Next Article in Journal
Stem Cell Therapies in Canine Cardiology: Comparative Efficacy, Emerging Trends, and Clinical Integration
Previous Article in Journal
IgG Biomarkers in Multiple Sclerosis: Deciphering Their Puzzling Protein A Connection
Previous Article in Special Issue
Casein Kinase 1 Phosphomimetic Mutations Negatively Impact Connexin-43 Gap Junctions in Human Pluripotent Stem Cell-Derived Cardiomyocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Overexpression of Cx43: Is It an Effective Approach for the Treatment of Cardiovascular Diseases?

1
Institute of Physiology, Justus-Liebig University, 35392 Giessen, Germany
2
Department of Clinical and Biological Sciences, University of Torino, 10125 Torino, Italy
3
Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3190601, Israel
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(3), 370; https://doi.org/10.3390/biom15030370
Submission received: 7 February 2025 / Revised: 26 February 2025 / Accepted: 28 February 2025 / Published: 4 March 2025
(This article belongs to the Special Issue Gap Junctions and Connexins in Health and Disease, 2nd Edition)

Abstract

:
In the heart, Connexin 43 (Cx43) is involved in intercellular communication through gap junctions and exosomes. In addition, Cx43-formed hemichannels at the plasma membrane are important for ion homeostasis and cellular volume regulation. Through its localization within nuclei and mitochondria, Cx43 influences the function of the respective organelles. Several cardiovascular diseases such as heart failure, ischemia/reperfusion injury, hypertrophic cardiomyopathy and arrhythmias are characterized by Cx43 downregulation and a dysregulated Cx43 function. Accordingly, a putative therapeutic approach of these diseases would include the induction of Cx43 expression in the damaged heart, albeit such induction may have both beneficial and detrimental effects. In this review we discuss the consequences of increasing cardiac Cx43 expression, and discuss this manipulation as a strategy for the treatment of cardiovascular diseases.

1. Localization and Function of Cx43 Within the Healthy Heart

The connexin protein family consists of 20 or 21 members in mice and humans [1,2], and connexins are named according to their approximate molecular weight of 20–62 kDa. In humans, connexin expression is mapped to more than 110 cell types, such as heart, brain and liver [3]. In addition to their wide distribution, connexin expression is highly dynamic, with temporal changes within cells and tissues, and specific combinations of expressed connexin isoforms due to the regulation of transcription and epigenetic mechanisms [4,5]. In the heart, Cx40 is mainly found in atrial cardiomyocytes, and, like Cx45, it is present in the atrioventricular node, the His-bundle and the ventricular conduction system. Cx45 is also localized in the sinoatrial node [6], and Cx30.2 in the conduction system [7]. In the present article, we focus on the expression and function of Cx43, which is encoded by the Gja1 gene and ubiquitously expressed in the heart, as well as in over 50 organs and many cell types, such as astrocytes, retinal cells, endothelial cells and alveolar cells [3,8,9,10,11].
Connexins are typically composed of intracellular N-terminal and C-terminal regions, four transmembrane domains, and two extracellular and one intracellular loops, and are synthesized in the endoplasmic reticulum; six connexins oligomerize into a connexon or hemichannel in the Golgi or trans-Golgi network, and traffic in vesicles along the microtubules to the plasma membrane [12,13]. Docking of two hemichannels from neighboring cells results in the formation of gap junctions, which mediate intercellular electrical and chemical cell–cell coupling [14].
Six identical connexins oligomerize into a homomeric hemichannel or connexon. Connexons consisting of two or more connexin isoforms are called heteromeric. Docking of hemichannels from adjacent cells results in the formation of homotypic (composed of two identical connexons), heterotypic (most typically composed of two different homomeric connexons), or heteromeric (composed of two different heteromeric connexons) connexin channels [9,15]. Accordingly, the different connexin channels display specific unitary conductance, charge selectivity and pore diameters [16,17]. In addition to the originally described function of connexins as gap junction proteins [14,18], recent studies suggest that connexins have additional functions. For example, Cx43 is detected within extracellular vesicles of different cellular origin [19,20,21,22], such as H9C2 cells [20], and phosphorylated Cx43 controls exosome release [23]. Thus, Cx43 not only contributes to the communication of neighboring cells, but also to that of cells located at greater distances. Non-paired hemichannels exist in the plasma membrane and, when open, allow communication with the extracellular space, including transporting molecules with a molecular weight of up to 1.2 kDa [24]. Whereas such hemichannels are predominantly closed under physiological conditions, they open under certain pathophysiological situations, such as in mdx mice (having a point mutation in the Dmd gene; a common mouse model of Duchenne muscular dystrophy), right ventricular arrhythmogenic cardiomyopathy in Plakophilin-2-(Pkp2)-deficient mice, and in ischemia/reperfusion (I/R) injury in mice [25,26,27]. Another subcellular location where Cx43 is detected are the mitochondria. However, cardiac Cx43 is not localized within all mitochondria, but is mainly present in subsarcolemmal mitochondria and not in interfibrillar mitochondria [28,29,30,31]. The subsarcolemmal mitochondria are located beneath the sarcolemma and differ from interfibrillar mitochondria in form and function [32,33]. The analysis of subsarcolemmal mitochondria shows that Cx43 is present in the inner mitochondrial membrane [34,35]. Regarding the orientation of Cx43 within the inner membrane, it is suggested that the C-terminus is facing the intermembrane space [28,36]. Within mitochondria, Cx43 regulates several functions, including respiration [37], potassium uptake [38], formation of reactive oxygen species [39] and opening of the mitochondrial permeability transition pore [35,40]. Recent data show that in addition to mitochondria, Cx43 is also found in nuclei of rat and mouse cardiomyocytes [41], as well as in nuclei of colon samples from patients with ulcerative colitis [42]. Whereas the pathways by which Cx43 reaches the nuclei are presently unclear, diffusion is excluded. Finally, Cx43 forms channels in the nuclear envelope [41] and influences gene expression in astrocytes [43] and cardiomyocytes [41].
The complexity of Cx43-mediated functions is not only evident in that it has several subcellular localizations, but also by the fact that in addition to the 43 kDa full-length protein, alternative translation generates a truncated 20 kDa form of Cx43 [44]. This N-terminally truncated form, termed Gja21-20k, is detected in different cell types and organs including the heart [44,45,46]. Gja1-20k is found within the Golgi apparatus and mitochondria. Like the full-length Cx43, mitochondrial Gja1-20k localizes to subsarcolemmal mitochondria [31], but unlike the 43kDa form, it is not found within the inner, but within the outer mitochondrial membrane [47]. Mitochondrial Gja1-20k affects their function in several aspects, including respiration, membrane potential and biogenesis [45,47]. Additionally, Gja1-20k influences oligomerization of hexamers, stabilization of actin filaments and the transport of Cx43-formed hexamers to the plasma membrane [48,49]. Moreover, Gja1-20k expression correlates with the intercellular transport of mitochondria from mesenchymal stromal cells to chondrocytes undergoing oxidative stress, a process suggested to contribute to tissue healing in degenerative diseases such as osteoarthritis [50]. The decisive role of Gja1-20k in the maintenance of cardiac function is demonstrated in Gja1-20k-deficient mice, which have reduced protein levels of full-length Cx43, abnormal cardiac conduction and a significantly reduced lifespan [51].
Cx43 function is regulated by post-translational modifications such as glycosylation, N-acetylation, S-nitrosylation, ubiquitination and phosphorylation (for review, see [52]). It is suggested that the cysteine residues in the extracellular loops function as redox sensors, thereby controlling connexin function [53]. Respecting Cx43 phosphorylation, the 21 amino acids (mainly serine and tyrosine residues) targeted by different protein kinases are located in the carboxy-terminus of the protein. Kinases phosphorylating Cx43 include protein kinase B (AKT, targets serine 373), protein kinase C (PKC, targets serine 368), casein kinase 1 (CK1, targets serines 325, 328, 330), mitogen-activated protein kinase (MAPK, targets serines 262, 279, 282) and pp60src kinase (targets tyrosine 247 and 265) (for review, see [54]). Cx43 phosphorylation is important for the regulation of its transport and assembly into gap junctions, gating of Cx43-formed channels and gap junction turnover [3,54]. In addition to the function of gap-junctional Cx43, the activities of hemichannels and mitochondria are also regulated by phosphorylation [25,55,56]. Whether Cx43 phosphorylation is a prerequisite for its import into mitochondria, or whether the protein is phosphorylated within the organelle by mitochondrially localized kinases, is presently unclear [57]. Nuclear Cx43 is also phosphorylated, thereby affecting its activity in the outer nuclear membrane [41]. Whereas the aforementioned studies show that phosphorylation controls Cx43 activity in the subcellular compartments, it is completely unknown whether there is an interplay between the phosphorylation sites in the specific compartments. The analysis of mitochondria isolated from mice with mutated Cx43 phosphorylation sites demonstrates alterations in the phosphorylation status of additional amino acids [55], showing that lack of phosphorylation at one amino acid influences the phosphorylation at other residues. The consequences of such secondary changes in Cx43 phosphorylation are yet to be investigated. Furthermore, it is unclear whether the interactions between the individual Cx43 phosphorylation sites are restricted to the mitochondria or whether they also occur in other compartments where Cx43 is localized. It needs to be investigated if Cx43 is phosphorylated in a specific pattern in the individual compartments, and if a dynamic interplay in the phosphorylation at the specific amino acids occurs in physiological as well as in pathological conditions.
Another approach to control Cx43 function is to adjust the amount of the protein, determined by the fine balance between synthesis and degradation. The half-life of cardiac Cx43 is relatively short (1–2 h) [58,59], implying that gap junctions are renewed several times/day [9] due to transcriptional modulation [60]. In addition, the epigenetic machinery regulates connexin expression via DNA methylation, histone acetylation or micro-RNAs [61,62]. Removal of gap junctions from the plasma membrane involves the internalization of gap junction plaques and the subsequent formation of annular gap junctions, also termed connexosomes [63]. Annular gap junctions are degraded via autophagosomal and endolysosomal pathways, whereas a fraction of newly synthesized Cx43 undergoes proteasomal degradation via ERAD (endoplasmic reticulum-associated degradation) [64,65]. Cx43 degradation is classically triggered by protein ubiquitination [66].
Taken together, knowledge about Cx43 function extends beyond its role in intercellular communication via gap junctions and hemichannels. Cx43 contributes to cell–cell communication also via its presence within exosomes. Furthermore, Cx43 is located within nuclei and mitochondria, and influences the function of the respective organelles. Whereas Cx43 participates in the regulation of cell functions via its specific localization, its own function is also controlled via alternative translation and post-translational modifications. As a protein with a short half-life, an additional aspect to control Cx43 function is transcriptional regulation, protein turnover and degradation. All these aspects highlight the wide range of options used to regulate Cx43 function under physiological and pathological conditions.

2. Cx43: Expression, Localization and Function in the Diseased Myocardium

Considering the essential role of Cx43 in the regulation of cardiac function, it is necessary to understand the changes in Cx43 activity associated with cardiovascular diseases such as hypertension, hypertrophy, myocardial infarction, heart failure and arrhythmias. The present review summarizes only general aspects of Cx43 dysfunction in cardiovascular diseases; for a more detailed description of these changes occurring in specific pathological conditions, please see previously published articles [6,67,68,69,70,71,72,73]. Despite the different pathomechanisms underlying the abovementioned diseases, there are several aspects of Cx43 dysregulation shared by these diseases. In hypertrophic heart disease, early stages are often associated with increased Cx43 expression, whereas decreased expression is reported in later (decompensated) stages [6,73,74]. Myocardial remodeling caused by hypertension also shows such two-step process in Cx43 expression; an upregulation in the compensated stage and a downregulation in the decompensated stage [74]. Reduced Cx43 expression is also evident in hearts with dilated and ischemic cardiomyopathy, following myocardial infarction and in failing hearts [6,72,75,76,77]. Cx43 degradation during I/R in rat hearts in vitro correlates with the induction of autophagy [66]. In addition, micro-RNA-1 plays a role in the regulation of Cx43 function and localization in cardiac hypertrophy in mice [61], in the protection from ischemic damage by telmisartan in rat myocardium [62] and in mice iPSC-CMs (induced pluripotent stem cell-derived cardiomyocytes) subjected to rapid pacing [78]. Cardiovascular diseases influence alternative Cx43 translation; for instance, in a mouse model of arrhythmogenic cardiomyopathy, the amount of Gja1-20k is reduced [79]. Furthermore, spontaneously hypertensive rats display diminished levels of Gja1-20k [45], whereas in mouse hearts undergoing I/R, the amounts of Gja1-20k are enhanced [47]. The quantity of Gja1-20k, in turn, may control the quantity of full-length Cx43; for example, in mice with mutated internal translation initiation sites, the levels of Cx43 are decreased [51]. The reduction in Cx43 can result in diminished intercellular coupling, and thereby to a decrease in conduction velocity. The influence of Cx43 on the conduction velocity is dependent on the extent of Cx43 protein reduction; a 50% reduction decreased conduction velocity by 44% in one study [80], but was without an effect in another study [81]. A further reduction to only 10% of baseline Cx43 leads to a ~33% decrease in the transverse conduction velocity [82]. Studies in which a specific contribution of mitochondrial Cx43 towards cardiac pathologies are investigated are scarce [56]; thus far, only a decrease in mitochondrial Cx43 in a rat model of dilated cardiomyopathy is described [83].
In general, Cx43 regulates a variety of cellular functions, such as conduction velocity, mitochondrial function [56], alterations in transcriptomics [43,84] and selective incorporation of micro-RNAs into extracellular vesicles [85]; all of these functions can be impaired by a reduction in Cx43 occurring in pathological conditions. In addition to the adverse consequences of reduced Cx43 in cardiac pathologies, at intercalated discs and probably also at other subcellular localizations, Cx43 lateralization at the plasma membrane alters Cx43 function and that of the cardiomyocytes. In the physiological situation, following Cx43 synthesis in the endoplasmic reticulum and oligomerization in the trans-Golgi network [86], the protein is directed to the intercalated discs in a process involving microtubules and actin filaments [87,88]. In pathological situations, there is often a shift of Cx43 from the intercalated discs to the lateral sides of the cardiomyocytes. Such lateralization occurs in several pathological states such as hypertension [89,90,91,92], post-myocardial infarction [93], and in hypertrophic, dilated and ischemic cardiomyopathy [6]. Lateralized Cx43 lowers the conduction velocity, thereby contributing to the development of arrhythmias [6], albeit there seems to be a large conduction reserve [94]. Hemichannels formed by lateralized Cx43 are activated under cardiac stress, which promotes the occurrence of arrhythmias and sudden death [95]. Data on the shift of Cx43 in cardiovascular pathologies at subcellular localizations other than the plasma membrane, such as mitochondria, nuclei and exosomes, are scarce. In addition to its amount and localization, Cx43 function is controlled by post-translational modifications such as phosphorylation. Interestingly, Cx43 phosphorylation also regulates the amount of Cx43 protein. In mice in which amino acids targeted by PKC, MAPK or CK1 are mutated to non-phosphorylatable residues, Cx43 expression is reduced [55]. However, mutation of the residues targeted by CK1 to phosphatase-resistant glutamic acids in human iPSC-CMs leads to decreased Cx43 expressions despite a tendency towards increased Cx43 mRNA levels [96]. The differences between the data relating to the consequences of mutated CK1-target sites may be explained by the different maturation stages of the cardiomyocytes.
Cx43 phosphorylation triggers its modification with lysine 63 (K63)-linked polyubiquitin chains and subsequent Cx43-internalization [97], with PKC being involved in Cx43 ubiquitination [98]. AKT is also implicated in this process, since the mutation of serine 373 to alanine renders Cx43 resistant to ubiquitination [66]. The kinases Src and ERK (extracellular-signal regulated kinase) and Cx43-S262 phosphorylation regulate connexosome formation [99,100]. In addition to the influence of Cx43 phosphorylation on its degradation, Cx43 phosphorylation status of Cx43 at specific residues is important for its lateralization. The mutation of residues targeted by CK1 to phospho-mimicking glutamic acids prevents Cx43 lateralization in a mouse model of Duchenne muscular dystrophy [25]. In the physiological situation, mutation of S368 to alanine does not induce a Cx43 shift away from the intercalated discs [101]. However, increased Cx43 lateralization in cardiomyocytes is associated with a dephosphorylation of the protein in cardiovascular diseases such as ischemia [102], heart failure [103,104] and hypertrophy [105]. In different cardiac diseases, Cx43 phosphorylation is altered such that the phosphorylation at serine 368 by PKC varies [106]. Cx43 phosphorylation at serine 368 by PKC influences Cx43 stability via conformational changes of Cx43-formed channels, which affect the channel permeability. Moreover, serine 368 phosphorylation influences Cx43 internalization and its degradation, leading to reduced intercellular communication [106]. Serine 373 targeted by AKT becomes increasingly phosphorylated in MDCK43 cells following 5–30 min of hypoxia, which contributes to the elimination of the protein–protein interaction between Cx43 and zonula occludens 1, in turn leading to increased gap junction size and intercellular communication. Following 60 min of hypoxia, serine 373 phosphorylation returns to control levels [107]. Increased gap junction size induced by serine 373 phosphorylation may indicate the first step in gap junction disassembly [54]. Phosphorylation at serines 325/328/330, which are targeted by CK1, is reduced in Chagas disease cardiomyopathy [108] and following ischemia in mouse hearts [109]. The amount of lateralized Cx43 increases following 30 min of ischemia in isolated mouse hearts, whereas lateralized Cx43 is not phosphorylated at serines 325/328/330, and the consequent reduced functionality of gap junctions may prevent aberrant cell–cell communication [109]. The loss of cardioprotection by ischemic preconditioning in mice with mutated serines 325/328/330 demonstrates the importance of these amino acids for myocardial I/R injury [55]. However, the influence of increased phosphorylation at serine 262, as well as decreased phosphorylation at serine 365 and 368, described in the mouse strain with mutated serines at positions 325/328/330 in the context of ischemic preconditioning, is presently unclear [55]. Ischemic preconditioning prevents the ischemia-induced dephosphorylation of Cx43 [110]. Regarding the importance of serine 282, it is shown that the mutation of this residue to alanine causes spontaneous arrhythmias in mice and rats [111,112]. Furthermore, mice with serines 325/328/330 mutated to alanine are susceptible to arrhythmias, whereas the mutation of these serines to phosphomimetic glutamic acids results in a diminished susceptibility to ventricular arrhythmias [113]. Generally, Cx43 dephosphorylation is associated with the occurrence of arrhythmias in heart failure and myocardial ischemia [114]. Details on the alterations of Cx43 phosphorylation in cardiac diseases such as I/R injury, heart failure, arrhythmia, hypertension and hypertrophy are reviewed elsewhere [115]. Importantly, the phosphorylation status of Cx43 is not only controlled by kinases, but also by phosphatase activity [116], and a dysregulation of protein phosphatases occurs in heart failure, cardiac ischemia and atrial fibrillation [114].
Collectively, cardiovascular pathologies are associated with Cx43 dysregulation, including decreased protein amounts, enhanced Cx43 lateralization at the plasma membrane, a modification of its phosphorylation status and changes of alternative translation. These parameters, which are crucial for the regulation of Cx43 function, show a variety of interactions that indicate a complex control of Cx43 function. The interactions of factors that contribute to the regulation of Cx43 amount and localization are summarized in Figure 1.
The Figure illustrates the relationships between factors important for the localization and the amount of Cx43 within cardiomyocytes, and therefore for Cx43-mediated function. The numbered blue boxes (1–4) in the schematic representation of Cx43 indicate transmembrane domains. The green dots represent Cx43 within mitochondria (inner mitochondrial membrane), nuclei and exosomes, and the orange dot represents Gja1-20k within mitochondria (outer mitochondrial membrane). The numbered gray dots represent the positions of some amino acids, which can be phosphorylated by protein kinases. The arrows indicate the interactions between Cx43 localizations and processes that regulate Cx43 function. Please note that the arrows only indicate which factor has an influence on another factor and not whether this influence is activating or inhibiting. For details of how the individual parameters relate to each other in terms of Cx43 localization and function, see the text and tables.

3. Overexpression of Cx43 as a Therapeutic Approach for the Treatment of Cardiovascular Diseases

The fundamental contribution of Cx43 to cardiac function makes it a potential target for the treatment of cardiovascular diseases. Pharmacological modulation of gap junctions and/or hemichannels can be accomplished by targeting various aspects of Cx43 function, including trafficking, channel gating and assembly or disassembly of gap junctions [12]. Whereas several chemicals (e.g., heptanol, carbenoxolone and 18α-glycyrrhetinic acid) affect Cx43-formed gap junctions or hemichannels [12], less information is available on their effects on Cx43 at other subcellular localizations such as mitochondria or nuclei. Peptide molecules (e.g., Gap19, Gap26, Gap27 or AAP10) are generally considered more specific than these chemicals, and are shown to interact with Cx43 or mimic its function, and can inhibit hemichannel function alone (Gap19) or also gap junctions (Gap26, Gap27) [117]. Analyzing the effects of these peptides which inhibit hemichannels while preserving gap-junctional communication reveals that they constitute a promising approach for treating heart diseases [118]. For detailed reviews summarizing the use of such peptides in cardiovascular pathologies, the reader is referred to references [119,120].
The demonstration of reduced Cx43 protein in several cardiovascular diseases (see Chapter 2) implies that overexpressing cardiac Cx43 may induce beneficial effects in the protection against these pathological conditions. To investigate the consequences of enhanced Cx43 expression in the physiological situation, transgenic mice were generated in which Cx43 is constitutively expressed under control of the cytomegalovirus promoter [121]. These mice show enhanced junctional communication in dye-coupling experiments, and although generally viable and fertile, have a reduced postnatal viability. Detailed analysis indicates that hearts of the Cx43-overexpressing mice have morphological defects in the right ventricle and the pulmonary outflow tract, as well as abnormalities in the coronary vasculature. The reduced viability and the malformations detected in these mice demonstrate that overexpressing Cx43 can trigger harmful effects. The mouse line described above is the only transgenic line published to date in which constitutive overexpression of Cx43 was achieved. Further studies, aimed at investigating the role of Cx43-overexpression, without using genetically modified animals, are required.
In this regard, Cx43 overexpression in iPSC-CMs results in enhanced Cx43 amounts, specifically at the sarcolemma, and augmentation of voltage-gated Na+-channel expression, as well as channel recruitment to the sarcolemma [122]. In turn, action potential upstroke velocity increases, which consequently leads to enhanced intercellular coupling. In a porcine model of atrial fibrillation induced by rapid pacing, adenoviral Cx43 gene transfer increases its protein levels and prevents the development of persistent atrial fibrillation [123]. Furthermore, it is shown that adenoviral Cx43 overexpression does not affect atrial conduction (P-wave duration and intra-atrial conduction time) in control animals, whereas it prevents atrial fibrillation in porcine myocardium [124]. In addition to its anti-arrhythmic effects, adenoviral Cx43 gene transfer enhances atrial Cx43 levels and prevents its lateralization, as evident in atrial fibrillation [124]. Injecting Cx43-expressing adenoviruses to the healed scar border of infarcted porcine myocardium improves conduction velocity and decreases ventricular tachycardia [125]. Whereas the amount of Cx43 increases following adenoviral gene transfer, the percentages of phosphorylated Cx43 and Cx43 located at the intercalated discs are similar between endogenous Cx43 and adenovirus-derived Cx43. In addition, injecting Cx43-expressing lentivirus in cryo-injured myocardium increases Cx43 expression and diminishes the occurrence of ventricular tachycardia in the post-infarct scar; this effect is most likely due to enhanced heterotypic cell–cell electrical coupling between the native myocardium and the infarct area [126].
The engraftment of Cx43-expressing skeletal myoblasts in mouse myocardial infarcts induced by cryoinjury protects against ventricular tachycardia [127]. However, these positive effects are not confirmed in a similarly structured study in infarcted rat hearts, in which the myoblasts are not from transgenic mice expressing Cx43 under the control of a skeletal muscle promoter, but are derived from cells transduced with a Cx43-carrying lentivirus [128]. The differences between these two studies may be explained by the different animal models and the level of Cx43 overexpression in the transplanted cells. In another attempt to treat myocardial infarction by cell transplantation, the delivery of mesenchymal stem cells combined with native or Cx43-overexpressing human skeletal myoblasts into the post-infarction mouse heart indicates no effect of Cx43 overexpression on the left or right ventricular ejection fraction or left/right ventricular mass [129]. Since (as mentioned above) there is an increase in both the overall amount of Cx43 and its lateralization in mdx mice (a common mouse model of Duchenne muscular dystrophy) and in human Duchenne hearts [95], it is proposed that Cx43 normalization (i.e., Cx43 downregulation), will alleviate cardiac dysfunction in this pathology. Indeed, crossing mdx mice with heterozygous Cx43-deficient mice decreases Cx43 expression to control values, reduces Cx43-lateralization and prevents the development of arrhythmias induced by an isoproterenol administration [130].
In porcine hearts in vivo, Cx43 translocation into mitochondria is associated with reduction of infarct size by ischemic preconditioning [131]. The cytoprotection by mitochondrial Cx43 is confirmed in stem cell antigen-1+ cells transfected with Cx43 fused to a mitochondrial targeting sequence [132]. Such a transfection increases the amounts of Cx43 at the inner mitochondrial membrane, reduces apoptosis and improves cell survival following oxygen–glucose deprivation. Augmenting the amount of Cx43 in HL1 cells by transfection leads to enhanced respiration [37]. However, the overexpression of mitochondrial Cx43 by injecting adeno-associated virus serotype 2/9 in mice induces mild diastolic dysfunction and increases the susceptibility to ventricular arrhythmia indicated by QTc, QT interval and JT interval prolongation [133]. Furthermore, doxorubicin-induced cardiotoxicity in H9C2 cells, which enhances the formation of reactive oxygen species (ROS), is characterized by increased amounts of mitochondrial Cx43 [134]. Therefore, these findings demonstrate that enhanced levels of mitochondrial Cx43 are not involved per se in protective signaling pathways; rather, the effects mediated by mitochondrial Cx43 seem to be dependent on the actual context (e.g., species, cell type, physiological or pathological situation) in which they are studied. A contribution of mitochondrial Gja1-20k to the cardioprotection by ischemic preconditioning is suggested, since Gja1-20k accumulates in mitochondria following a preconditioning stimulus [47]. The treatment of mice with adenoviruses to stimulate the Gja1-20k expression reduces ROS formation and infarct size following I/R induced by occlusion of the left anterior descending artery (LAD) in vivo as well as in Langendorff-perfused isolated mouse hearts [47]. Therefore, enhanced amounts of Gja1-20k in mouse hearts are associated with a cardioprotective phenotype. Table 1 summarizes studies in which direct effects of Cx43- or Gja1-20k-overexpression in cardiovascular cells are characterized.
There are several approaches for treating cardiovascular diseases which do not directly target Cx43, but are associated with changes in Cx43 amounts, and may therefore alter myocardial function. The results of such studies related to Cx43 expression and localization are summarized herein. Isolated rat hearts undergoing hypothermic I/R show an upregulation of miR-17, present increased apoptosis and develop arrhythmias. These alterations are associated with reduced amounts of Cx43. In contrast, miR-17 inhibition increases the conduction velocity and reduces arrhythmias. Inhibition of miR-17 in rat hearts undergoing hypothermic I/R is accompanied by enhanced Cx43 expression, al-though the amounts of Cx43 protein are significantly lower than in control hearts [135]. In H9C2 cells, hypoxia/reoxygenation (H/R) downregulates Cx43; the addition of lactate prior to H/R normalizes the Cx43 level to that of normoxic control cells [136]. In normoxic H9C2 cells, lactate augments the amounts of Cx43 mRNA and protein—an effect mediated by binding of the transcription factor EB to the Cx43 promoter. Ischemic cardiomyopathy in rats caused by 4-week LAD ligation decreases Cx43 expression, an effect counteracted by overexpressing caveolin-1, presumably via decreased phosphorylation of the tyrosine kinase cSrc [137]. Furthermore, I/R of isolated rat hearts reduces the Cx43 amount, which is partially prevented by injecting C2C12 cell-derived exosomes into the rat jugular vein prior to I/R [138]. A single targeted heavy ion radiation to rabbit hearts increases Cx43 at the intercalated discs and at the lateral sides of the cardiomyocytes, lasting for at least one year [139]. Despite the conceivable negative effects of Cx43 lateralization, no changes are detected in the ECG or echocardiogram up to one year post-radiation. These data indicate that in this model, Cx43 lateralization does not adversely affect cardiac function. In fact, with heavy ion radiation two weeks post-myocardial infarction (MI) in rabbit hearts in vivo, the moderate increase (~30% above control) in Cx43 and its lateralization are associated with reduced cardiac vulnerability to arrhythmias [140].
Rats treated with monocrotaline to induce pulmonary hypertension demonstrate Cx43 downregulation in right atrial tissue. The administration of the TRPV2 (transient receptor potential vanilloid type 2) inhibitor tranilast prevents the monocrotaline-stimulated remodeling of the right atrium and enhances Cx43 expression, while the amount of Cx43 is stabilized below the control level [141]. Another study describes the protective effects of monoterpene 1,8-cineole in monocrotaline-induced pulmonary hypertension, which are related predominantly to the prevention of Cx43-lateralization, rather than to the inhibition of Cx43 downregulation [92]. In contrast, pulmonary hypertension induced by chronic hypoxia promotes Cx43 expression in rat pulmonary arteries [142]. Moreover, enhanced amounts of Cx43 are detected in primary and third-level branches of mesenteric arterioles of spontaneously hypertensive rats [143].
Heart failure induced by transverse aortic constriction (TAC) in mice is accompanied by decreased Cx43 expression. The administration of carnosol (a phenolic compound found in rosemary) enhances Cx43 expression without attaining the levels in control animals, and reduces the vulnerability towards ventricular fibrillation [144]. Whereas Cx43 decreases in heart failure caused by high pacing in dogs, renal denervation increases its amount, yet below the control level [77]. In a heart failure model with preserved ejection fraction in mice induced by high-fat diet and L-NAME administration, Cx43 levels decline. This effect is partially reversed by the calcium-sensitizing drug levosimendan [145]. Cardiac remodeling induced by isoproterenol is associated with Cx43 downregulation, which is normalized to control levels by the administration of pinocembrin, a flavonoid with antioxidant and anti-inflammatory effects [146]. Pinocembrin also reduces the susceptibility to ventricular fibrillation.
Collectively, reduced Cx43 expression is a common feature of several cardiovascular diseases, although some pathological conditions such as the early stages of hypertrophy and hypertension are accompanied by enhanced Cx43 amounts. Accordingly, it is proposed that treating heart diseases associated with decreased Cx43 can be accomplished by enhancing Cx43 expression. Hence, whereas in some cases, Cx43 levels stagnate below those of respective controls, other settings demonstrate a complete restoration of Cx43 amounts. Of note, only one study describes Cx43 amounts above the control level, whereas all other studies report, at best, its normalization. Table 2 summarizes the studies in which therapeutic approaches to treat cardiovascular diseases do not directly target Cx43, yet are nevertheless associated with changes in Cx43 levels and/or Cx43 function. The specific interventions and their effects on Cx43 are provided in Table 2.

4. Conclusions

A common feature of several cardiovascular diseases is that they are associated with decreased amounts of Cx43. However, as biphasic responses (a short-term increase followed by a long-term decrease) evolve towards the pathological state, the conclusion as to whether up- or downregulation of Cx43 occurs is dependent on the time point analyzed. Further, some pathological conditions are associated with increased Cx43, implying that in these cases, reducing Cx43 levels can constitute a beneficial therapeutic approach. Importantly, in cardiovascular diseases associated with decreased Cx43, it should be noted that in most cases, the downregulation is shown only for total Cx43 (as mostly shown by Western blot analysis) or for gap-junctional Cx43 (mostly shown by immunohistochemistry). Such analysis does not take into account other localizations at which Cx43 is present, such as hemichannels, mitochondria, nuclei and exosomes, for which a specific Cx43 reduction in a certain pathological condition is possible, and thus should be determined. When considering the treatment of cardiovascular diseases associated with decreased Cx43, increasing Cx43 might be a beneficial therapeutic strategy. It is important to ensure that the Cx43 level is enhanced only in the cells and/or compartments where it was originally reduced. This approach takes into account that increasing Cx43 expression in sites presenting normal levels may induce adverse effects such as Cx43 lateralization, which may constitute a pro-arrhythmic substrate. Additionally, increased mitochondrial and nuclear Cx43 may adversely affect cellular energetics and gene expression, respectively. The complexity regarding the increase in mitochondrial Cx43 is further augmented by the fact that in cardiomyocytes, Cx43 is only localized in subsarcolemmal and not in interfibrillar mitochondria [56], and therefore Cx43 overexpression should specifically affect this mitochondrial subpopulation. It is noteworthy that only one of the abovementioned strategies for treating cardiovascular diseases is associated with increased Cx43 protein that exceeds the respective control group. Thus, it is unclear whether reaching the control values represents the maximal possible increase, or whether the amount of the protein is specifically stabilized at this level. Possibly, attaining the normal Cx43 levels and not beyond, in situations with decreased amounts of Cx43, is more beneficial than overexpressing Cx43, thereby preventing the adverse effects of excessive Cx43. It remains to be established whether modulation of Cx43 expression via interfering with micro-RNAs or protein degradation prevents Cx43 overexpression, thereby contributing to the Cx43 stabilization at control values. Accordingly, it will be advantageous to investigate the contribution of micro-RNAs and protein degradation to Cx43 regulation, and thus discover additional targets for controlling Cx43 expression. Additionally, since alternative translation of Cx43 is important for regulating the amount of full-length Cx43, its contribution towards normalization of Cx43 amounts in the treatment of cardiovascular diseases needs to be further investigated. Therefore, the analysis of factors affecting Cx43 levels under conditions in which Cx43 is gradually increased is highly relevant. The use of Cx43 as a therapeutic target in cardiovascular diseases associated with reduced Cx43 levels should not only ensure the increase in Cx43 protein levels; rather, Cx43 function should also be controlled in terms of its trafficking to and assembly into gap junctions, gating of Cx43-formed channels, functional control of mitochondria, nuclei and exosomes, as well as Cx43 degradation. Finally, when considering increasing Cx43 expression in the diseased myocardium, a physiological phosphorylation of the protein needs to be ensured to prevent aberrant Cx43 function.
In summary, the presence of reduced Cx43 levels in diseased myocardium suggests that Cx43 overexpression can represent a therapeutic approach for the treatment of such diseases; indeed, beneficial effects elicited by increased Cx43 protein amounts are demonstrated. However, it should be taken into consideration that Cx43 overexpression may be detrimental in the developing heart (as shown by constitutive Cx43 overexpression in transgenic mice) and that some cardiovascular diseases are associated with enhanced Cx43 levels. Thus, a moderate increase in Cx43 to physiological levels may be more effective in the treatment of cardiovascular disease than excessive overexpression, as this may circumvent potential negative effects of increased Cx43 amounts. The complex regulation of Cx43 through its different localizations in cellular compartments, phosphorylation, alternative translation or degradation necessitates tight control of its function at multiple levels. Accordingly, a detailed analysis of the parameters modulated by Cx43 is required to fully assess the consequences of increased Cx43 amounts on cell function. Further investigations are needed to study the interplay between the individual components of Cx43 regulation and their importance in the diseased myocardium.

Funding

This research received no funding.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CK1Casein kinase 1
CxConnexin
DMDDuchenne muscular dystrophy
ERADEndoplasmic reticulum-associated degradation
ERKExtracellular-signal regulated kinase
H/Rhypoxia/reoxygenation
K63Polyubiquitylation at lysine 63
LADLeft anterior descending artery
I/RIschemia/reperfusion
iPSC-CMsinduced pluripotent stem cell-derived cardiomyocytes
MAPKMitogen-activated protein kinase
MIMyocardial infarction
PKCProtein kinase C
ROSReactive oxygen species
TACTransverse aortic constriction
TRPV2Transient receptor potential vanilloid type 2

References

  1. Söhl, G.; Willecke, K. Gap junctions and the connexin protein family. Cardiovasc. Res. 2004, 62, 228–232. [Google Scholar] [CrossRef] [PubMed]
  2. Beyer, E.C.; Berthoud, V.M. Gap junction gene and protein families: Connexins, innexins, and pannexins. Biochim. Biophys. Acta Biomembr. 2018, 1860, 5–8. [Google Scholar] [CrossRef] [PubMed]
  3. Lampe, P.D.; Laird, D.W. Recent advances in connexin gap junction biology. Fac. Rev. 2022, 11, 14. [Google Scholar] [CrossRef] [PubMed]
  4. Lukowicz-Bedford, R.M.; Farnsworth, D.R.; Miller, A.C. Connexinplexity: The spatial and temporal expression of connexin genes during vertebrate organogenesis. G3 2022, 12, jkac062. [Google Scholar] [CrossRef]
  5. Aasen, T.; Johnstone, S.; Vidal-Brime, L.; Lynn, K.S.; Koval, M. Connexins: Synthesis, Post-Translational Modifications, and Trafficking in Health and Disease. Int. J. Mol. Sci. 2018, 19, 1296. [Google Scholar] [CrossRef]
  6. Fontes, M.S.; van Veen, T.A.; de Bakker, J.M.; van Rijen, H.V. Functional consequences of abnormal Cx43 expression in the heart. Biochim. Biophys. Acta 2012, 1818, 2020–2029. [Google Scholar] [CrossRef]
  7. Kreuzberg, M.M.; Schrickel, J.W.; Ghanem, A.; Kim, J.S.; Degen, J.; Janssen-Bienhold, U.; Lewalter, T.; Tiemann, K.; Willecke, K. Connexin30.2 containing gap junction channels decelerate impulse propagation through the atrioventricular node. Proc. Natl. Acad. Sci. USA 2006, 103, 5959–5964. [Google Scholar] [CrossRef]
  8. Lei, L.; Wang, Y.F.; Chen, C.Y.; Wang, Y.T.; Zhang, Y. Novel insight into astrocyte-mediated gliotransmission modulates the synaptic plasticity in major depressive disorder. Life Sci. 2024, 355, 122988. [Google Scholar] [CrossRef]
  9. Lucaciu, S.A.; Leighton, S.E.; Hauser, A.; Yee, R.; Laird, D.W. Diversity in connexin biology. J. Biol. Chem. 2023, 299, 105263. [Google Scholar] [CrossRef]
  10. González-Casanova, J.; Schmachtenberg, O.; Martínez, A.D.; Sanchez, H.A.; Harcha, P.A.; Rojas-Gomez, D. An Update on Connexin Gap Junction and Hemichannels in Diabetic Retinopathy. Int. J. Mol. Sci. 2021, 22, 3194. [Google Scholar] [CrossRef]
  11. Swartzendruber, J.A.; Nicholson, B.J.; Murthy, A.K. The Role of Connexin 43 in Lung Disease. Life 2020, 10, 363. [Google Scholar] [CrossRef] [PubMed]
  12. Leybaert, L.; Lampe, P.D.; Dhein, S.; Kwak, B.R.; Ferdinandy, P.; Beyer, E.C.; Laird, D.W.; Naus, C.C.; Green, C.R.; Schulz, R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharmacol. Rev. 2017, 69, 396–478. [Google Scholar] [PubMed]
  13. Laird, D.W.; Lampe, P.D. Therapeutic strategies targeting connexins. Nat. Rev. Drug Discov. 2018, 17, 905–921. [Google Scholar] [CrossRef] [PubMed]
  14. Beyer, E.C.; Paul, D.L.; Goodenough, D.A. Connexin43: A protein from rat heart homologous to a gap junction protein from liver. J. Cell Biol. 1987, 105, 2621–2629. [Google Scholar] [CrossRef]
  15. Harris, A.L. Emerging issues of connexin channels: Biophysics fills the gap. Q. Rev. Biophys. 2001, 34, 325–472. [Google Scholar] [CrossRef]
  16. Weber, P.A.; Chang, H.C.; Spaeth, K.E.; Nitsche, J.M.; Nicholson, B.J. The permeability of gap junction channels to probes of different size is dependent on connexin composition and permeant-pore affinities. Biophys. J. 2004, 87, 958–973. [Google Scholar] [CrossRef]
  17. Harris, A.L. Connexin channel permeability to cytoplasmic molecules. Prog. Biophys. Mol. Biol. 2007, 94, 120–143. [Google Scholar] [CrossRef]
  18. Beyer, E.C.; Kistler, J.; Paul, D.L.; Goodenough, D.A. Antisera directed against connexin43 peptides react with a 43-kD protein localized to gap junctions in myocardium and other tissues. J. Cell Biol. 1989, 108, 595–605. [Google Scholar] [CrossRef]
  19. Martins-Marques, T.; Pinho, M.J.; Zuzarte, M.; Oliveira, C.; Pereira, P.; Sluijter, J.P.; Gomes, C.; Girao, H. Presence of Cx43 in extracellular vesicles reduces the cardiotoxicity of the anti-tumour therapeutic approach with doxorubicin. J. Extracell. Vesicles 2016, 5, 32538. [Google Scholar] [CrossRef]
  20. Soares, A.R.; Martins-Marques, T.; Ribeiro-Rodrigues, T.; Ferreira, J.V.; Catarino, S.; Pinho, M.J.; Zuzarte, M.; Isabel Anjo, S.; Manadas, B.; Sluijter, J.P.G.; et al. Gap junctional protein Cx43 is involved in the communication between extracellular vesicles and mammalian cells. Sci. Rep. 2015, 5, 13243. [Google Scholar]
  21. Gemel, J.; Kilkus, J.; Dawson, G.; Beyer, E.C. Connecting Exosomes and Connexins. Cancers 2019, 11, 476. [Google Scholar] [CrossRef] [PubMed]
  22. Yang, Z.J.; Bi, Q.C.; Gan, L.J.; Zhang, L.L.; Wei, M.J.; Hong, T.; Liu, R.; Qiu, C.L.; Han, X.J.; Jiang, L.P. Exosomes Derived from Glioma Cells under Hypoxia Promote Angiogenesis through Up-regulated Exosomal Connexin 43. Int. J. Med. Sci. 2022, 19, 1205–1215. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, W.; Guo, Y.; Yang, W.; Chen, L.; Ren, D.; Wu, C.; He, B.; Zheng, P.; Tong, W. Phosphorylation of connexin 43 induced by traumatic brain injury promotes exosome release. J. Neurophysiol. 2018, 119, 305–311. [Google Scholar] [CrossRef] [PubMed]
  24. Sáez, J.C.; Schalper, K.A.; Retamal, M.A.; Orellana, J.A.; Shoji, K.F.; Bennett, M.V. Cell membrane permeabilization via connexin hemichannels in living and dying cells. Exp. Cell Res. 2010, 316, 2377–2389. [Google Scholar] [CrossRef]
  25. Himelman, E.; Lillo, M.A.; Nouet, J.; Gonzalez, J.P.; Zhao, Q.; Xie, L.H.; Li, H.; Liu, T.; Wehrens, X.H.; Lampe, P.D.; et al. Prevention of connexin-43 remodeling protects against Duchenne muscular dystrophy cardiomyopathy. J. Clin. Investig. 2020, 130, 1713–1727. [Google Scholar] [CrossRef]
  26. Kim, J.C.; Pérez-Hernández, M.; Alvarado, F.J.; Maurya, S.R.; Montnach, J.; Yin, Y.; Zhang, M.; Lin, X.; Vasquez, C.; Heguy, A.; et al. Disruption of Ca(2+)(i) Homeostasis and Connexin 43 Hemichannel Function in the Right Ventricle Precedes Overt Arrhythmogenic Cardiomyopathy in Plakophilin-2-Deficient Mice. Circulation 2019, 140, 1015–1030. [Google Scholar] [CrossRef]
  27. Wang, N.; De Vuyst, E.; Ponsaerts, R.; Boengler, K.; Palacios-Prado, N.; Wauman, J.; Lai, C.P.; De Bock, M.; Decrock, E.; Bol, M.; et al. Selective inhibition of Cx43 hemichannels by Gap19 and its impact on myocardial ischemia/reperfusion injury. Basic Res. Cardiol. 2013, 108, 309. [Google Scholar] [CrossRef]
  28. Boengler, K.; Stahlhofen, S.; van de Sand, A.; Gres, P.; Ruiz-Meana, M.; Garcia-Dorado, D.; Heusch, G.; Schulz, R. Presence of connexin 43 in subsarcolemmal, but not in interfibrillar cardiomyocyte mitochondria. Basic Res. Cardiol. 2009, 104, 141–147. [Google Scholar] [CrossRef]
  29. Sun, J.; Nguyen, T.; Aponte, A.M.; Menazza, S.; Kohr, M.J.; Roth, D.M.; Patel, H.H.; Murphy, E.; Steenbergen, C. Ischaemic preconditioning preferentially increases protein S-nitrosylation in subsarcolemmal mitochondria. Cardiovasc. Res. 2015, 106, 227–236. [Google Scholar] [CrossRef]
  30. Denuc, A.; Núñez, E.; Calvo, E.; Loureiro, M.; Miro-Casas, E.; Guarás, A.; Vázquez, J.; Garcia-Dorado, D. New protein-protein interactions of mitochondrial connexin 43 in mouse heart. J. Cell. Mol. Med. 2016, 20, 794–803. [Google Scholar] [CrossRef]
  31. Wang, M.; Smith, K.; Yu, Q.; Miller, C.; Singh, K.; Sen, C.K. Mitochondrial connexin 43 in sex-dependent myocardial responses and estrogen-mediated cardiac protection following acute ischemia/reperfusion injury. Basic Res. Cardiol. 2019, 115, 1. [Google Scholar] [CrossRef] [PubMed]
  32. Palmer, J.W.; Tandler, B.; Hoppel, C.L. Biochemical differences between subsarcolemmal and interfibrillar mitochondria from rat cardiac muscle: Effects of procedural manipulations. Arch. Biochem. Biophys. 1985, 236, 691–702. [Google Scholar] [CrossRef] [PubMed]
  33. Palmer, J.W.; Tandler, B.; Hoppel, C.L. Heterogeneous response of subsarcolemmal heart mitochondria to calcium. Am. J. Physiol. 1986, 250, H741–H748. [Google Scholar] [CrossRef] [PubMed]
  34. Rodriguez-Sinovas, A.; Boengler, K.; Cabestrero, A.; Gres, P.; Morente, M.; Ruiz-Meana, M.; Konietzka, I.; Miró, E.; Totzeck, A.; Heusch, G.; et al. Translocation of connexin 43 to the inner mitochondrial membrane of cardiomyocytes through the heat shock protein 90-dependent TOM pathway and its importance for cardioprotection. Circ. Res. 2006, 99, 93–101. [Google Scholar] [CrossRef]
  35. Srisakuldee, W.; Makazan, Z.; Nickel, B.E.; Zhang, F.; Thliveris, J.A.; Pasumarthi, K.B.; Kardami, E. The FGF-2-triggered protection of cardiac subsarcolemmal mitochondria from calcium overload is mitochondrial connexin 43-dependent. Cardiovasc. Res. 2014, 103, 72–80. [Google Scholar] [CrossRef]
  36. Miro-Casas, E.; Ruiz-Meana, M.; Agullo, E.; Stahlhofen, S.; Rodríguez-Sinovas, A.; Cabestrero, A.; Jorge, I.; Torre, I.; Vazquez, J.; Boengler, K.; et al. Connexin43 in cardiomyocyte mitochondria contributes to mitochondrial potassium uptake. Cardiovasc. Res. 2009, 83, 747–756. [Google Scholar] [CrossRef]
  37. Boengler, K.; Ruiz-Meana, M.; Gent, S.; Ungefug, E.; Soetkamp, D.; Miro-Casas, E.; Cabestrero, A.; Fernandez-Sanz, C.; Semenzato, M.; Di Lisa, F.; et al. Mitochondrial connexin 43 impacts on respiratory complex I activity and mitochondrial oxygen consumption. J. Cell. Mol. Med. 2012, 16, 1649–1655. [Google Scholar] [CrossRef]
  38. Boengler, K.; Ungefug, E.; Heusch, G.; Leybaert, L.; Schulz, R. Connexin 43 impacts on mitochondrial potassium uptake. Front. Pharmacol. 2013, 4, 73. [Google Scholar] [CrossRef]
  39. Soetkamp, D.; Nguyen, T.T.; Menazza, S.; Hirschhäuser, C.; Hendgen-Cotta, U.B.; Rassaf, T.; Schlüter, K.D.; Boengler, K.; Murphy, E.; Schulz, R. S-nitrosation of mitochondrial connexin 43 regulates mitochondrial function. Basic Res. Cardiol. 2014, 109, 433. [Google Scholar] [CrossRef]
  40. Gadicherla, A.K.; Wang, N.; Bulic, M.; Agullo-Pascual, E.; Lissoni, A.; De Smet, M.; Delmar, M.; Bultynck, G.; Krysko, D.V.; Camara, A.; et al. Mitochondrial Cx43 hemichannels contribute to mitochondrial calcium entry and cell death in the heart. Basic Res. Cardiol. 2017, 112, 27. [Google Scholar] [CrossRef]
  41. Martins-Marques, T.; Witschas, K.; Ribeiro, I.; Zuzarte, M.; Catarino, S.; Ribeiro-Rodrigues, T.; Caramelo, F.; Aasen, T.; Carreira, I.M.; Goncalves, L.; et al. Cx43 can form functional channels at the nuclear envelope and modulate gene expression in cardiac cells. Open Biol. 2023, 13, 230258. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, W.; Feng, Y.; Li, T.; Shi, T.; Hui, W.; Liu, H.; Gao, F. The role of enhanced expression of Cx43 in patients with ulcerative colitis. Open Med. 2024, 19, 20230885. [Google Scholar] [CrossRef]
  43. Iacobas, D.A.; Urban-Maldonado, M.; Iacobas, S.; Scemes, E.; Spray, D.C. Array analysis of gene expression in connexin-43 null astrocytes. Physiol. Genom. 2003, 15, 177–190. [Google Scholar] [CrossRef] [PubMed]
  44. Smyth, J.W.; Shaw, R.M. Autoregulation of connexin43 gap junction formation by internally translated isoforms. Cell Rep. 2013, 5, 611–618. [Google Scholar] [CrossRef] [PubMed]
  45. Fu, Y.L.; Tao, L.; Peng, F.H.; Zheng, N.Z.; Lin, Q.; Cai, S.Y.; Wang, Q. GJA1-20k attenuates Ang II-induced pathological cardiac hypertrophy by regulating gap junction formation and mitochondrial function. Acta Pharmacol. Sin. 2021, 42, 536–549. [Google Scholar] [CrossRef]
  46. Basheer, W.; Shaw, R. The “tail” of Connexin43: An unexpected journey from alternative translation to trafficking. Biochim. Biophys. Acta 2016, 1863, 1848–1856. [Google Scholar] [CrossRef]
  47. Basheer, W.A.; Fu, Y.; Shimura, D.; Xiao, S.; Agvanian, S.; Hernandez, D.M.; Hitzeman, T.C.; Hong, T.; Shaw, R.M. Stress response protein GJA1-20k promotes mitochondrial biogenesis, metabolic quiescence, and cardioprotection against ischemia/reperfusion injury. JCI Insight 2018, 3, e121900. [Google Scholar] [CrossRef]
  48. James, C.C.; Zeitz, M.J.; Calhoun, P.J.; Lamouille, S.; Smyth, J.W. Altered translation initiation of Gja1 limits gap junction formation during epithelial-mesenchymal transition. Mol. Biol. Cell 2018, 29, 797–808. [Google Scholar] [CrossRef]
  49. Basheer, W.A.; Xiao, S.; Epifantseva, I.; Fu, Y.; Kleber, A.G.; Hong, T.; Shaw, R.M. GJA1-20k Arranges Actin to Guide Cx43 Delivery to Cardiac Intercalated Discs. Circ. Res. 2017, 121, 1069–1080. [Google Scholar] [CrossRef]
  50. Irwin, R.M.; Thomas, M.A.; Fahey, M.J.; Mayán, M.D.; Smyth, J.W.; Delco, M.L. Connexin 43 regulates intercellular mitochondrial transfer from human mesenchymal stromal cells to chondrocytes. Stem Cell Res. Ther. 2024, 15, 359. [Google Scholar] [CrossRef]
  51. 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]
  52. D’Hondt, C.; Iyyathurai, J.; Vinken, M.; Rogiers, V.; Leybaert, L.; Himpens, B.; Bultynck, G. Regulation of connexin- and pannexin-based channels by post-translational modifications. Biol. Cell 2013, 105, 373–398. [Google Scholar] [CrossRef] [PubMed]
  53. Retamal, M.A.; García, I.E.; Pinto, B.I.; Pupo, A.; Báez, D.; Stehberg, J.; Del Rio, R.; González, C. Extracellular Cysteine in Connexins: Role as Redox Sensors. Front. Physiol. 2016, 7, 1. [Google Scholar] [CrossRef] [PubMed]
  54. Solan, J.L.; Lampe, P.D. Spatio-temporal regulation of connexin43 phosphorylation and gap junction dynamics. Biochim. Biophys. Acta Biomembr. 2018, 1860, 83–90. [Google Scholar] [CrossRef]
  55. Hirschhäuser, C.; Lissoni, A.; Görge, P.M.; Lampe, P.D.; Heger, J.; Schlüter, K.D.; Leybaert, L.; Schulz, R.; Boengler, K. Connexin 43 phosphorylation by casein kinase 1 is essential for the cardioprotection by ischemic preconditioning. Basic Res. Cardiol. 2021, 116, 21. [Google Scholar] [CrossRef]
  56. Boengler, K.; Leybaert, L.; Ruiz-Meana, M.; Schulz, R. Connexin 43 in Mitochondria: What Do We Really Know About Its Function? Front. Physiol. 2022, 13, 928934. [Google Scholar] [CrossRef]
  57. Boengler, K.; Eickelmann, C.; Kleinbongard, P. Mitochondrial Kinase Signaling for Cardioprotection. Int. J. Mol. Sci. 2024, 25, 4491. [Google Scholar] [CrossRef]
  58. Laird, D.W.; Puranam, K.L.; Revel, J.P. Turnover and phosphorylation dynamics of connexin43 gap junction protein in cultured cardiac myocytes. Biochem. J. 1991, 273 (Pt 1), 67–72. [Google Scholar] [CrossRef]
  59. Beardslee, M.A.; Laing, J.G.; Beyer, E.C.; Saffitz, J.E. Rapid turnover of connexin43 in the adult rat heart. Circ. Res. 1998, 83, 629–635. [Google Scholar] [CrossRef]
  60. Zhang, S.S.; Shaw, R.M. Multilayered regulation of cardiac ion channels. Biochim. Biophys. Acta 2013, 1833, 876–885. [Google Scholar] [CrossRef]
  61. Curcio, A.; Torella, D.; Iaconetti, C.; Pasceri, E.; Sabatino, J.; Sorrentino, S.; Giampà, S.; Micieli, M.; Polimeni, A.; Henning, B.J.; et al. MicroRNA-1 downregulation increases connexin 43 displacement and induces ventricular tachyarrhythmias in rodent hypertrophic hearts. PLoS ONE 2013, 8, e70158. [Google Scholar] [CrossRef] [PubMed]
  62. Trotta, M.C.; Ferraro, B.; Messina, A.; Panarese, I.; Gulotta, E.; Nicoletti, G.F.; D’Amico, M.; Pieretti, G. Telmisartan cardioprotects from the ischaemic/hypoxic damage through a miR-1-dependent pathway. J. Cell. Mol. Med. 2019, 23, 6635–6645. [Google Scholar] [CrossRef] [PubMed]
  63. Falk, M.M.; Bell, C.L.; Kells Andrews, R.M.; Murray, S.A. Molecular mechanisms regulating formation, trafficking and processing of annular gap junctions. BMC Cell Biol. 2016, 17 (Suppl. S1), 22. [Google Scholar] [CrossRef] [PubMed]
  64. Totland, M.Z.; Rasmussen, N.L.; Knudsen, L.M.; Leithe, E. Regulation of gap junction intercellular communication by connexin ubiquitination: Physiological and pathophysiological implications. Cell. Mol. Life Sci. 2020, 77, 573–591. [Google Scholar] [CrossRef]
  65. Su, V.; Cochrane, K.; Lau, A.F. Degradation of connexins through the proteasomal, endolysosomal and phagolysosomal pathways. J. Membr. Biol. 2012, 245, 389–400. [Google Scholar] [CrossRef]
  66. Martins-Marques, T.; Catarino, S.; Zuzarte, M.; Marques, C.; Matafome, P.; Pereira, P.; Girão, H. Ischaemia-induced autophagy leads to degradation of gap junction protein connexin43 in cardiomyocytes. Biochem. J. 2015, 467, 231–245. [Google Scholar] [CrossRef]
  67. Dhein, S.; Salameh, A. Remodeling of Cardiac Gap Junctional Cell-Cell Coupling. Cells 2021, 10, 2422. [Google Scholar] [CrossRef]
  68. Sykora, M.; Szeiffova Bacova, B.; Andelova, K.; Egan Benova, T.; Martiskova, A.; Kurahara, L.H.; Hirano, K.; Tribulova, N. Connexin43, A Promising Target to Reduce Cardiac Arrhythmia Burden in Pulmonary Arterial Hypertension. Int. J. Mol. Sci. 2024, 25, 3275. [Google Scholar] [CrossRef]
  69. Sykora, M.; Andelova, K.; Szeiffova Bacova, B.; Egan Benova, T.; Martiskova, A.; Knezl, V.; Tribulova, N. Hypertension Induces Pro-arrhythmic Cardiac Connexome Disorders: Protective Effects of Treatment. Biomolecules 2023, 13, 330. [Google Scholar] [CrossRef]
  70. Michela, P.; Velia, V.; Aldo, P.; Ada, P. Role of connexin 43 in cardiovascular diseases. Eur. J. Pharmacol. 2015, 768, 71–76. [Google Scholar] [CrossRef]
  71. de Jong, S.; van Veen, T.A.; van Rijen, H.V.; de Bakker, J.M. Fibrosis and cardiac arrhythmias. J. Cardiovasc. Pharmacol. 2011, 57, 630–638. [Google Scholar] [CrossRef] [PubMed]
  72. Yang, F.; Zhang, X.L.; Liu, H.H.; Qian, L.L.; Wang, R.X. Post translational modifications of connexin 43 in ventricular arrhythmias after myocardial infarction. Mol. Biol. Rep. 2024, 51, 329. [Google Scholar] [CrossRef] [PubMed]
  73. Kostin, S.; Dammer, S.; Hein, S.; Klovekorn, W.P.; Bauer, E.P.; Schaper, J. Connexin 43 expression and distribution in compensated and decompensated cardiac hypertrophy in patients with aortic stenosis. Cardiovasc. Res. 2004, 62, 426–436. [Google Scholar] [CrossRef] [PubMed]
  74. Egan Benova, T.; Szeiffova Bacova, B.; Viczenczova, C.; Diez, E.; Barancik, M.; Tribulova, N. Protection of cardiac cell-to-cell coupling attenuate myocardial remodeling and proarrhythmia induced by hypertension. Physiol. Res. 2016, 65 (Suppl. S1), S29–S42. [Google Scholar] [CrossRef]
  75. Laing, J.G.; Saffitz, J.E.; Steinberg, T.H.; Yamada, K.A. Diminished zonula occludens-1 expression in the failing human heart. Cardiovasc. Pathol. 2007, 16, 159–164. [Google Scholar] [CrossRef]
  76. Kostin, S. Zonula occludens-1 and connexin 43 expression in the failing human heart. J. Cell. Mol. Med. 2007, 11, 892–895. [Google Scholar] [CrossRef]
  77. Liang, X.; Shang, S.; Bai, Z.; Wang, Q.; Fan, Y.; Xiaokereti, J.; Lv, H.; Zhou, X.; Lu, Y.; Tang, B. Effects of Renal Denervation on Gap Junction in High-Pacing-Induced Heart Failure Dogs. Anatol. J. Cardiol. 2024, 28, 429–436. [Google Scholar] [CrossRef]
  78. Wahl, C.M.; Schmidt, C.; Hecker, M.; Ullrich, N.D. Distress-Mediated Remodeling of Cardiac Connexin-43 in a Novel Cell Model for Arrhythmogenic Heart Diseases. Int. J. Mol. Sci. 2022, 23, 10174. [Google Scholar] [CrossRef]
  79. Palatinus, J.A.; Valdez, S.; Taylor, L.; Whisenant, C.; Selzman, C.H.; Drakos, S.G.; Ranjan, R.; Hong, T.; Saffitz, J.E.; Shaw, R.M. GJA1-20k Rescues Cx43 Localization and Arrhythmias in Arrhythmogenic Cardiomyopathy. Circ. Res. 2023, 132, 744–746. [Google Scholar] [CrossRef]
  80. Guerrero, P.A.; Schuessler, R.B.; Davis, L.M.; Beyer, E.C.; Johnson, C.M.; Yamada, K.A.; Saffitz, J.E. Slow ventricular conduction in mice heterozygous for a connexin43 null mutation. J. Clin. Investig. 1997, 99, 1991–1998. [Google Scholar] [CrossRef]
  81. Morley, G.E.; Vaidya, D.; Samie, F.H.; Lo, C.; Delmar, M.; Jalife, J. Characterization of conduction in the ventricles of normal and heterozygous Cx43 knockout mice using optical mapping. J. Cardiovasc. Electrophysiol. 1999, 10, 1361–1375. [Google Scholar] [CrossRef] [PubMed]
  82. van Rijen, H.V.; Eckardt, D.; Degen, J.; Theis, M.; Ott, T.; Willecke, K.; Jongsma, H.J.; Opthof, T.; de Bakker, J.M. Slow conduction and enhanced anisotropy increase the propensity for ventricular tachyarrhythmias in adult mice with induced deletion of connexin43. Circulation 2004, 109, 1048–1055. [Google Scholar] [CrossRef] [PubMed]
  83. Shan, H.; Wei, J.; Zhang, M.; Lin, L.; Yan, R.; Zhang, R.; Zhu, Y.H. Suppression of PKCε-mediated mitochondrial connexin 43 phosphorylation at serine 368 is involved in myocardial mitochondrial dysfunction in a rat model of dilated cardiomyopathy. Mol. Med. Rep. 2015, 11, 4720–4726. [Google Scholar] [CrossRef] [PubMed]
  84. Iacobas, D.A.; Iacobas, S.; Urban-Maldonado, M.; Scemes, E.; Spray, D.C. Similar transcriptomic alterations in Cx43 knockdown and knockout astrocytes. Cell Commun. Adhes. 2008, 15, 195–206. [Google Scholar] [CrossRef]
  85. Martins-Marques, T.; Costa, M.C.; Catarino, S.; Simoes, I.; Aasen, T.; Enguita, F.J.; Girao, H. Cx43-mediated sorting of miRNAs into extracellular vesicles. EMBO Rep. 2022, 23, e54312. [Google Scholar] [CrossRef]
  86. Epifantseva, I.; Shaw, R.M. Intracellular trafficking pathways of Cx43 gap junction channels. Biochim. Biophys. Acta Biomembr. 2018, 1860, 40–47. [Google Scholar] [CrossRef]
  87. Smyth, J.W.; Vogan, J.M.; Buch, P.J.; Zhang, S.S.; Fong, T.S.; Hong, T.T.; Shaw, R.M. Actin cytoskeleton rest stops regulate anterograde traffic of connexin 43 vesicles to the plasma membrane. Circ. Res. 2012, 110, 978–989. [Google Scholar] [CrossRef]
  88. Fort, A.G.; Murray, J.W.; Dandachi, N.; Davidson, M.W.; Dermietzel, R.; Wolkoff, A.W.; Spray, D.C. In vitro motility of liver connexin vesicles along microtubules utilizes kinesin motors. J. Biol. Chem. 2011, 286, 22875–22885. [Google Scholar] [CrossRef]
  89. Dumitrescu, M.; Constantin, A.; Nemecz, A.M.; Drăgan, E.; Popov, L.D.; Tanko, G. Hypertension induces compensatory left ventricular hypertrophy by a mechanism involving gap junction lateralization and overexpression of CD36, PKC and MMP-2. Rom. J. Morphol. Embryol. 2021, 62, 713–721. [Google Scholar] [CrossRef]
  90. Chkourko, H.S.; Guerrero-Serna, G.; Lin, X.; Darwish, N.; Pohlmann, J.R.; Cook, K.E.; Martens, J.R.; Rothenberg, E.; Musa, H.; Delmar, M. Remodeling of mechanical junctions and of microtubule-associated proteins accompany cardiac connexin43 lateralization. Heart Rhythm 2012, 9, 1133–1140.e1136. [Google Scholar] [CrossRef]
  91. Fialová, M.; Dlugošová, K.; Okruhlicová, L.; Kristek, F.; Manoach, M.; Tribulová, N. Adaptation of the heart to hypertension is associated with maladaptive gap junction connexin-43 remodeling. Physiol. Res. 2008, 57, 7–11. [Google Scholar] [CrossRef] [PubMed]
  92. Alves-Silva, J.M.; Zuzarte, M.; Marques, C.; Viana, S.; Preguiça, I.; Baptista, R.; Ferreira, C.; Cavaleiro, C.; Domingues, N.; Sardão, V.A.; et al. 1,8-Cineole ameliorates right ventricle dysfunction associated with pulmonary arterial hypertension by restoring connexin43 and mitochondrial homeostasis. Pharmacol. Res. 2022, 180, 106151. [Google Scholar] [CrossRef] [PubMed]
  93. Macia, E.; Dolmatova, E.; Cabo, C.; Sosinsky, A.Z.; Dun, W.; Coromilas, J.; Ciaccio, E.J.; Boyden, P.A.; Wit, A.L.; Duffy, H.S. Characterization of gap junction remodeling in epicardial border zone of healing canine infarcts and electrophysiological effects of partial reversal by rotigaptide. Circ. Arrhythm. Electrophysiol. 2011, 4, 344–351. [Google Scholar] [CrossRef] [PubMed]
  94. Stein, M.; Boulaksil, M.; Engelen, M.A.; van Veen, T.A.; Hauer, R.N.; de Bakker, J.M.; van Rijen, H.V. Conduction reserve and arrhythmias. Neth. Heart J. 2006, 14, 113–116. [Google Scholar]
  95. Gonzalez, J.P.; Ramachandran, J.; Xie, L.H.; Contreras, J.E.; Fraidenraich, D. Selective Connexin43 Inhibition Prevents Isoproterenol-Induced Arrhythmias and Lethality in Muscular Dystrophy Mice. Sci. Rep. 2015, 5, 13490. [Google Scholar] [CrossRef]
  96. Al-Attar, R.; Jargstorf, J.; Romagnuolo, R.; Jouni, M.; Alibhai, F.J.; Lampe, P.D.; Solan, J.L.; Laflamme, M.A. Casein Kinase 1 Phosphomimetic Mutations Negatively Impact Connexin-43 Gap Junctions in Human Pluripotent Stem Cell-Derived Cardiomyocytes. Biomolecules 2024, 14, 61. [Google Scholar] [CrossRef]
  97. Kells-Andrews, R.M.; Margraf, R.A.; Fisher, C.G.; Falk, M.M. Connexin-43 K63-polyubiquitylation on lysines 264 and 303 regulates gap junction internalization. J. Cell Sci. 2018, 131, jcs204321. [Google Scholar] [CrossRef]
  98. Popolo, A.; Morello, S.; Sorrentino, R.; Pinto, A. Antiadrenergic effect of adenosine involves connexin 43 turn-over in H9c2 cells. Eur. J. Pharmacol. 2013, 715, 56–61. [Google Scholar] [CrossRef]
  99. Solan, J.L.; Lampe, P.D. Src Regulation of Cx43 Phosphorylation and Gap Junction Turnover. Biomolecules 2020, 10, 1596. [Google Scholar] [CrossRef]
  100. Norris, R.P.; Baena, V.; Terasaki, M. Localization of phosphorylated connexin 43 using serial section immunogold electron microscopy. J. Cell Sci. 2017, 130, 1333–1340. [Google Scholar] [CrossRef]
  101. Huang, G.Y.; Xie, L.J.; Linask, K.L.; Zhang, C.; Zhao, X.Q.; Yang, Y.; Zhou, G.M.; Wu, Y.J.; Marquez-Rosado, L.; McElhinney, D.B.; et al. Evaluating the role of connexin43 in congenital heart disease: Screening for mutations in patients with outflow tract anomalies and the analysis of knock-in mouse models. J. Cardiovasc. Dis. Res. 2011, 2, 206–212. [Google Scholar] [CrossRef] [PubMed]
  102. Brandenburger, T.; Huhn, R.; Galas, A.; Pannen, B.H.; Keitel, V.; Barthel, F.; Bauer, I.; Heinen, A. Remote ischemic preconditioning preserves Connexin 43 phosphorylation in the rat heart in vivo. J. Transl. Med. 2014, 12, 228. [Google Scholar] [CrossRef] [PubMed]
  103. Akar, F.G.; Spragg, D.D.; Tunin, R.S.; Kass, D.A.; Tomaselli, G.F. Mechanisms underlying conduction slowing and arrhythmogenesis in nonischemic dilated cardiomyopathy. Circ. Res. 2004, 95, 717–725. [Google Scholar] [CrossRef] [PubMed]
  104. Burstein, B.; Comtois, P.; Michael, G.; Nishida, K.; Villeneuve, L.; Yeh, Y.H.; Nattel, S. Changes in connexin expression and the atrial fibrillation substrate in congestive heart failure. Circ. Res. 2009, 105, 1213–1222. [Google Scholar] [CrossRef]
  105. Sasano, C.; Honjo, H.; Takagishi, Y.; Uzzaman, M.; Emdad, L.; Shimizu, A.; Murata, Y.; Kamiya, K.; Kodama, I. Internalization and dephosphorylation of connexin43 in hypertrophied right ventricles of rats with pulmonary hypertension. Circ. J. 2007, 71, 382–389. [Google Scholar] [CrossRef]
  106. Pun, R.; Kim, M.H.; North, B.J. Role of Connexin 43 phosphorylation on Serine-368 by PKC in cardiac function and disease. Front. Cardiovasc. Med. 2022, 9, 1080131. [Google Scholar] [CrossRef]
  107. Dunn, C.A.; Lampe, P.D. Injury-triggered Akt phosphorylation of Cx43: A ZO-1-driven molecular switch that regulates gap junction size. J. Cell Sci. 2014, 127, 455–464. [Google Scholar] [CrossRef]
  108. Barreto, B.C.; Neves, M.; Cardoso, C.M.A.; Meira, C.S.; Daltro, P.S.; Figueira, C.P.; Santos, G.C.; Silva, D.N.; Távora, F.; Neto, J.D.S.; et al. The effects of inflammation on connexin 43 in chronic Chagas disease cardiomyopathy. Front. Immunol. 2024, 15, 1440662. [Google Scholar] [CrossRef]
  109. Lampe, P.D.; Cooper, C.D.; King, T.J.; Burt, J.M. Analysis of Connexin43 phosphorylated at S325, S328 and S330 in normoxic and ischemic heart. J. Cell Sci. 2006, 119, 3435–3442. [Google Scholar] [CrossRef]
  110. Rusiecka, O.M.; Montgomery, J.; Morel, S.; Batista-Almeida, D.; Van Campenhout, R.; Vinken, M.; Girao, H.; Kwak, B.R. Canonical and Non-Canonical Roles of Connexin43 in Cardioprotection. Biomolecules 2020, 10, 1225. [Google Scholar] [CrossRef]
  111. Sun, Z.; Yang, Y.; Wu, L.; Talabieke, S.; You, H.; Zheng, Y.; Luo, D. Connexin 43-serine 282 modulates serine 279 phosphorylation in cardiomyocytes. Biochem. Biophys. Res. Commun. 2019, 513, 567–572. [Google Scholar] [CrossRef] [PubMed]
  112. 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] [PubMed]
  113. Remo, B.F.; Qu, J.; Volpicelli, F.M.; Giovannone, S.; Shin, D.; Lader, J.; Liu, F.Y.; Zhang, J.; Lent, D.S.; Morley, G.E.; et al. Phosphatase-resistant gap junctions inhibit pathological remodeling and prevent arrhythmias. Circ. Res. 2011, 108, 1459–1466. [Google Scholar] [CrossRef] [PubMed]
  114. Ai, X.; Yan, J.; Pogwizd, S.M. Serine-threonine protein phosphatase regulation of Cx43 dephosphorylation in arrhythmogenic disorders. Cell. Signal. 2021, 86, 110070. [Google Scholar] [CrossRef]
  115. Zhang, M.; Wang, Z.Z.; Chen, N.H. Connexin 43 Phosphorylation: Implications in Multiple Diseases. Molecules 2023, 28, 4914. [Google Scholar] [CrossRef]
  116. Hood, A.R.; Ai, X.; Pogwizd, S.M. Regulation of cardiac gap junctions by protein phosphatases. J. Mol. Cell. Cardiol. 2017, 107, 52–57. [Google Scholar] [CrossRef]
  117. Wang, N.; De Bock, M.; Antoons, G.; Gadicherla, A.K.; Bol, M.; Decrock, E.; Evans, W.H.; Sipido, K.R.; Bukauskas, F.F.; Leybaert, L. Connexin mimetic peptides inhibit Cx43 hemichannel opening triggered by voltage and intracellular Ca2+ elevation. Basic Res. Cardiol. 2012, 107, 304. [Google Scholar] [CrossRef]
  118. Leybaert, L.; De Smet, M.A.; Lissoni, A.; Allewaert, R.; Roderick, H.L.; Bultynck, G.; Delmar, M.; Sipido, K.R.; Witschas, K. Connexin hemichannels as candidate targets for cardioprotective and anti-arrhythmic treatments. J. Clin. Investig. 2023, 133, e168117. [Google Scholar] [CrossRef]
  119. Marsh, S.R.; Williams, Z.J.; Pridham, K.J.; Gourdie, R.G. Peptidic Connexin43 Therapeutics in Cardiac Reparative Medicine. J. Cardiovasc. Dev. Dis. 2021, 8, 52. [Google Scholar] [CrossRef]
  120. De Vuyst, E.; Boengler, K.; Antoons, G.; Sipido, K.R.; Schulz, R.; Leybaert, L. Pharmacological modulation of connexin-formed channels in cardiac pathophysiology. Br. J. Pharmacol. 2011, 163, 469–483. [Google Scholar] [CrossRef]
  121. Ewart, J.L.; Cohen, M.F.; Meyer, R.A.; Huang, G.Y.; Wessels, A.; Gourdie, R.G.; Chin, A.J.; Park, S.M.; Lazatin, B.O.; Villabon, S.; et al. Heart and neural tube defects in transgenic mice overexpressing the Cx43 gap junction gene. Development 1997, 124, 1281–1292. [Google Scholar] [CrossRef] [PubMed]
  122. Sottas, V.; Wahl, C.M.; Trache, M.C.; Bartolf-Kopp, M.; Cambridge, S.; Hecker, M.; Ullrich, N.D. Improving electrical properties of iPSC-cardiomyocytes by enhancing Cx43 expression. J. Mol. Cell. Cardiol. 2018, 120, 31–41. [Google Scholar] [CrossRef] [PubMed]
  123. Bikou, O.; Thomas, D.; Trappe, K.; Lugenbiel, P.; Kelemen, K.; Koch, M.; Soucek, R.; Voss, F.; Becker, R.; Katus, H.A.; et al. Connexin 43 gene therapy prevents persistent atrial fibrillation in a porcine model. Cardiovasc. Res. 2011, 92, 218–225. [Google Scholar] [CrossRef] [PubMed]
  124. Igarashi, T.; Finet, J.E.; Takeuchi, A.; Fujino, Y.; Strom, M.; Greener, I.D.; Rosenbaum, D.S.; Donahue, J.K. Connexin gene transfer preserves conduction velocity and prevents atrial fibrillation. Circulation 2012, 125, 216–225. [Google Scholar] [CrossRef]
  125. Greener, I.D.; Sasano, T.; Wan, X.; Igarashi, T.; Strom, M.; Rosenbaum, D.S.; Donahue, J.K. Connexin43 gene transfer reduces ventricular tachycardia susceptibility after myocardial infarction. J. Am. Coll. Cardiol. 2012, 60, 1103–1110. [Google Scholar] [CrossRef]
  126. Roell, W.; Klein, A.M.; Breitbach, M.; Becker, T.S.; Parikh, A.; Lee, J.; Zimmermann, K.; Reining, S.; Gabris, B.; Ottersbach, A.; et al. Overexpression of Cx43 in cells of the myocardial scar: Correction of post-infarct arrhythmias through heterotypic cell-cell coupling. Sci. Rep. 2018, 8, 7145. [Google Scholar] [CrossRef]
  127. Roell, W.; Lewalter, T.; Sasse, P.; Tallini, Y.N.; Choi, B.R.; Breitbach, M.; Doran, R.; Becher, U.M.; Hwang, S.M.; Bostani, T.; et al. Engraftment of connexin 43-expressing cells prevents post-infarct arrhythmia. Nature 2007, 450, 819–824. [Google Scholar] [CrossRef]
  128. Fernandes, S.; van Rijen, H.V.; Forest, V.; Evain, S.; Leblond, A.L.; Mérot, J.; Charpentier, F.; de Bakker, J.M.; Lemarchand, P. Cardiac cell therapy: Overexpression of connexin43 in skeletal myoblasts and prevention of ventricular arrhythmias. J. Cell. Mol. Med. 2009, 13, 3703–3712. [Google Scholar] [CrossRef]
  129. Fiedorowicz, K.; Wargocka-Matuszewska, W.; Ambrożkiewicz, K.A.; Rugowska, A.; Cheda, Ł.; Fiedorowicz, M.; Zimna, A.; Drabik, M.; Borkowski, S.; Świątkiewicz, M.; et al. Molecular Imaging of Human Skeletal Myoblasts (huSKM) in Mouse Post-Infarction Myocardium. Int. J. Mol. Sci. 2021, 22, 10885. [Google Scholar] [CrossRef]
  130. Gonzalez, J.P.; Ramachandran, J.; Himelman, E.; Badr, M.A.; Kang, C.; Nouet, J.; Fefelova, N.; Xie, L.H.; Shirokova, N.; Contreras, J.E.; et al. Normalization of connexin 43 protein levels prevents cellular and functional signs of dystrophic cardiomyopathy in mice. Neuromuscul. Disord. 2018, 28, 361–372. [Google Scholar] [CrossRef]
  131. Boengler, K.; Dodoni, G.; Rodriguez-Sinovas, A.; Cabestrero, A.; Ruiz-Meana, M.; Gres, P.; Konietzka, I.; Lopez-Iglesias, C.; Garcia-Dorado, D.; Di Lisa, F.; et al. Connexin 43 in cardiomyocyte mitochondria and its increase by ischemic preconditioning. Cardiovasc. Res. 2005, 67, 234–244. [Google Scholar] [CrossRef]
  132. Lu, G.; Haider, H.; Porollo, A.; Ashraf, M. Mitochondria-specific transgenic overexpression of connexin-43 simulates preconditioning-induced cytoprotection of stem cells. Cardiovasc. Res. 2010, 88, 277–286. [Google Scholar] [CrossRef] [PubMed]
  133. Wei, X.; Chang, A.C.H.; Chang, H.; Xu, S.; Xue, Y.; Zhang, Y.; Lei, M.; Chang, A.C.Y.; Zhang, Q. Hypoglycemia-Exacerbated Mitochondrial Connexin 43 Accumulation Aggravates Cardiac Dysfunction in Diabetic Cardiomyopathy. Front. Cardiovasc. Med. 2022, 9, 800185. [Google Scholar] [CrossRef] [PubMed]
  134. Pecoraro, M.; Marzocco, S.; Belvedere, R.; Petrella, A.; Franceschelli, S.; Popolo, A. Simvastatin Reduces Doxorubicin-Induced Cardiotoxicity: Effects beyond Its Antioxidant Activity. Int. J. Mol. Sci. 2023, 24, 7573. [Google Scholar] [CrossRef] [PubMed]
  135. Yi, J.; Chen, K.; Cao, Y.; Wen, C.; An, L.; Tong, R.; Wu, X.; Gao, H. Up-regulated novel-miR-17 promotes hypothermic reperfusion arrhythmias by negatively targeting Gja1 and mediating activation of the PKC/c-Jun signaling pathway. J. Mol. Cell. Cardiol. 2024, 193, 1–10. [Google Scholar] [CrossRef]
  136. Yang, Z.J.; Zhang, W.F.; Jin, Q.Q.; Wu, Z.R.; Du, Y.Y.; Shi, H.; Qu, Z.S.; Han, X.J.; Jiang, L.P. Lactate Contributes to Remote Ischemic Preconditioning-Mediated Protection Against Myocardial Ischemia Reperfusion Injury by Facilitating Autophagy via the AMP-Activated Protein Kinase-Mammalian Target of Rapamycin-Transcription Factor EB-Connexin 43 Axis. Am. J. Pathol. 2024, 194, 1857–1878. [Google Scholar] [CrossRef]
  137. Wu, S.J.; He, R.L.; Zhao, L.; Yu, X.Y.; Jiang, Y.N.; Guan, X.; Chen, Q.Y.; Ren, F.F.; Xie, Z.Y.; Wu, L.P.; et al. Cardiac-Specific Overexpression of Caveolin-1 in Rats With Ischemic Cardiomyopathy Improves Arrhythmogenicity and Cardiac Remodelling. Can. J. Cardiol. 2023, 39, 73–86. [Google Scholar] [CrossRef]
  138. Hu, T.; Duan, R.; Gao, H.; Bai, X.; Huang, X.; Yan, X.; An, L.; Ma, Y.; Chen, R.; Hong, S.; et al. Exosomes from myoblasts induced by hypoxic preconditioning improved ventricular conduction by increasing Cx43 expression in hypothermia ischemia reperfusion hearts. Cytotechnology 2024, 76, 533–546. [Google Scholar] [CrossRef]
  139. Amino, M.; Yoshioka, K.; Fujibayashi, D.; Hashida, T.; Furusawa, Y.; Zareba, W.; Ikari, Y.; Tanaka, E.; Mori, H.; Inokuchi, S.; et al. Year-long upregulation of connexin43 in rabbit hearts by heavy ion irradiation. Am. J. Physiol. Heart Circ. Physiol. 2010, 298, H1014–H1021. [Google Scholar] [CrossRef]
  140. Amino, M.; Yoshioka, K.; Tanabe, T.; Tanaka, E.; Mori, H.; Furusawa, Y.; Zareba, W.; Yamazaki, M.; Nakagawa, H.; Honjo, H.; et al. Heavy ion radiation up-regulates Cx43 and ameliorates arrhythmogenic substrates in hearts after myocardial infarction. Cardiovasc. Res. 2006, 72, 412–421. [Google Scholar] [CrossRef]
  141. Ye, T.; Song, Z.; Zhou, Y.; Liu, Z.; Yu, Y.; Yu, F.; Chu, Y.; Shi, J.; Wang, L.; Zhang, C.; et al. TRPV2 inhibitor tranilast prevents atrial fibrillation in rat models of pulmonary hypertension. Cell Calcium 2024, 117, 102840. [Google Scholar] [CrossRef] [PubMed]
  142. Cardouat, G.; Douard, M.; Bouchet, C.; Roubenne, L.; Kmecová, Z.; Esteves, P.; Brette, F.; Guignabert, C.; Tu, L.; Campagnac, M.; et al. NGF increases Connexin-43 expression and function in pulmonary arterial smooth muscle cells to induce pulmonary artery hyperreactivity. Biomed. Pharmacother. 2024, 174, 116552. [Google Scholar] [CrossRef] [PubMed]
  143. Wang, L.J.; Liu, W.D.; Zhang, L.; Ma, K.T.; Zhao, L.; Shi, W.Y.; Zhang, W.W.; Wang, Y.Z.; Li, L.; Si, J.Q. Enhanced expression of Cx43 and gap junction communication in vascular smooth muscle cells of spontaneously hypertensive rats. Mol. Med. Rep. 2016, 14, 4083–4090. [Google Scholar] [CrossRef] [PubMed]
  144. Fang, Z.; Lu, M.; Huang, R.; Wang, G.; Yushanjiang, F.; Jiang, X.; Li, J. Carnosol prevents cardiac remodeling and ventricular arrhythmias in pressure overload-induced heart failure mice. Phytother. Res. 2024, 38, 3763–3781. [Google Scholar] [CrossRef]
  145. Zhang, L.L.; Chen, G.H.; Tang, R.J.; Xiong, Y.Y.; Pan, Q.; Jiang, W.Y.; Gong, Z.T.; Chen, C.; Li, X.S.; Yang, Y.J. Levosimendan Reverses Cardiac Malfunction and Cardiomyocyte Ferroptosis During Heart Failure with Preserved Ejection Fraction via Connexin 43 Signaling Activation. Cardiovasc. Drugs. Ther. 2024, 38, 705–718. [Google Scholar] [CrossRef]
  146. Chen, X.; Wan, W.; Ran, Q.; Ye, T.; Sun, Y.; Liu, Z.; Liu, X.; Shi, S.; Qu, C.; Zhang, C.; et al. Pinocembrin mediates antiarrhythmic effects in rats with isoproterenol-induced cardiac remodeling. Eur. J. Pharmacol. 2022, 920, 174799. [Google Scholar] [CrossRef]
Figure 1. Interplay between factors regulating amount and localization of Cx43. The Figure illustrates the relationships between factors important for the localization and the amount of Cx43 within cardiomyocytes, and therefore for Cx43-mediated function. The numbered blue boxes (1–4) in the schematic representation of Cx43 indicate transmembrane domains. The green dots represent Cx43 within mitochondria (inner mitochondrial membrane), nuclei and exo-somes, and the orange dot represents Gja1-20k within mitochondria (outer mitochondrial mem-brane). The numbered gray dots represent the positions of some amino acids, which can be phosphorylated by protein kinases. The arrows indicate the interactions between Cx43 localizations and processes that regulate Cx43 function. Please note that the arrows only indicate which factor has an influence on another factor and not whether this influence is activating or inhibiting. For details of how the individual parameters relate to each other in terms of Cx43 localization and function, see the text and tables.
Figure 1. Interplay between factors regulating amount and localization of Cx43. The Figure illustrates the relationships between factors important for the localization and the amount of Cx43 within cardiomyocytes, and therefore for Cx43-mediated function. The numbered blue boxes (1–4) in the schematic representation of Cx43 indicate transmembrane domains. The green dots represent Cx43 within mitochondria (inner mitochondrial membrane), nuclei and exo-somes, and the orange dot represents Gja1-20k within mitochondria (outer mitochondrial mem-brane). The numbered gray dots represent the positions of some amino acids, which can be phosphorylated by protein kinases. The arrows indicate the interactions between Cx43 localizations and processes that regulate Cx43 function. Please note that the arrows only indicate which factor has an influence on another factor and not whether this influence is activating or inhibiting. For details of how the individual parameters relate to each other in terms of Cx43 localization and function, see the text and tables.
Biomolecules 15 00370 g001
Table 1. Direct modification of Cx43 expression and its effects in cardiovascular cells.
Table 1. Direct modification of Cx43 expression and its effects in cardiovascular cells.
ModelInterventionEffectReference
Transgenic miceTransgenic mice constitutively expressing Cx43 under control of the CMV promoterGap-junctional communication ↑
Postnatal viability ↓
Defects in the pulmonary outflow tract and coronary vasculature
[121]
iPSC-CMsAdenoviral transduction or transfection of iPSC-CMs with Cx43 expression plasmidCx43 expression ↑
Intercellular coupling ↑
Action potential upstroke velocity ↑
[122]
Atrial fibrillation in porcine heartsAdenoviral gene transfer of Cx43Cx43 protein ↑
Development of persistent atrial fibrillation ↓
[123]
Atrial fibrillation in porcine heartsAdenoviral gene transfer of Cx43Cx43 expression ↑
Atrial conduction in sham-operated
animals ↔
Atrial fibrillation ↓
Lateralization in atrial fibrillation ↓
[124]
Infarcted porcine myocardiumInjection of Cx43-expressing adenoviruses to the healed scar borderCx43 expression ↑
Conduction velocity ↑
Ventricular tachycardia ↓
[125]
Cryoinjured mouse myocardiumInjection of Cx43-expressing lentiviruses Cx43 expression ↑
Heterotypic cell–cell coupling ↑
Ventricular tachycardia ↓
[126]
Cryoinjured mouse myocardium Engraftment of skeletal myoblasts from transgenic mice expressing Cx43 under control of a skeletal muscle promoterVentricular tachycardia ↓[127]
Infarcted rat myocardium (permanent coronary artery ligation)Injection of skeletal myoblasts transduced with Cx43-expressing lentivirusIntercellular electrical coupling ↑ Arrhythmogenicity ↔[128]
Infarcted mouse (NOD/SCID) myocardium (permanent coronary artery ligation)Delivery of mesenchymal stem cells in combination with human skeletal myoblasts overexpressing Cx43Left or right ventricular ejection fraction, mass of the left or right ventricle ↔[129]
mdx miceCrossing of mdx-mice with heterozygous Cx43-deficient miceNormalization of Cx43 protein levels
Lateralization ↓
Arrhythmia ↓
[130]
Stem cell antigen-1+ cellsTransfection with plasmids for expression of Cx43 fused to a mitochondrial targeting sequenceCx43 expression ↑
Apoptosis ↓
Cell survival following oxygen–glucose deprivation ↑
[132]
Hl1-cellsTransduced to overexpress Cx43Cx43 expression ↑
Respiration ↑
[37]
Streptozotocin-induced diabetic cardio-myopathy in miceOverexpression of mitochondrial Cx43 using AAV2/9Cx43 amount in mitochondria ↑
Cardiac function ↓
Susceptibility to arrhythmias ↑
[133]
Isolated mouse hearts in vitroIschemic preconditioningMitochondrial Gja1-20k ↑[47]
Adenoviral overexpression of Gja1-20kROS formation ↓
Infarct size ↓
AAV2/9: adeno-associated virus serotype 2/9; CMV: cytomegalovirus promoter; iPSC-CM: induced pluripotent stem cell-derived cardiomyocytes; NOD/SCID: nonobese diabetic/severe combined immunodeficiency mice; mdx mice: mice with mutations in the dystrophin gene (a model of Duchenne muscular dystrophy); ↑: increase; ↓: decrease; ↔: unchanged.
Table 2. Interventions to treat cardiovascular diseases and their effects on Cx43 expression.
Table 2. Interventions to treat cardiovascular diseases and their effects on Cx43 expression.
ModelInterventionEffectReference
Porcine myocardiumI/R with ischemic preconditioningMitochondrial Cx43 ↑[131]
H9c2 cellsDoxorubicin-treatmentMitochondrial Cx43 ↑
ROS formation ↑
[134]
Isolated rat hearts in vitroHypothermic I/R
miR-17 ↑
Cx43 ↓
[135]
Hypothermic I/R + inhibition of miR-17Cx43 expression ↑ (level below control)
Arrhythmia ↓
H9c2 cellsHypoxia/reoxygenationCx43 ↓[136]
Hypoxia/reoxygenation + lactateCx43 ↑ (level comparable to control)
Rat hearts in vivoICM by coronary artery ligation for 4 wCx43 ↓[137]
ICM + cardiac specific overexpression of caveolin-1Cx43 ↑ (level comparable to control)
Isolated rat hearts in vitroI/RCx43 ↓[138]
Injection of exosomes from C2C12 cells before I/R Cx43 ↑ (level below control)
Rabbit hearts in vivosingle-targeted heavy ion irradiationCx43 ↑ (from 2 w to 12 m after irradiation)
Cx43 lateralization ↑
[139]
Rabbit hearts in vivoMyocardial infarction by microsphere injection
Cx43 ↓
[140]
Myocardial infarction + single targeted heavy ion irradiation after 2 wCx43 ↑ (level above control)
Cx43 lateralization ↑
Arrhythmia ↓
Rat hearts in vivoMonocrotaline-induced pulmonary hypertensionCx43 ↓[141]
Monocrotaline-induced pulmonary hypertension + tranilastCx43 ↑ (level below control)
Atrial fibrillation ↓
Rat hearts in vivoMonocrotaline-induced pulmonary hypertensionCx43 ↔
Lateralization ↑
[92]
Monocrotaline-induced pulmonary hypertension + 1,8-cineoleCx43 ↔
Lateralization ↔
Rat pulmonary arteriesPulmonary hypertension by chronic hypoxia in vivoCx43 ↑[142]
Rat mesenteric arteriolesSpontaneously hypertensive ratsCx43 ↑[143]
Mouse hearts in vivoHeart failure by transverse aortic constrictionCx43 ↓
[144]
Heart failure + carnosolCx43 ↑ (level below control)
Arrhythmia ↓
Dog hearts in vivoHeart failure by rapid pacingCx43 ↓[77]
Heart failure + renal denervationCx43 ↑ (level below control)
Mouse hearts in vivoHFpEF by high fat diet and L-NAME
HFpEF + levosimendan
Cx43 ↓[145]
Cx43 ↑ (level comparable to control)
Mitochondrial homeostasis ↑
Rat hearts in vivoCardiac remodeling by isoproterenolCx43 ↓[146]
Heart failure + carnosolCx43 ↑ (level comparable to control)
Arrhythmia ↓
HFpEF: heart failure with preserved ejection fraction; ICM: ischemic cardiomyopathy; I/R: ischemia/reperfusion; ROS: reactive oxygen species; ↑: increase; ↓: decrease; ↔: unchanged.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Boengler, K.; Mantuano, B.; Toledano, S.; Binah, O.; Schulz, R. Overexpression of Cx43: Is It an Effective Approach for the Treatment of Cardiovascular Diseases? Biomolecules 2025, 15, 370. https://doi.org/10.3390/biom15030370

AMA Style

Boengler K, Mantuano B, Toledano S, Binah O, Schulz R. Overexpression of Cx43: Is It an Effective Approach for the Treatment of Cardiovascular Diseases? Biomolecules. 2025; 15(3):370. https://doi.org/10.3390/biom15030370

Chicago/Turabian Style

Boengler, Kerstin, Beatrice Mantuano, Shira Toledano, Ofer Binah, and Rainer Schulz. 2025. "Overexpression of Cx43: Is It an Effective Approach for the Treatment of Cardiovascular Diseases?" Biomolecules 15, no. 3: 370. https://doi.org/10.3390/biom15030370

APA Style

Boengler, K., Mantuano, B., Toledano, S., Binah, O., & Schulz, R. (2025). Overexpression of Cx43: Is It an Effective Approach for the Treatment of Cardiovascular Diseases? Biomolecules, 15(3), 370. https://doi.org/10.3390/biom15030370

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