Next Article in Journal
Increased Prognostic Yield by Combined Assessment of Non-Contrast Computed Tomography Markers of Antithrombotic-Related Spontaneous Intracerebral Hemorrhage Expansion
Next Article in Special Issue
Pathogenesis and Management of Brugada Syndrome: Recent Advances and Protocol for Umbrella Reviews of Meta-Analyses in Major Arrhythmic Events Risk Stratification
Previous Article in Journal
Outcomes of Nonagenarians with Acute Myocardial Infarction with or without Coronary Intervention
Previous Article in Special Issue
In-Hospital and One-Year Outcomes of Patients after Early and Late Resuscitated Cardiac Arrest Complicating Acute Myocardial Infarction—Data from a Nationwide Database
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Caveolin-3 and Arrhythmias: Insights into the Molecular Mechanisms

Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan 430030, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2022, 11(6), 1595; https://doi.org/10.3390/jcm11061595
Submission received: 22 January 2022 / Revised: 2 March 2022 / Accepted: 9 March 2022 / Published: 14 March 2022

Abstract

:
Caveolin-3 is a muscle-specific protein on the membrane of myocytes correlated with a variety of cardiovascular diseases. It is now clear that the caveolin-3 plays a critical role in the cardiovascular system and a significant role in cardiac protective signaling. Mutations in the gene encoding caveolin-3 cause a broad spectrum of clinical phenotypes, ranging from persistent elevations in the serum levels of creatine kinase in asymptomatic humans to cardiomyopathy. The influence of Caveolin-3(CAV-3) mutations on current density parallels the effect on channel trafficking. For example, mutations in the CAV-3 gene promote ventricular arrhythmogenesis in long QT syndrome 9 by a combined decrease in the loss of the inward rectifier current (IK1) and gain of the late sodium current (INa-L). The functional significance of the caveolin-3 has proved that caveolin-3 overexpression or knockdown contributes to the occurrence and development of arrhythmias. Caveolin-3 overexpression could lead to reduced diastolic spontaneous Ca2+ waves, thus leading to the abnormal L-Type calcium channel current-induced ventricular arrhythmias. Moreover, CAV-3 knockdown resulted in a shift to more negative values in the hyperpolarization-activated cyclic nucleotide channel 4 current (IHCN4) activation curve and a significant decrease in IHCN4 whole-cell current density. Recent evidence indicates that caveolin-3 plays a significant role in adipose tissue and is related to obesity development. The role of caveolin-3 in glucose homeostasis has attracted increasing attention. This review highlights the underlining mechanisms of caveolin-3 in arrhythmia. Progress in this field may contribute to novel therapeutic approaches for patients prone to developing arrhythmia.

1. Introduction

The novel subcellular structures, named plasmalemmal vesicles, were first detected by Palade, G.E. et al. in 1953, and then renamed as caveolae intracellulares by Yamada, E. et al. due to their resemblance to ‘little caves’ [1]. Caveolins are the most essential proteins in caveolae, presenting in three isoforms: caveolin-1, caveolin-2, and caveolin-3. Caveolin-1 and caveolin-2 are co-expressed across many cell types, whereas caveolin-3 is specifically found in muscle tissues, such as skeletal and cardiac myocytes [2,3,4]. With the development of biochemical, cell biological, and genetic approaches, especially molecular markers, the molecular functions of caveolae have gradually been discovered, which involve the participation of homeostasis, most notably endocytosis, mechano-protection, and signal transduction. Composed of 151 amino acids, caveolin-3 has four major structural domains: the N-terminal domain, the scaffolding domain, the intramembrane domain, and the C-terminal domain [5,6,7]. Caveolin-3 exerts its effects as a scaffolding and regulatory protein for signaling molecules and moderators of ion channels and has already been linked to numerous human disease states, such as long QT syndrome, sudden infant death syndrome, myocardial hypertrophy, and diabetic cardiomyopathy [8,9,10] (Table 1). Furthermore, a study has demonstrated that the caveolin-3 protein can modify integrin function and mechanotransduction in the cardiac myocytes and intact heart; thus, modifications in caveolin-3 can result in the dysregulation of integrin function and predispose the heart to develop a myopathic phenotype [11]. Multiple ion channels are expressed in the caveolae in cardiomyocites, such as the L-Type calcium channel (LTCC), T-Type calcium channel (TTCC), voltage dependent sodium channel 1.5 (Nav1.5), the voltage-dependent K channel (Kv1.5), and inward rectifier potassium channel (Kir2.x) [12,13,14,15].

2. Caveolin-3 and Electrical Signal Propagation

2.1. The Sodium Current (INa)

Several studies have confirmed that caveolin-3 colocalizes with the cardiac sodium channel and interacts with the dystrophin–glycoprotein complex to target multiple ion channels including SCN5A-encoded cardiac sodium channels (SCN5A, also termed Nav1.5) to the cell surface membrane [12,16,24]. CAV-3 is a novel Long QT syndrome (LQTS)-associated gene with mutations producing a gain-of-function, LQT3-like molecular/cellular phenotype, as a pathogenic basis of sudden infant death syndrome (SIDS) [25]. It has been reported that the functional alteration of SCN5A resulting from the mutation of CAV-3(V14L, T78M, and L79R) is presumed to be the cause of sudden cardiac death in infants because of a significant fivefold increase in late sodium current compared with controls, just like LQT3 [16,17,26] (Table 2). Recent evidence indicates that neural nitric oxide synthase (nNOS), which mediates nitric oxide (NO) synthesis, and SCN5A form a complex with caveolin-3 in the heart, and the direct binding of caveolin-3 to nNOS suppresses the catalytic activity of nNOS [27,28]. Excessive NO synthesis and release mediated by nNOS in cardiomyocytes was shown to increase late INa [29]. It has been well documented that increased late INa caused by CAV-3 mutation in ventricular muscle was reversed by NOS inhibitor L-NMMA, which meant that caveolin-3 was identified as an important negative regulator for cardiac late INa through a NOS-associated mechanism and contributed to both inherited arrhythmia syndromes and acquired arrhythmias in conditions [30,31]. Moreover, beta-adrenergic receptor regulation was reported to increase current densities of caveolar SCN5A through both protein kinase A (PKA)-dependent phosphorylation of sodium channels and direct Gas interaction with caveolin-3 which promoted the presentation of SCN5A containing caveolae to the surface membrane [32]. The roles of caveolin-3 in prolonging action potential by binding to calmodulin, which binds SCN5A and increases its slow inactivation kinetics in response to the regional increase in the concentration of calcium, have been gradually discovered. Although the above studies have confirmed the important role of caveolin-3, its specific mechanism is still unclear and may be closely related to the regulation of ion channels.

2.2. The K+ Current (Ik)

The inward rectifier current (IK1), dominating the terminal phases of cellular repolarization, maintains the resting membrane potential close to the potassium equilibrium potential and contributes to final phase 3 repolarization [33]. Ik1 is mainly comprised of the rectifier potassium channel Kir2.1, which is one member of Kir2.x family in the human cardiac ventricle and is mainly encoded by KCNJ2A gene on chromosome 17 [34,35]. Kir2.x family have a unique intracellular pattern of distribution in association with specific caveolin-3 domains, which critically depends on interaction with Kir2.x- caveolin-3 binding motifs [36]. An early study demonstrated that CAV-3 gene mutation decreased the surface expression of Kir2.1 and Kir2.2 [36,37]. Its mechanism may be closely related to the disruption in normal membrane trafficking of caveolin-3, regulating cell surface expression of Kir2.x channels, then causing decreased Kir2.x current density. Decreased Kir2.1 protein expression or function can decrease phase 3 rapid repolarization current magnitude of action potential, and prolong QT intermittent period or action potential duration, causing cardiac arrhythmia [37,38,39,40]. Moreover, a study has revealed that caveolin-3 participated in common early anterograde trafficking mechanism of the NaV1.5-Kir2.1 channelsome, which may have contributed to the potential for arrhythmias [40].
The transient outward K+ current (Ito), responsible for rapid initial repolarization manifested as phase 1 of the AP, was partly constituted by Kv4.2 in human ventricular myocardium. Caveolin-3 is comparably co-localized with Kv4.2 channels by co-immunoprecipitation analysis. Overwhelming evidence has suggested that CAV-3 gene mutation results in not only slower activation and recovery of IKv4.2, which could cause a reduction of Ito at physiological heart rates, but decreased IKv4.2, which contributes to cardiac arrhythmia [31]. It has been reported that CAV-3 gene mutation leads to dysfunction of AngII contributing to modulate a variety of ionic currents in atrial myocytes, including Ito. In the heart, AngII binds two types of angiotensin receptors, namely, AngII receptor 1 (Ang1R) and AngII receptor 2 (Ang2R), with Ang1Rs mediating most of the AngII effects [41]. Ang1R is associated with caveolin-3 in mouse atrial myocytes that is required for the reduction in Ito by AngII [42]. There is increasing evidence that downstream signaling of Ang1R activation includes activation of protein kinase C (PKC) which catalyzes the hydrolysis of the membrane phospholipid, phosphatidylinositol biphosphate, producing diacylglycerol (DAG) [41,43,44,45]. Although the above studies have confirmed that the crucial role of caveolin-3 in the modulation of ionic currents, its specific mechanism is still unclear and need to be further elucidated (Figure 1).

2.3. The Ca+ Current (ICa)

Voltage-gated Ca2+ channels, including both the LTCC and TTCC, are the major handlers for the excitation-contraction coupling (ECC) and pacing activity of the heart [46,47,48]. TTCC have three different isoforms, CaV3.1(α1G), CaV3.2 (α1H), and CaV3.3 (α1I), which were consistently absent in human adult atrial and ventricular myocardium. In the heart, ICa,T participates in Ca2+ entry and Ca2+-dependent hormonal secretion, pacemaker activity, and arrhythmia. T-type CaV3.1 (α1G) Ca2+ channels play important roles in the spontaneous activity of pacemaker cells. CaV3.3 is not expressed in the heart. In the normal condition, CaV3.2 isoforms are undetectable in normal adult ventricular myocytes but are re-expressed during conditions of cardiac hypertrophy [49,50,51]. Considerable evidence indicates that the N terminus region of caveolin-3 closely interacts with CaV3.2 channels [52]. It has been reported that caveolin-3 could regulate protein kinase A modulation of the Ca(V)3.2 (alpha1H) T-type Ca2+ channels and attenuate cardiac hypertrophy via inhibition of T-type Ca2+ current modulated by protein kinase Cα in cardiomyocytes [52,53,54]. Caveolin-3 specifically inhibited the increased expression of ICav3.2 and suppressed the Ca2+-dependent hypertrophic calcineurin-NFAT (calcineurin/nuclear factor of activated T cell) signaling pathway under pathological cardiac hypertrophy condition, thus significantly decreased the peak ICav3.2 current density to improve cardiac function [53].
L-type Ca channels (LTCCs) plays the vital role in cardiac ECC initiated by the action potential. Recent work has shown that caveolin-3 was associated with t-tubule formation, LTCC current to the t-tubules, and localization of LTCC regulatory proteins, such as protein kinase A (PKA) and β2-adrenoceptors [15,55,56,57]. In the recently reported study, global loss of caveolin-3, rather than cardiac-specific deletion of caveolin-3 protein, resulting in the pathological loss of t-tubular ICa contributes to impaired excitation-contraction coupling and thereby cardiac function in vivo [58]. Cardiac contractile performance is mainly regulated by circulating catecholamines, which bind to β-adrenergic receptors, the main catecholamine-responsive receptors on the surface of cardiac myocytes. β-adrenergic receptors(β-ARs) have two isoforms, β1-AR and β2-ARs. Multiple studies report the clustering of β2-adrenergic receptors(β2-ARs), coupled to Gs proteins and thus, inducing cAMP production, have also been shown to co-immunoprecipitate with caveoin-3 [59]. PKA is a key mediator in the molecular mechanism of the caveolin-3 affecting LTCCs by stimulating β2-adrenoceptor expression [15,56].
Type 2 ryanodine receptor (RyR2), a key component of ECC in cardiomyocytes, is a cation-selective receptor channel located in the cardiac sarcoplasmic reticulum (SR) in the mammalian heart. A few Ca2+ ions pass through Cav1.2 to trigger a much larger Ca2+ release at concentration levels into the myoplasm through RyR2, thus activating action potential [60,61]. Recently, many studies have provided evidence that the RyR2 colocalized with caveolin-3 [62]. RyR2 and caveolin-3-associated dihydropyridine receptor (DHPR) forms a triple complex with HLP family which is one member of the cysteine-rich protein (Crip) [63]. The RyR2 modulates Ca2+ entry through caveolin-3-associated DHPR by regulating itself [15]. Caveolin-3 overexpression could lead to the reduced diastolic spontaneous Ca2+ waves by inhibiting the hyperphosphorylation of ryanodine receptor-2 (RyR2) at Ser2814, thus leading to the abnormal LTCC current-induced ventricular arrhythmias [64].
Table 2. The arrhythmia mechanism associated with change of CAV-3 expression.
Table 2. The arrhythmia mechanism associated with change of CAV-3 expression.
CAV-3 ExpressionFunctional AlterationArrhythmia ImplicationsRelated Mechanisms Ref.
CAV-3 mutation (V14L, T78M, L79R)Increased late sodium currentLQT3NOS-dependent S-nitrosylation of SCN5ACheng. J. et al. [30]
Decreased Kir2.x current densityLQT9Downstream Ang1R signaling involves the activation of PKCTyan. L. et al. [42]
Caveolin-3 overexpressionReduced diastolic spontaneous Ca2+ wavesVentricular arrhythmiasInhibition of RYR2 hyperphosphorylationZhang. ZH. et al. [64]
CAV-3 mutation (S141R)Increased HCN4 current densityLQTSNANA
Caveolin-3 downregulated expressionActivated ICl, swellAtrial fibrillation NANA
NOS, Nitric oxide synthase; RYR2, Ryanodine receptor-2; Ang1R, AngII receptor 1; PKC, Protein kinase C. NA, Not Available.
Striatin (STRN) is a dynamic protein that was originally purified from highly active adenylyl cyclase rich fractions and was found to bind caveolin-3 and calmodulin (CaM) in a calcium-sensitive manner [65]. Further studies attributed the role of caveolin-3 in the interaction between STRN and CaM regulating the maturation and organization of the ECC in ventricular cardiomyocytes. The expression level of STRN was inversely proportional to the interaction of caveolin-3 with the CaM/STRN complex. Thus, caveolin-3 may mediate STRN expression by producing the opposite phenotype to silence the STRN gene, but the specific mechanism needs to be further elucidated [66].

2.4. The Hyperpolarization-Activated Cyclic Nucleotide Channel 4 Current (IHCN4)

Hyperpolarization-activated cyclic nucleotide channel 4(HCN4), also named the pacemaker channel, is the dominant isoform of the sinus node region interacting with caveolin-3 [67]. HCN4 channel function is negatively modulated by caveolin-3 to modulate the channel’s activity [68]. Of note, increased IHCN4 activity in the ventricle was shown to provoke ventricular automaticity resulting in ventricular arrhythmias [69]. Interestingly, LQTs-associated CAV-3 mutations differentially modulate HCN4 channel function indicating a pathophysiological role in clinical manifestations. One study indicated that HCN4 current properties were differentially modulated by LQTS-associated CAV-3 mutations. S141R, a CAV-3 LQTS-associated mutation, significantly increased HCN4 current density. CAV-3 KO resulted in a shift to more negative values in the IHCN4 activation curve and a significant decrease in IHCN4 whole-cell current density, which indicates that caveolin-3 mutations significantly accelerated the activation kinetics of HCN4 [68].

2.5. The Volume-Activated Cl-Channel Current (ICl,swell)

Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia, and it has been associated with an increased risk of stroke, heart failure, and eventually, contributing to an increased risk of cardiac and total mortality. A high level of CAV-3 expression had a significant relationship with AF participants. Caveolin-3 concentrations in the serum samples were much higher in the group with persistent AF than the group with paroxysmal AF. Concentrations of caveolin-3 might be associated with the frequency and duration of AF [70]. AF associated with elevated chronic stretch is linked to a decrease in cardiomyocyte caveolae density and downregulation of the caveolin-3 [53,71].
Volume-activated Cl-channels are localized in the caveolae microdomains and can be activated on the mechanical stretch. Downregulation of caveolin-3 expression facilitates activation of ICl,swell and increases sensitivity to stretch 5- to 10-fold, promoting the development of AF. Caveolin-3-mediated activation of mechanosensitive ICl,swell is a critical cause of the triggering impulses that can initiate AF including AF, and this mechanism is exacerbated in the setting of chronically elevated blood pressures [72].

3. Caveolin-3 and Intercellular Communication

3.1. Caveolin-3 and ConnexIn 43

Gap junctions are collections of multiple intercellular channels comprised of connexons, to permit the rapid cell–cell transfer of action potentials, ensuring the coordinated contraction of the cardiomyocytes. ConnexIn 43 (Cx43) is the main component in cardiac gap junctions and is expressed in all atrial and ventricular myocytes. Reduced Cx43 expression can reduce function of gap junctions, resulting in promoting reentrant arrhythmias. There is extensive evidence that Cx43 plays a crucial role for rapid action potential transmission and signaling molecules that are associated with cardiac arrhythmias [73,74]. When the levels of full-length Cx43 protein are markedly reduced via an M213L mutation associated with an absence of GJA1-20k which is an auxiliary subunit for the trafficking of Cx43, the abnormal propagation of electrical impulse, including decrease in R wave amplitude, elongation of QRS complex duration, and increase in frequency of premature ventricular contractions (PVC), occurred in rats undergoing the loss of Cx43 gap junction that contributes to arrhythmias [75,76]. Oppositely, when Cx43 is activated by pinocembrin which is a flavonoid compound originated from propolis, Cx43 can be upregulated to alleviate ventricular arrhythmia in I/R rats [77]. Stem cell therapy in combination with enhanced protein expression of connexin-43 is a promising strategy against myocardial dysfunction, such as the post-infarction arrhythmias. More recently, data from the Rugowska, A et al. demonstrated that the increased expression of connexin-43 in human skeletal muscle-derived stem/progenitor cells reduced arrhythmogenic phenomena after their transplantation into the post-infarcted myocardium [78]. This is probably because that increased expression of Cx43 contributes to the reduction of inflammation and improvement of intercellular communication thus causing an indirect positive effect on calcium intracellular circulation.
Cx43 has been shown to interact with caveolin-3, and the mechanism of the relationship between Cx43 and caveolin-3 is still being explored. One study attributed a role of caveolin-1 in the regulation of Cx43. It maintains cardiac homeostasis by modulating cSrc activity. cSrc became activated to downregulate Cx43 without caveolin-1 expression. This process reduced ventricular conduction velocity and increased propensity for ventricular arrhythmias. Caveolin-3 regulating Cx43 function/expression may be a similar way as the Cx43 regulation of caveolin-1. However, more studies need to further confirm this hypothesis [79].
A study has shown that caveolin-3 was not only co-localized but interacts with the gap junction protein Cx43, by methods of double-hybridization and co-immunoprecipitation [80]. Moreover, the changes in caveolin-3 levels over time perfectly paralleled the pattern of changes in total Cx43 when phoneutria nigriventer spider venom (PNV) disrupt blood–brain barrier, which raise the possibility of cross-talk between Cx43 and caveolin-3 [81]. A previous study demonstrated that downregulation of caveolin-3 leads to inhibition of Cx43 gap junction communication in the lipopolysaccharide (LPS) treatment of astrocytes. Further, the specific knockout of caveolin-3 causes a downregulation of Cx43 and thus has a significant inhibitory effect on gap junctional intercellular communication (GJIC). All of those results indicated that caveolin-3 may play a crucial role in regulating Cx43 expression, but the signaling pathway between caveolin-3 and Cx43 merited further investigations [82].

3.2. Caveolin-3 and Dystrophin

The dystrophin, fundamental for muscle integrity, is a critical component of the dystrophin-glycoprotein complex that acts as a connection between the extracellular matrix and intracellular cytoskeleton [83]. Mutations in the dystrophin gene contribute to dystrophinopathies, which are mostly associated with cardiomyopathies [84]. A heart lacking functional dystrophin is mechanically weak [85]. Doyle, D.D. et al. verify that dystrophin co-localizes, co-fractionates, and co-immunoprecipitates with caveolin-3, which serves to suggest directions for further research that caveolin-3 may regulate physiological functions of dystrophin as upstream regulator [86]. Recent evidence indicates that dystrophin and its associated glycoproteins are downregulated in caveolin-3 overexpression heart, causing severe cardiac tissue degeneration, fibrosis and a reduction in cardiac functions [87]. Caveolin-3 may regulate the normal processing or stoichiometry of the dystrophin complex at the protein level [88].

3.3. Caveolin-3 and Adiponectin Receptor

Adiponectin (APN) is a benign adipokine secreted cytokine with reduced expression in obesity and diabetes [89]. The primary function of APN is to increase insulin sensitivity by sensitizing the insulin receptor signaling pathway. Adiponectin receptors are G protein-coupled receptors (GPCRs) and two receptor subtypes: AdipoR1 and AdipoR2. In both AdipoR1 and AdipoR2, the N-terminal domain exists in intracellular space and C-terminal domain presents in the external region of cells. Many studies have indicated that AdipoR1 colocalized with caveolin-3, forming AdipoR1/ caveolin-3 complex via specific caveolin-3 scaffolding domain binding motifs [90]. By interacting with AdipoR1, caveolin-3 corrals downstream molecules in close proximity with AdipoR1, thus enabling proper transmembrane signaling and cardioprotection. Recently, many studies have provided evidence that high fat diet-induced diabetes disrupted the expression of CAV-3, which deranged eNOS signaling in diabetic myocardium and then diminished the cardioprotective effects of APN [91,92,93,94]. Overall, caveolin-3 plays an essential role in APN-induced ceramidase recruitment and activation, although the involved detailed molecular mechanisms are still unknown.

4. Caveolin-3 and Metabolic Perturbation

4.1. Caveolin-3 and Insulin Resistance

Insulin resistance is the important defect in the pathophysiology of type 2 diabetes (T2DM). A major complication of diabetes is diabetic cardiomyopathy. The ion channel remodeling has been reported in the animal models of diabetic cardiomyopathy which could lead to a variety of arrhythmias, such as AF [95,96].
In recent years, the role of caveolin-3 in glucose homeostasis has attracted increasing attention. A previous study demonstrated that a variety of mutations were present in the CAV-3 gene among 1–1000 patients with T2DM. A previous assessment of patients with newly developed T2DM demonstrated that K15N muta tion located in the N-terminus of caveolin-3 may lead to changes in caveolin-3 secondary structure, thus causing decreased recombinant caveolin-3 expression [97]. It has been found that decreased expression and reduced localization of caveolin-3 by CAV-3-P104L mutation inhibited glucose uptake and glycogen synthesis [98]. The IR/PI3K/AKT/GLUT signaling pathway has been demonstrated to be a major mechanism in the development of insulin resistance [99,100]. Insulin receptor (IR) and GLUT-4 are associated with glucose metabolism, with their expression being regulated by caveolin-3 on the cell membrane. GLUT4 is one of the most important glucose transporters and is responsible for regulating and transporting 50% to 80% of body glucose. Caveolin-3 can enhance the expression of IR by stimulating IR kinase activity and activating PI3K/AKT signaling pathway. Activated AKT promoted the translocation of GLUT-4 to the plasma membrane and enhanced glucose uptake. There is evidence that increased CAV-3 expression contributed to GLUT4 translocation and thus ameliorated high-fat-diet (HFD)-induced glucose intolerance and insulin resistance, which indicated a positive correlation between the alternation of glucose metabolism and the level of caveolin-3 [93,101]. Conversely, decreased expression levels of CAV-3 were observed in diabetes animal models [102]. Decreased CAV-3 expression inhibited Akt phosphorylation, and thus protein expression of GLUT4 which are molecules downstream of Akt, was significantly decreased [98]. Moreover, caveolin-3 may play a pivotal role in 17β-estradiol (E2) actions on glucose metabolism, and there is evidence that E2 intervention reduces the incidence of diabetes [92,103]. Diabetes could cause inhibition of CAV-3 expression by regulating the activities of various enzymes and ion channels. Those changes lead to insulin resistance, inhibition of Cx43 gap junction communication and modulation of ion channel function, which contribute to susceptibility to arrhythmias.

4.2. Caveolin-3 and Adiposity

Obesity is a major concern because it is a risk factor for metabolic diseases that increase mortality rates. Recent evidence indicates that caveolin-3 plays a significant role in adipose tissue and is related to obesity development. A study has demonstrated that CAV-3-knockout mice increased adiposity [104]. Obesity is a complex multifactorial condition that contributes significantly to cardiovascular risk including AF. In animal models with adipose tissue, it has been found that obesity is also associated with a modest increase in QTc and QT dispersion [105,106]. Moreover, high-fat-diet-fed animals showed metabolic alterations, obesity, and insulin resistance along with the induced expression of muscle-specific caveolin-3 in retroperitoneal adipocytes and skeletal muscle in the initial phase. The mechanism was partly explained by the fact that continued exposure to the high-fat diet in the initial phase produces an increase in circulating glucose and insulin levels which could induce CAV-3 expression [107,108]. Indeed, previous studies have reported that animals fed on a high fat diet show increased oxidative stress in muscle tissue, which also could be partly explained by the induced expression of CAV-3 [109]. In a late phase, insulin resistance becomes apparent, accompanied by an impairment of caveolin-3 levels in skeletal muscle.

5. Perspective

Since it was discovered approximately 60 years ago, important functional roles and biochemical properties for caveolae have been identified. It is now clear that the caveolin-3 plays a critical role in the cardiovascular system and a significant role in cardiac protective signaling. CAV-3 mutations have been linked to the LQT9, and the cause of underlying action potential duration prolongation is incompletely understood. However, there are numerous difficulties to be overcome in the process from basic laboratory research to clinical application. Caveolin-3 dysfunctions have been responsible for inherited arrhythmias. The importance of caveolin-3 changes in arrhythmias and downstream microdomain dysregulation may have important implications for arrhythmia generation.
Understanding the composition and functional roles of caveolin-3 in the heart as well as their contribution to arrhythmia syndromes is only the beginning. Many critical questions remain to be answered. Further basic science research and eventual randomized clinical trials are needed to define the precise mechanisms and therapeutic potential of caveolin-3 in patients with arrhythmias.

Author Contributions

M.H. and J.Q. performed the study and drafted the manuscript; G.C. and Y.B. organized the study and edited the manuscript; G.C. and Y.W. revised the study. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (No. 82070383).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

We declare that we have no financial or personal relationships with other people or organizations that could inappropriately influence our work.

References

  1. Yamada, E. The fine structure of the gall bladder epithelium of the mouse. J. Biophys. Biochem. Cytol. 1955, 1, 445–458. [Google Scholar] [CrossRef] [Green Version]
  2. Rothberg, K.G.; Heuser, J.E.; Donzell, W.C.; Ying, Y.S.; Glenney, J.R.; Anderson, R.G. Caveolin, a protein component of caveolae membrane coats. Cell 1992, 68, 673–682. [Google Scholar] [CrossRef]
  3. Song, K.S.; Scherer, P.E.; Tang, Z.; Okamoto, T.; Li, S.; Chafel, M.; Chu, C.; Kohtz, D.S.; Lisanti, M.P. Expression of caveolin-3 in skeletal, cardiac, and smooth muscle cells. Caveolin-3 is a component of the sarcolemma and co-fractionates with dystrophin and dystrophin-associated glycoproteins. J. Biol. Chem. 1996, 271, 15160–15165. [Google Scholar] [CrossRef] [Green Version]
  4. Balijepalli, R.C.; Kamp, T.J. Caveolae, ion channels and cardiac arrhythmias. Prog. Biophys. Mol. Biol. 2008, 98, 149–160. [Google Scholar] [CrossRef] [Green Version]
  5. Kim, J.H.; Peng, D.; Schlebach, J.P.; Hadziselimovic, A.; Sanders, C.R. Modest effects of lipid modifications on the structure of caveolin-3. Biochemistry 2014, 53, 4320–4322. [Google Scholar] [CrossRef] [PubMed]
  6. Kim, J.H.; Schlebach, J.P.; Lu, Z.; Peng, D.; Reasoner, K.C.; Sanders, C.R. A pH-Mediated Topological Switch within the N-Terminal Domain of Human Caveolin-3. Biophys. J. 2016, 110, 2475–2485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Fernandez, I.; Ying, Y.; Albanesi, J.; Anderson, R.G. Mechanism of caveolin filament assembly. Proc. Natl. Acad. Sci. USA 2002, 99, 11193–11198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Koga, A.; Oka, N.; Kikuchi, T.; Miyazaki, H.; Kato, S.; Imaizumi, T. Adenovirus-mediated overexpression of caveolin-3 inhibits rat cardiomyocyte hypertrophy. Hypertension 2003, 42, 213–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Tsutsumi, Y.M.; Horikawa, Y.T.; Jennings, M.M.; Kidd, M.W.; Niesman, I.R.; Yokoyama, U.; Head, B.P.; Hagiwara, Y.; Ishikawa, Y.; Miyanohara, A.; et al. Cardiac-specific overexpression of caveolin-3 induces endogenous cardiac protection by mimicking ischemic preconditioning. Circulation 2008, 118, 1979–1988. [Google Scholar] [CrossRef] [Green Version]
  10. Parton, R.G. Caveolae: Structure, Function, and Relationship to Disease. Annu. Rev. Cell Dev. Biol. 2018, 34, 111–136. [Google Scholar] [CrossRef] [PubMed]
  11. Israeli-Rosenberg, S.; Chen, C.; Li, R.; Deussen, D.N.; Niesman, I.R.; Okada, H.; Patel, H.H.; Roth, D.M.; Ross, R.S. Caveolin modulates integrin function and mechanical activation in the cardiomyocyte. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2015, 29, 374–384. [Google Scholar] [CrossRef] [Green Version]
  12. Yarbrough, T.L.; Lu, T.; Lee, H.C.; Shibata, E.F. Localization of cardiac sodium channels in caveolin-rich membrane domains: Regulation of sodium current amplitude. Circ. Res. 2002, 90, 443–449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Martens, J.R.; Sakamoto, N.; Sullivan, S.A.; Grobaski, T.D.; Tamkun, M.M. Isoform-specific localization of voltage-gated K+ channels to distinct lipid raft populations. Targeting of Kv1.5 to caveolae. J. Biol. Chem. 2001, 276, 8409–8414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Bossuyt, J.; Taylor, B.E.; James-Kracke, M.; Hale, C.C. The cardiac sodium-calcium exchanger associates with caveolin-3. Ann. N. Y. Acad. Sci. 2002, 976, 197–204. [Google Scholar] [CrossRef] [PubMed]
  15. Balijepalli, R.C.; Foell, J.D.; Hall, D.D.; Hell, J.W.; Kamp, T.J. Localization of cardiac L-type Ca(2+) channels to a caveolar macromolecular signaling complex is required for beta(2)-adrenergic regulation. Proc. Natl. Acad. Sci. USA 2006, 103, 7500–7505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Vatta, M.; Ackerman, M.J.; Ye, B.; Makielski, J.C.; Ughanze, E.E.; Taylor, E.W.; Tester, D.J.; Balijepalli, R.C.; Foell, J.D.; Li, Z.; et al. Mutant caveolin-3 induces persistent late sodium current and is associated with long-QT syndrome. Circulation 2006, 114, 2104–2112. [Google Scholar] [CrossRef] [Green Version]
  17. Cronk, L.B.; Ye, B.; Kaku, T.; Tester, D.J.; Vatta, M.; Makielski, J.C.; Ackerman, M.J. Novel mechanism for sudden infant death syndrome: Persistent late sodium current secondary to mutations in caveolin-3. Hear. Rhythm 2007, 4, 161–166. [Google Scholar] [CrossRef] [Green Version]
  18. .Catteruccia, M.; Sanna, T.; Santorelli, F.M.; Tessa, A.; Di Giacopo, R.; Sauchelli, D.; Verbo, A.; Monaco, M.L.; Servidei, S. Rippling muscle disease and cardiomyopathy associated with a mutation in the CAV3 gene. Neuromuscul. Disord. 2009, 19, 779–783. [Google Scholar] [CrossRef]
  19. Traverso, M.; Gazzerro, E.; Assereto, S.; Sotgia, F.; Biancheri, R.; Stringara, S.; Giberti, L.; Pedemonte, M.; Wang, X.; Scapolan, S.; et al. Caveolin-3 T78M and T78K missense mutations lead to different phenotypes in vivo and in vitro. Lab. Investig. 2008, 88, 275–283. [Google Scholar] [CrossRef] [Green Version]
  20. Hayashi, T.; Arimura, T.; Ueda, K.; Shibata, H.; Hohda, S.; Takahashi, M.; Hori, H.; Koga, Y.; Oka, N.; Imaizumi, T.; et al. Identification and functional analysis of a caveolin-3 mutation associated with familial hypertrophic cardiomyopathy. Biochem. Biophys. Res. Commun. 2004, 313, 178–184. [Google Scholar] [CrossRef]
  21. Ohsawa, Y.; Tokoet, H.; Katsura, M.; Morimoto, K.; Yamada, H.; Ichikawa, Y.; Murakami, T.; Ohkuma, S.; Komuro, I.; Sunada, Y. Overexpression of P104L mutant caveolin-3 in mice develops hypertrophic cardiomyopathy with enhanced contractility in association with increased endothelial nitric oxide synthase activity. Hum. Mol. Genet. 2004, 13, 151–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Bruno, G.; Puoti, G.; Oliva, M.; Colavito, D.; Allegorico, L.; Napolitano, F.; Sampaoloet, S. A novel missense mutation in CAV3 gene in an Italian family with persistent hyperCKemia, myalgia and hypercholesterolemia: Double-trouble. Clin. Neurol. Neurosurg. 2020, 191. [Google Scholar] [CrossRef] [PubMed]
  23. Gal, D.B.; Wojciak, J.; Perera, J.; Tanel, R.E.; Patel, A.R. Atrial standstill in a pediatric patient with associated caveolin-3 mutation. Hear. Case Rep. 2017, 3, 513–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Head, B.P.; Patel, H.H.; Roth, D.M.; Lai, N.C.; Niesman, I.R.; Farquhar, M.G.; Insel, P.A. G-protein-coupled receptor signaling components localize in both sarcolemmal and intracellular caveolin-3-associated microdomains in adult cardiac myocytes. J. Biol. Chem. 2005, 280, 31036–31044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Lariccia, V.; Nasti, A.A.; Alessandrini, F.; Pesaresi, M.; Gratteri, S.; Tagliabracci, A.; Amoroso, S. Identification and functional analysis of a new putative caveolin-3 variant found in a patient with sudden unexplained death. J. Biomed. Sci. 2014, 21, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Ackerman, M.J.; Siu, B.L.; Sturner, W.Q.; Tester, D.J.; Valdivia, C.R.; Makielski, J.C.; Towbin, J.A. Postmortem molecular analysis of SCN5A defects in sudden infant death syndrome. JAMA 2001, 286, 2264–2269. [Google Scholar] [CrossRef] [PubMed]
  27. Ahern, G.P.; Hsu, S.F.; Klyachko, V.A.; Jackson, M.B. Induction of persistent sodium current by exogenous and endogenous nitric oxide. J. Biol. Chem. 2000, 275, 28810–28815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Venema, V.J.; Ju, H.; Zou, R.; Venema, R.C. Interaction of neuronal nitric-oxide synthase with caveolin-3 in skeletal muscle. Identification of a novel caveolin scaffolding/inhibitory domain. J. Biol. Chem. 1997, 272, 28187–28190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Tan, B.H.; Pundi, K.N.; Van Norstrand, D.W.; Valdivia, C.R.; Tester, D.J.; Medeiros-Domingo, A.; Makielski, J.C.; Ackerman, M.J. Sudden infant death syndrome-associated mutations in the sodium channel beta subunits. Heart Rhythm 2010, 7, 771–778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Cheng, J.; Valdivia, C.R.; Vaidyanathan, R.; Balijepalli, R.C.; Ackerman, M.J.; Makielski, J.C. Caveolin-3 suppresses late sodium current by inhibiting nNOS-dependent S-nitrosylation of SCN5A. J. Mol. Cell. Cardiol. 2013, 61, 102–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Tyan, L.; Foell, J.D.; Vincent, K.P.; Woon, M.T.; Mesquitta, W.T.; Lang, D.; Best, J.M.; Ackerman, M.J.; McCulloch, A.D.; Glukhov, A.V.; et al. Long QT syndrome caveolin-3 mutations differentially modulate K(v) 4 and Ca(v) 1.2 channels to contribute to action potential prolongation. J. Physiol. 2019, 597, 1531–1551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Palygin, O.A.; Pettus, J.M.; Shibata, E.F. Regulation of caveolar cardiac sodium current by a single Gsalpha histidine residue. Am. J. Physiology. Heart Circ. Physiol. 2008, 294, H1693–H1699. [Google Scholar] [CrossRef] [PubMed]
  33. Miake, J.; Marbán, E.; Nuss, H.B. Functional role of inward rectifier current in heart probed by Kir2.1 overexpression and dominant-negative suppression. J. Clin. Investig. 2003, 111, 1529–1536. [Google Scholar] [CrossRef]
  34. Liu, G.X.; Derst, C.; Schlichthörl, G.; Heinen, S.; Seebohm, G.; Brüggemann, A.; Kummer, W.; Veh, R.W.; Daut, J.; Preisig-Müller, R. Comparison of cloned Kir2 channels with native inward rectifier K+ channels from guinea-pig cardiomyocytes. J. Physiol. 2001, 532, 115–126. [Google Scholar] [CrossRef] [PubMed]
  35. Anumonwo, J.M.; Lopatin, A.N. Cardiac strong inward rectifier potassium channels. J. Mol. Cell. Cardiol. 2010, 48, 45–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Vaidyanathan, R.; Van Ert, H.; Haq, K.T.; Morotti, S.; Esch, S.; McCune, E.C.; Grandi, E.; Eckhardt, L.L. Inward Rectifier Potassium Channels (Kir2.x) and Caveolin-3 Domain-Specific Interaction: Implications for Purkinje Cell-Dependent Ventricular Arrhythmias. Circ. Arrhythmia Electrophysiol. 2018, 11, e005800. [Google Scholar] [CrossRef] [PubMed]
  37. Vaidyanathan, R.; Vega, A.L.; Song, C.; Zhou, Q.; Tan, B.H.; Berger, S.; Makielski, J.C.; Eckhardt, L.L. The interaction of caveolin 3 protein with the potassium inward rectifier channel Kir2.1: Physiology and pathology related to long qt syndrome 9 (LQT9). J. Biol. Chem. 2013, 288, 17472–17480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Chang, Y.; Yu, T.; Yang, H.; Peng, Z. Exhaustive exercise-induced cardiac conduction system injury and changes of cTnT and Cx43. Int. J. Sports Med. 2015, 36, 1–8. [Google Scholar] [CrossRef] [PubMed]
  39. Vaidyanathan, R.; Reilly, L.; Eckhardt, L.L. Caveolin-3 Microdomain: Arrhythmia Implications for Potassium Inward Rectifier and Cardiac Sodium Channel. Front. Physiol. 2018, 9, 1548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Ponce-Balbuena, D.; Guerrero-Serna, G.; Valdivia, C.R.; Caballero, R.; Diez-Guerra, F.J.; Jiménez-Vázquez, E.N.; Ramírez, R.J.; Monteiro da Rocha, A.; Herron, T.J.; Campbell, K.F.; et al. Cardiac Kir2.1 and Na(V)1.5 Channels Traffic Together to the Sarcolemma to Control Excitability. Circ. Res. 2018, 122, 1501–1516. [Google Scholar] [CrossRef] [PubMed]
  41. Nguyen Dinh Cat, A.; Touyz, R.M. A new look at the renin-angiotensin system--focusing on the vascular system. Peptides 2011, 32, 2141–2150. [Google Scholar] [CrossRef]
  42. Tyan, L.; Turner, D.; Komp, K.R.; Medvedev, R.Y.; Lim, E.; Glukhov, A.V. Caveolin-3 is required for regulation of transient outward potassium current by angiotensin II in mouse atrial myocytes. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H787–H797. [Google Scholar] [CrossRef] [PubMed]
  43. Rogers, T.B.; Lokuta, A.J. Angiotensin II signal transduction pathways in the cardiovascular system. Trends Cardiovasc. Med. 1994, 4, 110–116. [Google Scholar] [CrossRef]
  44. Scholz, E.P.; Welke, F.; Joss, N.; Seyler, C.; Zhang, W.; Scherer, D.; Völkers, M.; Bloehs, R.; Thomas, D.; Katus, H.A.; et al. Central role of PKCα in isoenzyme-selective regulation of cardiac transient outward current Ito and Kv4.3 channels. J. Mol. Cell. Cardiol. 2011, 51, 722–729. [Google Scholar] [CrossRef] [PubMed]
  45. Jansen, H.J.; Mackasey, M.; Moghtadaei, M.; Belke, D.D.; Egom, E.E.; Tuomi, J.M.; Rafferty, S.A.; Kirkby, A.W.; Rose, R.A. Distinct patterns of atrial electrical and structural remodeling in angiotensin II mediated atrial fibrillation. J. Mol. Cell. Cardiol. 2018, 124, 12–25. [Google Scholar] [CrossRef] [PubMed]
  46. Kawai, M.; Hussain, M.; Orchard, C.H. Excitation-contraction coupling in rat ventricular myocytes after formamide-induced detubulation. Am. J. Physiol. 1999, 277, H603–H609. [Google Scholar] [CrossRef]
  47. Shan, J.; Xie, W.; Betzenhauser, M.; Reiken, S.; Chen, B.X.; Wronska, A.; Marks, A.R. Calcium leak through ryanodine receptors leads to atrial fibrillation in 3 mouse models of catecholaminergic polymorphic ventricular tachycardia. Circ. Res. 2012, 111, 708–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Nikolaev, V.O.; Moshkov, A.; Lyon, A.R.; Miragoli, M.; Novak, P.; Paur, H.; Lohse, M.J.; Korchev, Y.E.; Harding, S.E.; Gorelik, J. Beta2-adrenergic receptor redistribution in heart failure changes cAMP compartmentation. Science 2010, 327, 1653–1657. [Google Scholar] [CrossRef] [PubMed]
  49. Beuckelmann, D.J.; Näbauer, M.; Erdmann, E. Characteristics of calcium-current in isolated human ventricular myocytes from patients with terminal heart failure. J. Mol. Cell. Cardiol. 1991, 23, 929–937. [Google Scholar] [CrossRef]
  50. Ouadid, H.; Séguin, J.; Richard, S.; Chaptal, P.A.; Nargeot, J. Properties and Modulation of Ca channels in adult human atrial cells. J. Mol. Cell. Cardiol. 1991, 23, 41–54. [Google Scholar] [CrossRef]
  51. Li, G.R.; Nattel, S. Properties of human atrial ICa at physiological temperatures and relevance to action potential. Am. J. Physiol. 1997, 272, H227–H235. [Google Scholar] [CrossRef] [PubMed]
  52. Markandeya, Y.S.; Fahey, J.M.; Pluteanu, F.; Cribbs, L.L.; Balijepalli, R.C. Caveolin-3 regulates protein kinase A modulation of the Ca(V)3.2 (alpha1H) T-type Ca2+ channels. J. Biol. Chem. 2011, 286, 2433–2444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Markandeya, Y.S.; Phelan, L.J.; Woon, M.T.; Keefe, A.M.; Reynolds, C.R.; August, B.K.; Hacker, T.A.; Roth, D.M.; Patel, H.H.; Balijepalli, R.C. Caveolin-3 Overexpression Attenuates Cardiac Hypertrophy via Inhibition of T-type Ca2+ Current Modulated by Protein Kinase Cα in Cardiomyocytes. J. Biol. Chem. 2015, 290, 22085–22100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Chemin, J.; Taiakina, V.; Monteil, A.; Piazza, M.; Guan, W.; Stephens, R.F.; Kitmitto, A.; Pang, Z.P.; Dolphin, A.C.; Perez-Reyes, E.; et al. Calmodulin regulates Ca(v)3 T-type channels at their gating brake. J. Biol. Chem. 2017, 292, 20010–20031. [Google Scholar] [CrossRef] [Green Version]
  55. Parton, R.G.; Way, M.; Zorzi, N.; Stang, E. Caveolin-3 associates with developing T-tubules during muscle differentiation. J. Cell Biol. 1997, 136, 137–154. [Google Scholar] [CrossRef] [PubMed]
  56. Bryant, S.; Kimura, T.E.; Kong, C.H.; Watson, J.J.; Chase, A.; Suleiman, M.S.; James, A.F.; Orchard, C.H. Stimulation of ICa by basal PKA activity is facilitated by Caveolin-3 in cardiac ventricular myocytes. J. Mol. Cell. Cardiol. 2014, 68, 47–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Galbiati, F.; Engelman, J.A.; Volonte, D.; Zhang, X.L.; Minetti, C.; Li, M.; Hou, H., Jr.; Kneitz, B.; Edelmann, W.; Lisanti, M.P. Caveolin-3 null mice show a loss of caveolae, changes in the microdomain distribution of the dystrophin-glycoprotein complex, and t-tubule abnormalities. J. Biol. Chem. 2001, 276, 21425–21433. [Google Scholar] [CrossRef] [Green Version]
  58. Bryant, S.M.; Kong, C.H.T.; Watson, J.J.; Gadeberg, H.C.; James, A.F.; Cannell, M.B.; Orchard, C.H. Caveolin 3-dependent loss of t-tubular I(Ca) during hypertrophy and heart failure in mice. Exp. Physiol. 2018, 103, 652–665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Bhogal, N.K.; Hasan, A.; Gorelik, J. The Development of Compartmentation of cAMP Signaling in Cardiomyocytes: The Role of T-Tubules and Caveolae Microdomains. J. Cardiovasc. Dev. Dis. 2018, 5, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Asghari, P.; Scriven, D.R.; Hoskins, J.; Fameli, N.; van Breemen, C.; Moore, E.D. The structure and functioning of the couplon in the mammalian cardiomyocyte. Protoplasma 2012, 249 (Suppl. 1), S31–S38. [Google Scholar] [CrossRef] [PubMed]
  61. Fill, M.; Copello, J.A. Ryanodine receptor calcium release channels. Physiol. Rev. 2002, 82, 893–922. [Google Scholar] [CrossRef] [Green Version]
  62. Wong, J.; Baddeley, D.; Bushong, E.A.; Yu, Z.; Ellisman, M.H.; Hoshijima, M.; Soeller, C. Nanoscale distribution of ryanodine receptors and caveolin-3 in mouse ventricular myocytes: Dilation of t-tubules near junctions. Biophys. J. 2013, 104, L22-24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Song, D.W.; Lee, K.E.; Ryu, J.Y.; Jeon, H.; Kim, D.H. The molecular interaction of heart LIM protein (HLP) with RyR2 and caveolin-3 is essential for Ca(2+)-induced Ca(2+) release in the heart. Biochem. Biophys. Res. Commun. 2015, 463, 975–981. [Google Scholar] [CrossRef] [PubMed]
  64. Zhang, Z.; Fang, Q.; Du, T.; Chen, G.; Wang, Y.; Wang, D.W. Cardiac-Specific Caveolin-3 Overexpression Prevents Post-Myocardial Infarction Ventricular Arrhythmias by Inhibiting Ryanodine Receptor-2 Hyperphosphorylation. Cardiology 2020, 145, 136–147. [Google Scholar] [CrossRef]
  65. Castets, F.; Rakitina, T.; Gaillard, S.; Moqrich, A.; Mattei, M.G.; Monneron, A. Zinedin, SG2NA, and striatin are calmodulin-binding, WD repeat proteins principally expressed in the brain. J. Biol. Chem. 2000, 275, 19970–19977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Nader, M.; Alotaibi, S.; Alsolme, E.; Khalil, B.; Abu-Zaid, A.; Alsomali, R.; Bakheet, D.; Dzimiri, N. Cardiac striatin interacts with caveolin-3 and calmodulin in a calcium sensitive manner and regulates cardiomyocyte spontaneous contraction rate. Can. J. Physiol. Pharmacol. 2017, 95, 1306–1312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Barbuti, A.; Terragni, B.; Brioschi, C.; DiFrancesco, D. Localization of f-channels to caveolae mediates specific beta2-adrenergic receptor modulation of rate in sinoatrial myocytes. J. Mol. Cell. Cardiol. 2007, 42, 71–78. [Google Scholar] [CrossRef]
  68. Motloch, L.J.; Larbig, R.; Darabi, T.; Reda, S.; Motloch, K.A.; Wernly, B.; Lichtenauer, M.; Gebing, T.; Schwaiger, A.; Zagidullin, N.; et al. Long-QT syndrome-associated Caveolin-3 mutations differentially regulate the hyperpolarization-activated cyclic nucleotide gated channel 4. Physiol. Int. 2017, 104, 130–138. [Google Scholar] [CrossRef]
  69. Kuwabara, Y.; Kuwahara, K.; Takano, M.; Kinoshita, H.; Arai, Y.; Yasuno, S.; Nakagawa, Y.; Igata, S.; Usami, S.; Minami, T.; et al. Increased expression of HCN channels in the ventricular myocardium contributes to enhanced arrhythmicity in mouse failing hearts. J. Am. Heart Assoc. 2013, 2, e000150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Sun, L.Y.; Qu, X.; Chen, L.Z.; Zheng, G.S.; Wu, X.L.; Chen, X.X.; Huang, W.J.; Zhou, H. Potential Roles of Serum Caveolin-3 Levels in Patients with Atrial Fibrillation. Front. Aging Neurosci. 2017, 9, 90. [Google Scholar] [CrossRef]
  71. Reilly, S.N.; Liu, X.; Carnicer, R.; Recalde, A.; Muszkiewicz, A.; Jayaram, R.; Carena, M.C.; Wijesurendra, R.; Stefanini, M.; Surdo, N.C.; et al. Up-regulation of miR-31 in human atrial fibrillation begets the arrhythmia by depleting dystrophin and neuronal nitric oxide synthase. Sci. Transl. Med. 2016, 8, 340ra374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Egorov, Y.V.; Lang, D.; Tyan, L.; Turner, D.; Lim, E.; Piro, Z.D.; Hernandez, J.J.; Lodin, R.; Wang, R.; Schmuck, E.G.; et al. Caveolae-Mediated Activation of Mechanosensitive Chloride Channels in Pulmonary Veins Triggers Atrial Arrhythmogenesis. J. Am. Heart Assoc. 2019, 8, e012748. [Google Scholar] [CrossRef] [PubMed]
  73. Andelova, K.; Egan Benova, T.; Szeiffova Bacova, B.; Sykora, M.; Prado, N.J.; Diez, E.R.; Hlivak, P.; Tribulova, N. Cardiac Connexin-43 Hemichannels and Pannexin1 Channels: Provocative Antiarrhythmic Targets. Int. J. Mol. Sci. 2020, 22, 260. [Google Scholar] [CrossRef] [PubMed]
  74. Xue, J.; Yan, X.; Yang, Y.; Chen, M.; Wu, L.; Gou, Z.; Sun, Z.; Talabieke, S.; Zheng, Y.; Luo, D. Connexin 43 dephosphorylation contributes to arrhythmias and cardiomyocyte apoptosis in ischemia/reperfusion hearts. Basic Res. Cardiol. 2019, 114, 40. [Google Scholar] [CrossRef]
  75. Saffitz, J.E. Arrhythmogenic cardiomyopathy and abnormalities of cell-to-cell coupling. Heart Rhythm 2009, 6, S62–S65. [Google Scholar] [CrossRef] [PubMed]
  76. Xiao, S.; Shimura, D.; Baum, R.; Hernandez, D.M.; Agvanian, S.; Nagaoka, Y.; Katsumata, M.; Lampe, P.D.; Kleber, A.G.; Hong, T.; et al. Auxiliary trafficking subunit GJA1-20k protects connexin-43 from degradation and limits ventricular arrhythmias. J. Clin. Investig. 2020, 130, 4858–4870. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, P.; Xu, J.; Hu, W.; Yu, D.; Bai, X. Effects of Pinocembrin Pretreatment on Connexin 43 (Cx43) Protein Expression After Rat Myocardial Ischemia-Reperfusion and Cardiac Arrhythmia. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2018, 24, 5008–5014. [Google Scholar] [CrossRef]
  78. Rugowska, A.; Wiernicki, B.; Maczewski, M.; Mackiewicz, U.; Chojnacka, K.; Bednarek-Rajewska, K.; Kluk, A.; Majewski, P.; Kolanowski, T.; Malcher, A.; et al. Human skeletal muscle-derived stem/progenitor cells modified with connexin-43 prevent arrhythmia in rat post-infarction hearts and influence gene expression in the myocardium. J. Physiol. Pharmacol. Off. J. Pol. Physiol. Soc. 2019, 70. [Google Scholar] [CrossRef]
  79. Yang, K.C.; Rutledge, C.A.; Mao, M.; Bakhshi, F.R.; Xie, A.; Liu, H.; Bonini, M.G.; Patel, H.H.; Minshall, R.D.; Dudley, S.C., Jr. Caveolin-1 modulates cardiac gap junction homeostasis and arrhythmogenecity by regulating cSrc tyrosine kinase. Circ. Arrhythmia Electrophysiol. 2014, 7, 701–710. [Google Scholar] [CrossRef] [Green Version]
  80. Liu, L.; Li, Y.; Lin, J.; Liang, Q.; Sheng, X.; Wu, J.; Huang, R.; Liu, S.; Li, Y. Connexin43 interacts with Caveolin-3 in the heart. Mol. Biol. Rep. 2010, 37, 1685–1691. [Google Scholar] [CrossRef] [PubMed]
  81. Soares, E.S.; Mendonça, M.C.; Rocha, T.; Kalapothakis, E.; da Cruz-Höfling, M.A. Are Synchronized Changes in Connexin-43 and Caveolin-3 a Bystander Effect in a Phoneutria nigriventer Venom Model of Blood-Brain Barrier Breakdown? J. Mol. Neurosci. MN 2016, 59, 452–463. [Google Scholar] [CrossRef] [PubMed]
  82. Liao, C.K.; Wang, S.M.; Chen, Y.L.; Wang, H.S.; Wu, J.C. Lipopolysaccharide-induced inhibition of connexin43 gap junction communication in astrocytes is mediated by downregulation of caveolin-3. Int. J. Biochem. Cell Biol. 2010, 42, 762–770. [Google Scholar] [CrossRef]
  83. Valera, I.C.; Wacker, A.L.; Hwang, H.S.; Holmes, C.; Laitano, O.; Landstrom, A.P.; Parvatiyar, M.S. Essential roles of the dystrophin-glycoprotein complex in different cardiac pathologies. Adv. Med. Sci. 2021, 66, 52–71. [Google Scholar] [CrossRef] [PubMed]
  84. Sinagra, G.; Dal Ferro, M.; Gigli, M. The heart of dystrophinopathies. Eur. J. Heart Fail. 2021, 23, 1287–1289. [Google Scholar] [CrossRef]
  85. Kamdar, F.; Garry, D.J. Dystrophin-Deficient Cardiomyopathy. J. Am. Coll. Cardiol. 2016, 67, 2533–2546. [Google Scholar] [CrossRef] [PubMed]
  86. Doyle, D.D.; Goings, G.; Upshaw-Earley, J.; Ambler, S.K.; Mondul, A.; Palfrey, H.C.; Page, E. Dystrophin associates with caveolae of rat cardiac myocytes: Relationship to dystroglycan. Circ. Res. 2000, 87, 480–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Aravamudan, B.; Volonte, D.; Ramani, R.; Gursoy, E.; Lisanti, M.P.; London, B.; Galbiati, F. Transgenic overexpression of caveolin-3 in the heart induces a cardiomyopathic phenotype. Hum. Mol. Genet. 2003, 12, 2777–2788. [Google Scholar] [CrossRef] [Green Version]
  88. Sotgia, F.; Lee, J.K.; Das, K.; Bedford, M.; Petrucci, T.C.; Macioce, P.; Sargiacomo, M.; Bricarelli, F.D.; Minetti, C.; Sudol, M.; et al. Caveolin-3 directly interacts with the C-terminal tail of beta -dystroglycan. Identification of a central WW-like domain within caveolin family members. J. Biol. Chem. 2000, 275, 38048–38058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Fang, H.; Judd, R.L. Adiponectin Regulation and Function. Compr. Physiol. 2018, 8, 1031–1063. [Google Scholar] [CrossRef] [PubMed]
  90. Wang, Y.; Wang, X.; Jasmin, J.F.; Lau, W.B.; Li, R.; Yuan, Y.; Yi, W.; Chuprun, K.; Lisanti, M.P.; Koch, W.J.; et al. Essential role of caveolin-3 in adiponectin signalsome formation and adiponectin cardioprotection. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 934–942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Yi, W.; Sun, Y.; Gao, E.; Wei, X.; Lau, W.B.; Zheng, Q.; Wang, Y.; Yuan, Y.; Wang, X.; Tao, L.; et al. Reduced cardioprotective action of adiponectin in high-fat diet-induced type II diabetic mice and its underlying mechanisms. Antioxid. Redox Signal. 2011, 15, 1779–1788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Lei, S.; Li, H.; Xu, J.; Liu, Y.; Gao, X.; Wang, J.; Ng, K.F.; Lau, W.B.; Ma, X.L.; Rodrigues, B.; et al. Hyperglycemia-induced protein kinase C β2 activation induces diastolic cardiac dysfunction in diabetic rats by impairing caveolin-3 expression and Akt/eNOS signaling. Diabetes 2013, 62, 2318–2328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Tan, Z.; Zhou, L.J.; Mu, P.W.; Liu, S.P.; Chen, S.J.; Fu, X.D.; Wang, T.H. Caveolin-3 is involved in the protection of resveratrol against high-fat-diet-induced insulin resistance by promoting GLUT4 translocation to the plasma membrane in skeletal muscle of ovariectomized rats. J. Nutr. Biochem. 2012, 23, 1716–1724. [Google Scholar] [CrossRef] [PubMed]
  94. Li, H.; Yao, W.; Liu, Z.; Xu, A.; Huang, Y.; Ma, X.L.; Irwin, M.G.; Xia, Z. Hyperglycemia Abrogates Ischemic Postconditioning Cardioprotection by Impairing AdipoR1/Caveolin-3/STAT3 Signaling in Diabetic Rats. Diabetes 2016, 65, 942–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Hegyi, B.; Bers, D.M.; Bossuyt, J. CaMKII signaling in heart diseases: Emerging role in diabetic cardiomyopathy. J. Mol. Cell. Cardiol. 2019, 127, 246–259. [Google Scholar] [CrossRef]
  96. Lau, D.H.; Nattel, S.; Kalman, J.M.; Sanders, P. Modifiable Risk Factors and Atrial Fibrillation. Circulation 2017, 136, 583–596. [Google Scholar] [CrossRef]
  97. Huang, Y.; Deng, Y.; Shang, L.; Yang, L.; Huang, J.; Ma, J.; Liao, X.; Zhou, H.; Xian, J.; Liang, G.; et al. Effect of type 2 diabetes mellitus caveolin-3 K15N mutation on glycometabolism. Exp. Ther. Med. 2019, 18, 2531–2539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Shang, L.; Chen, T.; Xian, J.; Deng, Y.; Huang, Y.; Zhao, Q.; Liang, G.; Liang, Z.; Lian, F.; Wei, H.; et al. The caveolin-3 P104L mutation in LGMD-1C patients inhibits non-insulin-stimulated glucose metabolism and growth but promotes myocyte proliferation. Cell Biol. Int. 2019, 43, 669–677. [Google Scholar] [CrossRef] [PubMed]
  99. Gao, Y.; Zhang, M.; Wu, T.; Xu, M.; Cai, H.; Zhang, Z. Effects of D-Pinitol on Insulin Resistance through the PI3K/Akt Signaling Pathway in Type 2 Diabetes Mellitus Rats. J. Agric. Food Chem. 2015, 63, 6019–6026. [Google Scholar] [CrossRef]
  100. Yang, M.; Ren, Y.; Lin, Z.; Tang, C.; Jia, Y.; Lai, Y.; Zhou, T.; Wu, S.; Liu, H.; Yang, G.; et al. Krüppel-like factor 14 increases insulin sensitivity through activation of PI3K/Akt signal pathway. Cell. Signal. 2015, 27, 2201–2208. [Google Scholar] [CrossRef]
  101. Mu, P.; Tan, Z.; Cui, Y.; Liu, H.; Xu, X.; Huang, Q.; Zeng, L.; Wang, T. 17β-Estradiol attenuates diet-induced insulin resistance and glucose intolerance through up-regulation of caveolin-3. Ir. J. Med. Sci. 2011, 180, 221–227. [Google Scholar] [CrossRef] [PubMed]
  102. Lei, S.; Su, W.; Xia, Z.Y.; Wang, Y.; Zhou, L.; Qiao, S.; Zhao, B.; Xia, Z.; Irwin, M.G. Hyperglycemia-Induced Oxidative Stress Abrogates Remifentanil Preconditioning-Mediated Cardioprotection in Diabetic Rats by Impairing Caveolin-3-Modulated PI3K/Akt and JAK2/STAT3 Signaling. Oxidative Med. Cell. Longev. 2019, 2019, 9836302. [Google Scholar] [CrossRef] [PubMed]
  103. Margolis, K.L.; Bonds, D.E.; Rodabough, R.J.; Tinker, L.; Phillips, L.S.; Allen, C.; Bassford, T.; Burke, G.; Torrens, J.; Howard, B.V. Effect of oestrogen plus progestin on the incidence of diabetes in postmenopausal women: Results from the Women’s Health Initiative Hormone Trial. Diabetologia 2004, 47, 1175–1187. [Google Scholar] [CrossRef] [PubMed]
  104. Capozza, F.; Combs, T.P.; Cohen, A.W.; Cho, Y.R.; Park, S.Y.; Schubert, W.; Williams, T.M.; Brasaemle, D.L.; Jelicks, L.A.; Scherer, P.E.; et al. Caveolin-3 knockout mice show increased adiposity and whole body insulin resistance, with ligand-induced insulin receptor instability in skeletal muscle. Am. J. Physiology. Cell Physiol. 2005, 288, C1317–C1331. [Google Scholar] [CrossRef] [PubMed]
  105. Lin, Y.K.; Chen, Y.C.; Chen, J.H.; Chen, S.A.; Chen, Y.J. Adipocytes modulate the electrophysiology of atrial myocytes: Implications in obesity-induced atrial fibrillation. Basic Res. Cardiol. 2012, 107, 293. [Google Scholar] [CrossRef] [PubMed]
  106. Omran, J.; Bostick, B.P.; Chan, A.K.; Alpert, M.A. Obesity and Ventricular Repolarization: A Comprehensive Review. Prog. Cardiovasc. Dis. 2018, 61, 124–135. [Google Scholar] [CrossRef] [PubMed]
  107. Gómez-Ruiz, A.; Milagro, F.I.; Campión, J.; Martínez, J.A.; de Miguel, C. Caveolin expression and activation in retroperitoneal and subcutaneous adipocytes: Influence of a high-fat diet. J. Cell. Physiol. 2010, 225, 206–213. [Google Scholar] [CrossRef] [PubMed]
  108. Gómez-Ruiz, A.; Milagro, F.I.; Campión, J.; Martínez, J.A.; de Miguel, C. High-fat diet feeding alters metabolic response to fasting/non fasting conditions. Effect on caveolin expression and insulin signalling. Lipids Health Dis. 2011, 10, 55. [Google Scholar] [CrossRef] [Green Version]
  109. Feillet-Coudray, C.; Sutra, T.; Fouret, G.; Ramos, J.; Wrutniak-Cabello, C.; Cabello, G.; Cristol, J.P.; Coudray, C. Oxidative stress in rats fed a high-fat high-sucrose diet and preventive effect of polyphenols: Involvement of mitochondrial and NAD(P)H oxidase systems. Free Radic. Biol. Med. 2009, 46, 624–632. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cartoon illustration of arrhythmia mechanism caused by Cav3 mutation (F97C). NOS, Nitric oxide synthase; NO, Nitric oxide.
Figure 1. Cartoon illustration of arrhythmia mechanism caused by Cav3 mutation (F97C). NOS, Nitric oxide synthase; NO, Nitric oxide.
Jcm 11 01595 g001
Table 1. Pathogenic CAV3 mutation associated with cardiovascular diseases.
Table 1. Pathogenic CAV3 mutation associated with cardiovascular diseases.
PhenotypeCAV3 MutationSerum CK ConcentrationsRef.
LQTSp.A85TNAVatta, M. et al. [16]
Sudden infant death syndromep.V14 LNACronk, L.B. et al. [17]
Dilated cardiomyopathyp.A46VHighCatteruccia, M. et al. [18]
Dilated cardiomyopathyp.T78MHighTraverso, M. et al. [19]
Hypertrophic cardiomyopathyp.T63SNormalHayashi, T. et al. [20]
Hypertrophic cardiomyopathyP104LNAOhsawa, Y. et al. [21]
Hypercholesterolemiap. Val44MetHighBruno, G. et al. [22]
Atrial standstillp. Leu84ProNAGal, D. B. et al. [23]
NA, Not Available; LQTS, Long QT syndrome.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

He, M.; Qiu, J.; Wang, Y.; Bai, Y.; Chen, G. Caveolin-3 and Arrhythmias: Insights into the Molecular Mechanisms. J. Clin. Med. 2022, 11, 1595. https://doi.org/10.3390/jcm11061595

AMA Style

He M, Qiu J, Wang Y, Bai Y, Chen G. Caveolin-3 and Arrhythmias: Insights into the Molecular Mechanisms. Journal of Clinical Medicine. 2022; 11(6):1595. https://doi.org/10.3390/jcm11061595

Chicago/Turabian Style

He, Miaomiao, Jie Qiu, Yan Wang, Yang Bai, and Guangzhi Chen. 2022. "Caveolin-3 and Arrhythmias: Insights into the Molecular Mechanisms" Journal of Clinical Medicine 11, no. 6: 1595. https://doi.org/10.3390/jcm11061595

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