T.; Laitinen, P.J.; Reiken, S.R.; Deng, S.-X.; Cheng, Z.; Landry, D.W.; Kontula, K.; Swan, H.; Marks, A.R. Sudden death in familial polymorphic ventricular tachycardia associated with calcium release channel (ryanodine receptor) leak. Circulation 2004, 109, 3208–3214. [Google Scholar] [CrossRef] [Green Version]
  138. Lehnart, S.E.; Terrenoire, C.; Reiken, S.; Wehrens, X.H.T.; Song, L.-S.; Tillman, E.J.; Mancarella, S.; Coromilas, J.; Lederer, W.J.; Kass, R.S.; et al. Stabilization of cardiac ryanodine receptor prevents intracellular calcium leak and arrhythmias. Proc. Natl. Acad. Sci. USA 2006, 103, 7906–7910. [Google Scholar] [CrossRef] [Green Version]
  139. Wehrens, X.H.T.; Lehnart, S.E.; Reiken, S.; van der Nagel, R.; Morales, R.; Sun, J.; Cheng, Z.; Deng, S.-X.; de Windt, L.J.; Landry, D.W.; et al. Enhancing calstabin binding to ryanodine receptors improves cardiac and skeletal muscle function in heart failure. Proc. Natl. Acad. Sci. USA 2005, 102, 9607–9612. [Google Scholar] [CrossRef] [Green Version]
  140. Lehnart, S.E.; Mongillo, M.; Bellinger, A.; Lindegger, N.; Chen, B.-X.; Hsueh, W.; Reiken, S.; Wronska, A.; Drew, L.J.; Ward, C.W.; et al. Leaky Ca2+ release channel/ryanodine receptor 2 causes seizures and sudden cardiac death in mice. J. Clin. Investig. 2008, 118, 2230–2245. [Google Scholar] [CrossRef] [Green Version]
  141. Kushnir, A.; Marks, A.R. Ryanodine receptor patents. Recent Pat. Biotechnol. 2012, 6, 157–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Fauconnier, J.; Meli, A.C.; Thireau, J.; Roberge, S.; Shan, J.; Sassi, Y.; Reiken, S.R.; Rauzier, J.-M.; Marchand, A.; Chauvier, D.; et al. Ryanodine receptor leak mediated by caspase-8 activation leads to left ventricular injury after myocardial ischemia-reperfusion. Proc. Natl. Acad. Sci. USA 2011, 108, 13258–13263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. RyR2 inhibitors used in ryanopathies.
Table 1. RyR2 inhibitors used in ryanopathies.
RyanopathyNameOriginal UseRyR-Specific DerivativeStatus of Trial
Congestive Heart Failure (CHF)CarvedilolBeta-blocker clinical
VK-II-86preclinical
DantroleneRyR1 preclinical
PhenytoinNa+ channel preclinical
K201 (JTV519)RyR2 preclinical
Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT)Carvedilol clinical
VK-II-86preclinical
Flecainide Na+ channel clinical
Dantrolene preclinical
(Tetracaine) EL20preclinical
K201 (JTV519) preclinical
Calcium Release Deficiency Syndrome (CRDS)Flecainide clinical
N-methylatedpreclinical
Arrhythmogenic Right Ventricular Dysplasia type 2 (ARVD2)Carvedilol clinical
Flecainide clinical
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Szentandrássy, N.; Magyar, Z.É.; Hevesi, J.; Bányász, T.; Nánási, P.P.; Almássy, J. Therapeutic Approaches of Ryanodine Receptor-Associated Heart Diseases. Int. J. Mol. Sci. 2022, 23, 4435. https://doi.org/10.3390/ijms23084435

AMA Style

Szentandrássy N, Magyar ZÉ, Hevesi J, Bányász T, Nánási PP, Almássy J. Therapeutic Approaches of Ryanodine Receptor-Associated Heart Diseases. International Journal of Molecular Sciences. 2022; 23(8):4435. https://doi.org/10.3390/ijms23084435

Chicago/Turabian Style

Szentandrássy, Norbert, Zsuzsanna É. Magyar, Judit Hevesi, Tamás Bányász, Péter P. Nánási, and János Almássy. 2022. "Therapeutic Approaches of Ryanodine Receptor-Associated Heart Diseases" International Journal of Molecular Sciences 23, no. 8: 4435. https://doi.org/10.3390/ijms23084435

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop