Next Article in Journal
The Influence of Adipocyte Secretome on Selected Metabolic Fingerprints of Breast Cancer Cell Lines Representing the Four Major Breast Cancer Subtypes
Next Article in Special Issue
Advances in the Generation of Constructed Cardiac Tissue Derived from Induced Pluripotent Stem Cells for Disease Modeling and Therapeutic Discovery
Previous Article in Journal
Genetic and Pharmacological YAP Activation Induces Proliferation and Improves Survival in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes
Previous Article in Special Issue
Human ApoE2 Endows Stronger Contractility in Rat Cardiomyocytes Enhancing Heart Function
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Desmosomes in Cell Fate Determination: From Cardiogenesis to Cardiomyopathy

Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
*
Author to whom correspondence should be addressed.
Cells 2023, 12(17), 2122; https://doi.org/10.3390/cells12172122
Submission received: 18 July 2023 / Revised: 16 August 2023 / Accepted: 17 August 2023 / Published: 22 August 2023
(This article belongs to the Special Issue Model Systems and Candidate Genes for Inherited Cardiomyopathies)

Abstract

:
Desmosomes play a vital role in providing structural integrity to tissues that experience significant mechanical tension, including the heart. Deficiencies in desmosomal proteins lead to the development of arrhythmogenic cardiomyopathy (AC). The limited availability of preventative measures in clinical settings underscores the pressing need to gain a comprehensive understanding of desmosomal proteins not only in cardiomyocytes but also in non-myocyte residents of the heart, as they actively contribute to the progression of cardiomyopathy. This review focuses specifically on the impact of desmosome deficiency on epi- and endocardial cells. We highlight the intricate cross-talk between desmosomal proteins mutations and signaling pathways involved in the regulation of epicardial cell fate transition. We further emphasize that the consequences of desmosome deficiency differ between the embryonic and adult heart leading to enhanced erythropoiesis during heart development and enhanced fibrogenesis in the mature heart. We suggest that triggering epi-/endocardial cells and fibroblasts that are in different “states” involve the same pathways but lead to different pathological outcomes. Understanding the details of the different responses must be considered when developing interventions and therapeutic strategies.

1. Cardiogenesis and Mechanical Cues

Cardiogenesis is the orchestrated act of cell proliferation, differentiation, and migration that results in the formation of a resilient and durable contractile organ. The uninterrupted rhythmic contractions and the specific arrangement of cardiomyocytes and non-cardiomyocytes are the essential pillars of cardiac function. Deviation from cardiac cell identity and the canonical cell arrangement therefore not only alters cardiac morphogenesis but also impairs heart function. Our knowledge about the key genes that drive cardiogenesis comes from decades of animal studies [1]. It is increasingly recognized that the genetic profile and signaling molecules are not the only regulators of cardiac cell biology, but that mechanical cues also play a definitive role [2]. For example, the stiffness of the extracellular matrix modulates signaling in cardiomyocytes during development and disease [3,4]. Such multifactorial pathogenesis has been discussed in various cardiomyopathies including the still poorly understood genetic heart disease arrhythmogenic cardiomyopathy (AC).
Fifty percent of AC cases have been linked to mutations in genes that contribute to the formation of desmosomes, which are prominent cell–cell adhesion sites between cardiomyocytes [5,6]. Disease hallmarks are arrhythmia, loss of cardiomyocytes, and progressive formation of fibro–fatty structures, which lead to impairment of heart function and ultimately to heart failure [7]. Several transgenic animal models have been created to study AC pathogenesis [6]. The emerging insights categorize AC as a multifaceted disease involving disruption of mechanical and signaling pathways and a surge of immune responses [8]. This paper does not intend to provide a comprehensive review of AC pathogenesis and its clinical outcomes. Readers are referred to recent excellent reviews [9,10,11,12]. Instead, we will focus on the much less investigated and poorly understood functions of desmosomes in embryogenesis.
Desmosomal proteins are indispensable for early embryogenesis [13,14,15] and cardiogenesis [16,17]. Desmosomal proteins are enriched in intercalated discs of cardiomyocytes, but have been detected at low levels also in non-cardiomyocytes such as epicardial cells [18,19,20] and cardiac mesenchymal cells [21,22,23]. Importantly, cardiac mesenchymal cells also express desmosomal proteins [24] and desmosomal protein deficiency can enhance differentiation of these cells into fibrous or adipose tissue. In a recent study, a role of desmosomes has been suggested in regulation of cardiac mesenchymal cell fate by direct modulation of Ca2+ signaling at the level of gene expression [25].
During the last decades, research in the AC field was dominated by investigating the role of desmosomal proteins in adult cardiomyocytes [8,26]. It, however, remains to be explored whether desmosomal deficiency triggers “primary events” for the renewal or differentiation of cardiac progenitors both in the developing and mature heart. Of note, molecular pathways that are activated during embryonic development can be reactivated in pathological conditions [27]. This review focuses on the role of desmosomal proteins in cardial and epicardial morphogenesis. We begin with an overview of desmosomal proteins and structural differences between embryonic and adult hearts. This is followed by describing the development of cardiac progenitors and the formation of junctions in embryonic hearts. We further summarize current knowledge about the cross-talk between desmosomes and signaling pathways that mediate epicardial cell fate transition. In conclusion, we present a model whereby desmosomal deficiency manifests in different phenotypes depending on the pre-existing status of non-cardiomyocytes.

2. Molecular Structure of Desmosomes

Desmosomes are specialized structures, which support the physical stability and integrity of epithelial and heart tissue. Cardiac desmosomes are formed by clustering of the Ca2+-dependent adhesion molecules (cadherins) desmoglein 2 (Dsg2) and desmocollin 2 (Dsc2) in the plasma membrane. They interact with each other via their extracellular domains to link neighboring cells. Intracellularly, the clustered desmosomal cadherins are connected to plakophilin 2 (Pkp2) and plakoglobin (PG), both of which contain multiple copies of the 42 amino acid-long armadillo repeat. They serve not only as structural linkers in desmosomes but fulfill additional cellular functions impacting adherens junctions, cytoskeletal organization, and gene transcription. The functions of the large cytolinker desmoplakin (Dsp), on the other hand, are much more restricted to desmosomes. Dsp is essential for the linkage between the clustered desmosomal cadherins with their associated armadillo-repeat proteins and the intermediate filament cytoskeleton, which consists of desmin polypeptides in cardiomyocytes (Figure 1A) [28,29]. Although the molecular composition of desmosomes is the same in the embryonic and adult heart, the arrangement of desmosomes differs. A distinctive characteristic of mammalian adult cardiomyocytes is their maturation process, which is initiated postnatally. Round-shaped embryonic cardiomyocytes initially form independent adherens junctions and desmosomes with neighboring cells throughout their entire borders. Concurrent with myofibril elongation and organization, desmosomes and adherens junctions concentrate at the apical surfaces of cardiomyocytes where they serve important mechanical functions [30]. Postnatally, the junctions amalgamate forming hybrid junctions that are composed of tightly integrated desmosomes and adherens junctions, which are in close apposition to gap junctions and membrane channels. This supercomplex has been referred to as area composita and connexome [31,32]. It was suggested that the maturation phase is essential to ensure life-long contraction of cardiomyocytes [31,33].

3. Cardiogenesis

3.1. Contribution of Different Heart Fields

Lineage tracing and anatomical studies revealed that Mesp1+ cardiac progenitors appear in the anterior splanchnic mesoderm layer at the lateral sides of the primitive streak [34]. The cardiac progenitor cells are arranged in two different heart fields, i.e., the first heart field (FHF) and the second heart field (SHF). They develop in a temporally and spatially distinct manner and participate in the formation of discrete parts of the heart [35], each with a unique gene profile [36,37]. The FHF comprises cardiac progenitors, which appear first and differentiate rapidly. These fast-differentiating cells express the transcription factors NKX2-5, Tbx5, Hand1, and GATA4 as well as the chromatin regulatory factor BAF60c. Together, these factors drive the expression of sarcomeric proteins [38]. Morphologically, the cells in the lateral regions of the FHF migrate toward the midline and form a tubular heart at embryonic day (E) 8 in mice and during the third week of gestation in humans. Slowly differentiating but rapidly proliferating cardiac progenitors appear posterior to the FHF to form the SHF. They express the transcription factors Isl-1 and TBX-1 [39,40,41]. Following heart tube formation, the SHF cells migrate into the heart tube from the atrial and venous poles. The coordinated movement of SHF and FHF cells is facilitated via receptor-ligand interaction and leads to the elongation of the heart tube and the formation of the right ventricle and outflow tract [42].
The lumen of the heart is lined by endocardial cells, which are a unique type of endothelial cells, both in terms of their origin and differentiation capacities [43]. Endocardial cells appear at the same time as cardiac progenitors with whom they share a common origin [44]. The endocardial and myocardial cell layers are separated by an extracellular matrix, which is composed of hyaluronic acid, fibronectin, collagen IV, and proteoglycans [45,46,47,48]. Endocardial cells can undergo endothelial-to-mesenchymal transition and transform into mesenchymal endocardial cushion cells, which subsequently remodel to form cardiac valves and separate the outflow track into the pulmonary artery and aorta. Endocardial cells exhibit remarkable plasticity differentiating into various lineages including endothelial cells of the capillary network, adipocytes, fibroblasts, and hematopoietic cells [44,49].
At mid-gestation (E10.5), two types of cardiomyocytes are present in the ventricular myocardium. The majority (86%) are immature cardiomyocytes, which are primarily located in the compact myocardium [50]. They have a spherical shape and contain little cytoplasm with loosely arranged myofilaments. The remaining cardiomyocytes (14%) are elongated and have regularly arranged sarcomeres [50]. They are generated from the compact myocardium by proliferation and delamination of cells and make up the trabecular myocardium [51]. Adherens and desmosomal junctions are present in both cardiomyocyte types. But the spherical-shaped myocytes contain intercellular junctions along all sides whereas the trabecular myocytes restrict the junctions to intercalated discs for the most part (Figure 1B,C).

3.2. Development of Epicardium and Epicardial-Derived Cells

Epicardial cells emerge from cell clusters that are referred to as the proepicardium (PE). The PE is located close to the liver primordium and sinus venosus. PE cells migrate toward the looped heart tube around E9.5, attach to the myocardium, and form the epicardial cell layer [52]. Prior to the attachment of epicardial cells to the myocardium at E9.5, the outer layer of cardiomyocytes is covered by a thin and patchy layer of fibronectin, laminin, and collagen IV, generating a basement membrane-like structure [53]. Epicardial cells initially contact myocytes directly through their α4 integrin receptor (CD49d), which binds VCAM-1 on adjacent cardiomyocytes [54]. Later, an extracellular matrix builds up between both cell layers. After formation of the epicardial layer, around E12 some epicardial cells undergo epithelial to mesenchymal transition (EMT), migrate into the sub-epicardial space and subsequently into the myocardium, where they can differentiate into fibroblasts, endothelial cells, and the smooth muscle cells surrounding arteries [55,56] as well as into mesenchymal stem cells [57].
At the same time, paracrine communication between epicardial and myocardial cells promotes myocardial growth [58]. In accordance, co-culture of embryonic epicardium-derived cells enhances the proliferation, maturation, and alignment of cardiomyocytes in vitro. This cross-talk involves increased expression of Cx43, N-cadherin, focal adhesion kinase, and sarcoplasmic reticulum Ca2+ ATPase [59]. Similarly, the promotion of structural and metabolic maturation of cardiomyocytes has been observed in co-cultures of cardiac fibroblasts (the derivatives of epicardial cells) with cardiomyocytes [60].

4. Development of Intercellular Junctions in Embryonic Cardiomyocytes

N-cadherin is the main cadherin of classical adherens junctions that are formed in cardiac progenitors as they appear in the cardiac crescent [61,62]. Immature and spherical cardiomyocytes establish multiple contacts with neighboring cells through N-cadherin-based junctions and maintain them as they are required for cardiomyocyte differentiation and organization [30,63,64,65]. During cardiogenesis, the localization of N-cadherins to intercalated discs is closely followed by the appearance of desmosomes [66]. The formation of adherens junctions is a prerequisite for desmosome formation. Loss of N-cadherin, therefore, destabilizes intercalated discs and desmosomes in adult cardiomyocytes [67]. It is even more detrimental during cardiogenesis inducing the formation of a disorganized myocardium with adhesion-deficient cardiomyocytes, reduced trabeculation, loss of cell polarity, and outward migration of cardiomyocytes to the pericardial cavity [51,68].
After the establishment of adherens junctions, the spherical cardiomyocytes of the compact myocardium establish additional contact points by forming desmosomal adhesions with neighboring cardiomyocytes. Desmosomal proteins such as Pkp2 and Dsp can be identified in cardiomyocytes as early as embryonic day 9.5 [17,62]. This goes along with prominent shape changes from spheroidal to elongated. After the appearance of desmosomes, gap junctions are formed and expanded in the plasma membrane [62]. In mature cardiomyocytes of the adult, desmosomes are clustered together with other junctions in the intercalated disc region leaving the lateral membranes desmosome-free.
In the following section, we will summarize the evidence, which indicates that a deficiency of desmosomal proteins can alter the fate of epicardial cells and will describe pathways that may play a role in converting this fate.

5. The Impact of Desmosomal Proteins on Cardiac Morphogenesis

Of all desmosomal proteins, Dsp and Dsg2 were found to be essential for early embryogenesis [13,69]. In particular, Dsp was identified as a common factor mediating both reprograming and regeneration, and its deficiency delayed in vitro reprogramming [70]. Rescue of Dsp mutation in extraembryonic tissue results in the progression of embryonic development up to E10, but then the mutants show drastic abnormalities in the organization of the myocardium with reduced mass in spite of unaltered apoptosis levels [16]. Subsequent studies revealed the importance of desmosomes for heart morphogenesis in several zebrafish and mouse models (Table 1; [16,17,71,72,73,74]).
Mutations in Jup (gene encoding PG) and Pkp2 induce similar cardiogenesis abnormalities [74,76,77]. The hallmark of the Jup−/ phenotype is edema with the appearance of blood cells in the pericardial cavity along with reduced cell mass in the compact and trabecular myocardium. Despite normal development up to E9.5, Jup−/, embryos die between E10.5 and E12.5. This time window corresponds to myocardial growth, the formation of epicardial cells, and epicardial to mesenchymal transition—events that are regulated by cross-talk between myocardial and epicardial cells. Similarly, Pkp2 expression is indispensable for heart development in zebrafish and mice [74,78,79]. Immunostaining of Pkp2 null mice showed perturbed localization of desmosomal proteins in the myocardium [17], suggesting a key role of Pkp2 in stabilizing desmosome structure.
Despite the inability to document a rupture in Pkp2 or Jup mutants, accumulating blood in the pericardial cavity was interpreted as leakage of blood due to weak cardiomyocyte junctions [17,74]. Our detailed histological assessment of embryonic hearts with Dsg2 mutation suggests that rupture is not the cause of pericardial blood cell accumulation in most instances [72]. The analyses revealed that desmosome deficiency does not result in myocyte rupture or leakiness but that the accumulating blood in the pericardial cavity is caused by differentiation of endocardial/epicardial cells into hematopoietic Runx1+ cells, which excessively proliferate and subsequently transmigrate into the pericardial cavity [72]. Interestingly, the loss of cuboidal endothelial cell morphology and reduction of cell junctions between endocardial cells have also been observed in zebrafish treated with Jup morpholinos [73]. The reported blood-filled pericardial cavities in Dsg2, Pkp2, and Jup mutants suggest that the transition of epi-/endocardial cells to a hemogenic fate may be a common consequence of perturbed desmosome formation during cardiogenesis. Whether this transition of epi-/endocardial fate is a consequence of altered myocardium integrity or a direct consequence of desmosome deficiency in epicardial cells remains to be clarified.

6. Epithelial to Mesenchymal Transition in Desmosome Deficient Models

A wide spectrum of signaling molecules including the TGF-β superfamily, FGF, Notch, and Wnt mediate the cross-talk of epicardial and myocardial cells and prompt EMT (comprehensive recent reviews in [27,80,81]). Here, we will focus on activators of EMT that have been specifically associated with desmosome deficiency (Table 2). Furthermore, we will describe the divergent outcomes of desmosome deficiency/EMT between embryonic and adult hearts.

6.1. Desmosomes Communicate with Gap Junctions in the Regulation of Epicardial EMT

The interaction between desmosomes and the gap junction protein connexin 43 (Cx43) plays a role in maintaining the stability of plasma membrane-bound Cx43 in cardiomyocytes and epicardium of neonatal rats, albeit through different mechanisms [83,85]. It was observed that reducing Pkp2 levels using siRNA leads to a decrease in Cx43 plasma membrane localization [83]. In cardiomyocytes, super-resolution microscopy and proximity ligation assays demonstrated the localization of Pkp2 in Cx43-containing gap junction plaques indicating that Pkp2 has a direct role in Cx43 trafficking [86]. Furthermore, Dsp stabilizes Cx43 in the plasma membrane by inhibiting the activation of ERK1/2-MAPK and the phosphorylation of S278/282 residues in Cx43. This inhibition prevents clathrin-mediated internalization of Cx43, subsequently averting its lysosomal degradation [85].
During cardiogenesis, the loss of Cx43 results in reduced epicardial adhesion to the extracellular matrix as indicated by diminished ZO-1 immunostaining and smaller sized focal adhesions [87]. Morphologically, this manifests as detachment of epicardium from the myocardium, referred to as a “blistering phenotype” [87]. Mechanistically, reduction of Cx43 in epicardial cells is associated with alteration in actin cytoskeletal organization, increased migration speed, but loss of migration directionality, all of which reflects impaired EMT [87]. The in vivo consequence of impaired EMT in Cx43 deficiency is the abnormal development of coronary arteries in mice [88].
Numerous studies documented the cross-talk between Cx43 and the TGF-β signaling pathway, which includes the modulation of SMAD4 nuclear localization [89] as well as signaling involving Snail-1 [90,91,92]. However, some of these reports present conflicting results. For instance, blocking Cx43 in osteoblasts and retinal pigment epithelial cells inhibits TGF-β2-mediated EMT [93,94], while Cx43 siRNA leads to nuclear localization of SMAD-4 and activation of TGF-β targets in HL-1 cells [89]. Further exploration is required to understand the impact of desmosome deficiency on gap junction stability and activation of downstream pathways that influence the propensity of epicardial cells to undergo EMT in embryonic hearts.

6.2. Desmosome-TGF-β Cross-Talk in the Regulation of Epicardium Development

TGF-β signaling is one of the key drivers of cardiogenesis, and it is intriguing to explore whether desmosomal proteins regulate TGF-β signaling during cardiogenesis. TGF-β signaling is a well-recognized pathway in the regulation of EMT during development [95]. Given the intricate nature of the upstream activators of TGF-β signaling and its downstream targets, uncovering the potential interaction between desmosomes and regulators of TGF-β signaling is a nontrivial undertaking. But multiple pieces of evidence point to desmosome-TGF-β cross-talk. A deficiency of Pkp2 in cultured neonatal cardiomyocytes activates TGF-β1 expression via activation of p38 MAP kinase, leading to the expression of profibrotic genes [84]. In two murine AC disease models with constitutive or myocardial-restricted Dsg2 deficiency Tgf-β mRNA isoforms were upregulated [82]. In another AC disease model caused by adhesion-deficient Dsg2 activation of the profibrotic TGF-β pathway was reported and linked to the enrichment of ITG αV/β6 in intercalated discs [75]. Interestingly, epicardium-specific depletion of Dsp was shown to be sufficient to induce activation of the FGF2 and TGF-β1 pathways [21]. Altered TGF-β signaling in endothelial cells is most likely irrelevant since desmosomal proteins are not expressed in endothelial cells. Nevertheless, there is considerable ambiguity about the involvement of endocardial/cardiac endothelial cells in desmosome-deficient pathogenesis.
The previous literature, which reported activation of TGF-β signaling in epicardial-derived cells [21] is particularly interesting with regard to the observed expansion of blood clusters in embryonic Dsg- and Pkp-mutant murine myocardium [17,72,76]. Another complementary result is the expansion of blood cells in the pericardium of embryos with VCAM-1 null mutation [54]. In accordance, the null mutation of the myocardial VCAM1 ligand integrin β4 (ITG-β4) results in a similar phenotype [96]. VCAM-1 antagonizes TGF-β stimulated weakening of intercellular junctions via modulation of stress fibers and diminishing Rho activity [97]. Therefore, the loss of epicardial cell attachment and expansion of blood cells in the pericardium can be explained by the activation of TGF-β signaling in the absence of VCAM-1 and ITG-β4. In epi/endocardial cells, TGF-β can induce EMT through multiple downstream signaling pathways involving phosphatidylinositol-3 kinase [98], wnt/β-catenin signaling [99], Rho [100], SMAD signaling [101], and the Jagged-1/Notch pathway [102,103]. Notably, epicardial activation of Notch signaling can induce subepicardial expansion of erythrocytes (known as epicardial blistering) in embryonic hearts [104]—a phenotype that is similar to that in Dsg2 mutant embryos [72]. Another significant study focused on the generation of human PKP2 null iPSC-derived epicardial cells. This study revealed that the long-term consequence of desmosome deficiency in epicardial cells includes the enrichment of mesenchymal markers such as CD73 and CD29. Additionally, there is a loss of Wt-1 expression, an elevation of markers associated with adipose and connective tissue, and activation of the transcription factor TFAP2A, which mediates EMT [22]. These findings are further supported by the validation of TFAP2A expression in cardiac biopsies of AC patients [22]. Importantly and in line with our argument, TFAP2A can activate TGF-β mediated EMT [105,106].

6.3. Desmosomes and Modulation of YAP/TAZ Signaling

Another critical pathway involved in heart development and cardiomyopathy is the YAP/TAZ pathway, which has been extensively reviewed (e.g., [107]). This pathway is activated upon disruption of cell polarity, loss of cell–cell contacts, mechanical stress, and increased extracellular matrix stiffness [108]. Similar situations are encountered in desmosome deficiency. In accordance, altered YAP signaling has been documented in AC animal models [109,110]. But, there exists still a considerable knowledge gap linking the YAP/TAZ pathway with desmosomal adhesion during cardiogenesis. Analyses conducted in different systems suggest that Cx43 expression counteracts YAP signaling. Specifically, the downregulation of Cx43 in astrocytes facilitated YAP nuclear translocation and regulated mesenchymal transition [111]. Moreover, the YAP/TAZ pathway has been found to converge with SMAD-mediated TGF-β signaling [112,113].

7. Conclusions and Future Directions

Taken together, we conclude that the combination of mechanical cues and signaling factors provides a microenvironment that promotes the escape of epicardial cells from their restricted fate. In embryonic hearts, desmosome deficiency-induced EMT leads to unrestricted expansion of hematopoietic stem cells [72]. Conversely, in adult hearts, the same mutation activates profibrotic pathways and the expansion of fibroblasts (Figure 2) [114]. We, therefore, suggest that the impact of desmosome deficiency in the heart depends primarily on the intrinsic, development-specific potentials of cells encountered in the heart including, besides cardiomyocytes, epi-, and endocardial cells.

Author Contributions

Conceptualization, H.M. and R.E.L.; writing—original draft preparation, H.M. and M.D.B.; writing—review and editing, H.M. and R.E.L.; visualization, H.M. and M.D.B.; supervision, R.E.L.; funding acquisition, H.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research project and APC were funded by the START Program of the Faculty of Medicine RWTH Aachen University, grant number [81-02.04.2020.A323].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank Adam Breitscheidel for his contribution to creating the graphic illustrations. We also thank Sabine Eisner for providing electron microscopy micrographs and Claudia Krusche for helpful discussions and for providing the image of adult heart fibrosis. We are also thankful for the support received from the START Funding of the faculty of medicine at RWTH University, which was granted to H.M.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Paige, S.L.; Plonowska, K.; Xu, A.; Wu, S.M. Molecular regulation of cardiomyocyte differentiation. Circ. Res. 2015, 116, 341–353. [Google Scholar] [CrossRef] [PubMed]
  2. Bartman, T.; Hove, J. Mechanics and function in heart morphogenesis. Dev. Dyn. 2005, 233, 373–381. [Google Scholar] [CrossRef] [PubMed]
  3. Gaetani, R.; Zizzi, E.A.; Deriu, M.A.; Morbiducci, U.; Pesce, M.; Messina, E. When Stiffness Matters: Mechanosensing in Heart Development and Disease. Front. Cell Dev. Biol. 2020, 8, 334. [Google Scholar] [CrossRef] [PubMed]
  4. Munch, J.; Abdelilah-Seyfried, S. Sensing and Responding of Cardiomyocytes to Changes of Tissue Stiffness in the Diseased Heart. Front. Cell Dev. Biol. 2021, 9, 642840. [Google Scholar] [CrossRef]
  5. Xu, T.; Yang, Z.; Vatta, M.; Rampazzo, A.; Beffagna, G.; Pilichou, K.; Scherer, S.E.; Saffitz, J.; Kravitz, J.; Zareba, W.; et al. Compound and digenic heterozygosity contributes to arrhythmogenic right ventricular cardiomyopathy. J. Am. Coll. Cardiol. 2010, 55, 587–597. [Google Scholar] [CrossRef] [PubMed]
  6. Gerull, B.; Brodehl, A. Genetic Animal Models for Arrhythmogenic Cardiomyopathy. Front. Physiol. 2020, 11, 624. [Google Scholar] [CrossRef]
  7. Corrado, D.; Basso, C.; Judge, D.P. Arrhythmogenic Cardiomyopathy. Circ. Res. 2017, 121, 784–802. [Google Scholar] [CrossRef]
  8. Gao, S.; Puthenvedu, D.; Lombardi, R.; Chen, S.N. Established and Emerging Mechanisms in the Pathogenesis of Arrhythmogenic Cardiomyopathy: A Multifaceted Disease. Int. J. Mol. Sci. 2020, 21, 6320. [Google Scholar] [CrossRef]
  9. Coscarella, I.L.; Landim-Vieira, M.; Pinto, J.R.; Chelko, S.P. Arrhythmogenic Cardiomyopathy: Exercise Pitfalls, Role of Connexin-43, and Moving beyond Antiarrhythmics. Int. J. Mol. Sci. 2022, 23, 8753. [Google Scholar] [CrossRef]
  10. Thiene, G.; Basso, C.; Pilichou, K.; Bueno Marinas, M. Desmosomal Arrhythmogenic Cardiomyopathy: The Story Telling of a Genetically Determined Heart Muscle Disease. Biomedicines 2023, 11, 2018. [Google Scholar] [CrossRef]
  11. Reisqs, J.B.; Moreau, A.; Sleiman, Y.; Boutjdir, M.; Richard, S.; Chevalier, P. Arrhythmogenic cardiomyopathy as a myogenic disease: Highlights from cardiomyocytes derived from human induced pluripotent stem cells. Front. Physiol. 2023, 14, 1191965. [Google Scholar] [CrossRef] [PubMed]
  12. Peretto, G.; Sommariva, E.; Di Resta, C.; Rabino, M.; Villatore, A.; Lazzeroni, D.; Sala, S.; Pompilio, G.; Cooper, L.T. Myocardial Inflammation as a Manifestation of Genetic Cardiomyopathies: From Bedside to the Bench. Biomolecules 2023, 13, 646. [Google Scholar] [CrossRef] [PubMed]
  13. Eshkind, L.; Tian, Q.; Schmidt, A.; Franke, W.W.; Windoffer, R.; Leube, R.E. Loss of desmoglein 2 suggests essential functions for early embryonic development and proliferation of embryonal stem cells. Eur. J. Cell Biol. 2002, 81, 592–598. [Google Scholar] [CrossRef]
  14. Den, Z.; Cheng, X.; Merched-Sauvage, M.; Koch, P.J. Desmocollin 3 is required for pre-implantation development of the mouse embryo. J. Cell Sci. 2006, 119, 482–489. [Google Scholar] [CrossRef] [PubMed]
  15. Park, J.; Son, Y.; Lee, N.G.; Lee, K.; Lee, D.G.; Song, J.; Lee, J.; Kim, S.; Cho, M.J.; Jang, J.H.; et al. DSG2 Is a Functional Cell Surface Marker for Identification and Isolation of Human Pluripotent Stem Cells. Stem. Cell Rep. 2018, 11, 115–127. [Google Scholar] [CrossRef]
  16. Gallicano, G.I.; Bauer, C.; Fuchs, E. Rescuing desmoplakin function in extra-embryonic ectoderm reveals the importance of this protein in embryonic heart, neuroepithelium, skin and vasculature. Development 2001, 128, 929–941. [Google Scholar] [CrossRef]
  17. Grossmann, K.S.; Grund, C.; Huelsken, J.; Behrend, M.; Erdmann, B.; Franke, W.W.; Birchmeier, W. Requirement of plakophilin 2 for heart morphogenesis and cardiac junction formation. J. Cell Biol. 2004, 167, 149–160. [Google Scholar] [CrossRef] [PubMed]
  18. Viragh, S.; Gittenberger-de Groot, A.C.; Poelmann, R.E.; Kalman, F. Early development of quail heart epicardium and associated vascular and glandular structures. Anat. Embryol. 1993, 188, 381–393. [Google Scholar] [CrossRef]
  19. Eroglu, E.; Yen, C.Y.T.; Tsoi, Y.L.; Witman, N.; Elewa, A.; Joven Araus, A.; Wang, H.; Szattler, T.; Umeano, C.H.; Sohlmer, J.; et al. Epicardium-derived cells organize through tight junctions to replenish cardiac muscle in salamanders. Nat. Cell Biol. 2022, 24, 645–658. [Google Scholar] [CrossRef]
  20. Matthes, S.A.; Taffet, S.; Delmar, M. Plakophilin-2 and the migration, differentiation and transformation of cells derived from the epicardium of neonatal rat hearts. Cell Commun. Adhes. 2011, 18, 73–84. [Google Scholar] [CrossRef]
  21. Yuan, P.; Cheedipudi, S.M.; Rouhi, L.; Fan, S.; Simon, L.; Zhao, Z.; Hong, K.; Gurha, P.; Marian, A.J. Single-Cell RNA Sequencing Uncovers Paracrine Functions of the Epicardial-Derived Cells in Arrhythmogenic Cardiomyopathy. Circulation 2021, 143, 2169–2187. [Google Scholar] [CrossRef] [PubMed]
  22. Kohela, A.; van Kampen, S.J.; Moens, T.; Wehrens, M.; Molenaar, B.; Boogerd, C.J.; Monshouwer-Kloots, J.; Perini, I.; Goumans, M.J.; Smits, A.M.; et al. Epicardial differentiation drives fibro-fatty remodeling in arrhythmogenic cardiomyopathy. Sci. Transl. Med. 2021, 13, eabf2750. [Google Scholar] [CrossRef] [PubMed]
  23. Reant, P.; Hauer, A.D.; Castelletti, S.; Pantazis, A.; Rosmini, S.; Cheang, M.H.; Peyrou, J.; Tome-Esteban, M.; Syrris, P.; Lafitte, S.; et al. Epicardial myocardial strain abnormalities may identify the earliest stages of arrhythmogenic cardiomyopathy. Int. J. Cardiovasc. Imaging 2016, 32, 593–601. [Google Scholar] [CrossRef] [PubMed]
  24. Lombardi, R.; Chen, S.N.; Ruggiero, A.; Gurha, P.; Czernuszewicz, G.Z.; Willerson, J.T.; Marian, A.J. Cardiac Fibro-Adipocyte Progenitors Express Desmosome Proteins and Preferentially Differentiate to Adipocytes Upon Deletion of the Desmoplakin Gene. Circ. Res. 2016, 119, 41–54. [Google Scholar] [CrossRef]
  25. Maione, A.S.; Faris, P.; Iengo, L.; Catto, V.; Bisonni, L.; Lodola, F.; Negri, S.; Casella, M.; Guarino, A.; Polvani, G.; et al. Ca(2+) dysregulation in cardiac stromal cells sustains fibro-adipose remodeling in Arrhythmogenic Cardiomyopathy and can be modulated by flecainide. J. Transl. Med. 2022, 20, 522. [Google Scholar] [CrossRef]
  26. Gerull, B.; Brodehl, A. Insights Into Genetics and Pathophysiology of Arrhythmogenic Cardiomyopathy. Curr. Heart Fail. Rep. 2021, 18, 378–390. [Google Scholar] [CrossRef]
  27. Quijada, P.; Trembley, M.A.; Small, E.M. The Role of the Epicardium During Heart Development and Repair. Circ. Res. 2020, 126, 377–394. [Google Scholar] [CrossRef]
  28. Holthofer, B.; Windoffer, R.; Troyanovsky, S.; Leube, R.E. Structure and function of desmosomes. Int. Rev. Cytol. 2007, 264, 65–163. [Google Scholar] [CrossRef]
  29. Hegazy, M.; Perl, A.L.; Svoboda, S.A.; Green, K.J. Desmosomal Cadherins in Health and Disease. Annu. Rev. Pathol. 2022, 17, 47–72. [Google Scholar] [CrossRef]
  30. Hirschy, A.; Schatzmann, F.; Ehler, E.; Perriard, J.C. Establishment of cardiac cytoarchitecture in the developing mouse heart. Dev. Biol. 2006, 289, 430–441. [Google Scholar] [CrossRef]
  31. Franke, W.W.; Borrmann, C.M.; Grund, C.; Pieperhoff, S. The area composita of adhering junctions connecting heart muscle cells of vertebrates. I. Molecular definition in intercalated disks of cardiomyocytes by immunoelectron microscopy of desmosomal proteins. Eur. J. Cell Biol. 2006, 85, 69–82. [Google Scholar] [CrossRef] [PubMed]
  32. Leo-Macias, A.; Liang, F.X.; Delmar, M. Ultrastructure of the intercellular space in adult murine ventricle revealed by quantitative tomographic electron microscopy. Cardiovasc. Res. 2015, 107, 442–452. [Google Scholar] [CrossRef] [PubMed]
  33. Pieperhoff, S.; Franke, W.W. The area composita of adhering junctions connecting heart muscle cells of vertebrates - IV: Coalescence and amalgamation of desmosomal and adhaerens junction components - late processes in mammalian heart development. Eur. J. Cell Biol. 2007, 86, 377–391. [Google Scholar] [CrossRef] [PubMed]
  34. Buckingham, M.; Meilhac, S.; Zaffran, S. Building the mammalian heart from two sources of myocardial cells. Nat. Rev. Genet. 2005, 6, 826–835. [Google Scholar] [CrossRef] [PubMed]
  35. Martin-Puig, S.; Wang, Z.; Chien, K.R. Lives of a heart cell: Tracing the origins of cardiac progenitors. Cell Stem Cell 2008, 2, 320–331. [Google Scholar] [CrossRef] [PubMed]
  36. Lescroart, F.; Wang, X.; Lin, X.; Swedlund, B.; Gargouri, S.; Sanchez-Danes, A.; Moignard, V.; Dubois, C.; Paulissen, C.; Kinston, S.; et al. Defining the earliest step of cardiovascular lineage segregation by single-cell RNA-seq. Science 2018, 359, 1177–1181. [Google Scholar] [CrossRef]
  37. Ma, Q.; Zhou, B.; Pu, W.T. Reassessment of Isl1 and Nkx2-5 cardiac fate maps using a Gata4-based reporter of Cre activity. Dev. Biol. 2008, 323, 98–104. [Google Scholar] [CrossRef]
  38. Lopez-Sanchez, C.; Garcia-Martinez, V. Molecular determinants of cardiac specification. Cardiovasc. Res. 2011, 91, 185–195. [Google Scholar] [CrossRef]
  39. Bruneau, B.G.; Logan, M.; Davis, N.; Levi, T.; Tabin, C.J.; Seidman, J.G.; Seidman, C.E. Chamber-specific cardiac expression of Tbx5 and heart defects in Holt-Oram syndrome. Dev. Biol. 1999, 211, 100–108. [Google Scholar] [CrossRef]
  40. Cai, C.L.; Liang, X.; Shi, Y.; Chu, P.H.; Pfaff, S.L.; Chen, J.; Evans, S. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev. Cell 2003, 5, 877–889. [Google Scholar] [CrossRef]
  41. Galdos, F.X.; Wu, S.M. Single-Cell Delineation of Who's on First and Second Heart Fields During Development. Circ. Res. 2019, 125, 411–413. [Google Scholar] [CrossRef] [PubMed]
  42. Xiong, H.; Luo, Y.; Yue, Y.; Zhang, J.; Ai, S.; Li, X.; Wang, X.; Zhang, Y.L.; Wei, Y.; Li, H.H.; et al. Single-Cell Transcriptomics Reveals Chemotaxis-Mediated Intraorgan Crosstalk During Cardiogenesis. Circ. Res. 2019, 125, 398–410. [Google Scholar] [CrossRef] [PubMed]
  43. Misfeldt, A.M.; Boyle, S.C.; Tompkins, K.L.; Bautch, V.L.; Labosky, P.A.; Baldwin, H.S. Endocardial cells are a distinct endothelial lineage derived from Flk1+ multipotent cardiovascular progenitors. Dev. Biol. 2009, 333, 78–89. [Google Scholar] [CrossRef] [PubMed]
  44. Dye, B.; Lincoln, J. The Endocardium and Heart Valves. Cold Spring Harb. Perspect. Biol. 2020, 12, a036723. [Google Scholar] [CrossRef] [PubMed]
  45. Silva, A.C.; Pereira, C.; Fonseca, A.; Pinto-do, O.P.; Nascimento, D.S. Bearing My Heart: The Role of Extracellular Matrix on Cardiac Development, Homeostasis, and Injury Response. Front. Cell Dev. Biol. 2020, 8, 621644. [Google Scholar] [CrossRef]
  46. Rienks, M.; Papageorgiou, A.P.; Frangogiannis, N.G.; Heymans, S. Myocardial extracellular matrix: An ever-changing and diverse entity. Circ. Res. 2014, 114, 872–888. [Google Scholar] [CrossRef]
  47. Jallerat, Q.; Feinberg, A.W. Extracellular Matrix Structure and Composition in the Early Four-Chambered Embryonic Heart. Cells 2020, 9, 285. [Google Scholar] [CrossRef]
  48. Kalman, F.; Viragh, S.; Modis, L. Cell surface glycoconjugates and the extracellular matrix of the developing mouse embryo epicardium. Anat. Embryol. 1995, 191, 451–464. [Google Scholar] [CrossRef]
  49. Zhang, H.; Lui, K.O.; Zhou, B. Endocardial Cell Plasticity in Cardiac Development, Diseases and Regeneration. Circ. Res. 2018, 122, 774–789. [Google Scholar] [CrossRef]
  50. Zhang, F.; Pasumarthi, K.B. Ultrastructural and immunocharacterization of undifferentiated myocardial cells in the developing mouse heart. J. Cell Mol. Med. 2007, 11, 552–560. [Google Scholar] [CrossRef]
  51. Wu, M. Mechanisms of Trabecular Formation and Specification During Cardiogenesis. Pediatr. Cardiol. 2018, 39, 1082–1089. [Google Scholar] [CrossRef] [PubMed]
  52. Männer, J.; Pérez-Pomares, J.M.; Macías, D.; Muñoz-Chápuli, R. The origin, formation and developmental significance of the epicardium: A review. Cells Tissues Organs 2001, 169, 89–103. [Google Scholar] [CrossRef] [PubMed]
  53. Nahirney, P.C.; Mikawa, T.; Fischman, D.A. Evidence for an extracellular matrix bridge guiding proepicardial cell migration to the myocardium of chick embryos. Dev. Dyn. 2003, 227, 511–523. [Google Scholar] [CrossRef] [PubMed]
  54. Yang, J.T.; Rayburn, H.; Hynes, R.O. Cell adhesion events mediated by alpha 4 integrins are essential in placental and cardiac development. Development 1995, 121, 549–560. [Google Scholar] [CrossRef]
  55. Cao, Y.; Duca, S.; Cao, J. Epicardium in Heart Development. Cold Spring Harb. Perspect. Biol. 2020, 12, a037192. [Google Scholar] [CrossRef]
  56. Dettman, R.W.; Denetclaw, W., Jr.; Ordahl, C.P.; Bristow, J. Common epicardial origin of coronary vascular smooth muscle, perivascular fibroblasts, and intermyocardial fibroblasts in the avian heart. Dev. Biol. 1998, 193, 169–181. [Google Scholar] [CrossRef]
  57. Chong, J.J.; Chandrakanthan, V.; Xaymardan, M.; Asli, N.S.; Li, J.; Ahmed, I.; Heffernan, C.; Menon, M.K.; Scarlett, C.J.; Rashidianfar, A.; et al. Adult cardiac-resident MSC-like stem cells with a proepicardial origin. Cell Stem Cell 2011, 9, 527–540. [Google Scholar] [CrossRef]
  58. Boezio, G.L.M.; Zhao, S.; Gollin, J.; Priya, R.; Mansingh, S.; Guenther, S.; Fukuda, N.; Gunawan, F.; Stainier, D.Y.R. The developing epicardium regulates cardiac chamber morphogenesis by promoting cardiomyocyte growth. Dis. Model Mech. 2023, 16, dmm049571. [Google Scholar] [CrossRef]
  59. Weeke-Klimp, A.; Bax, N.A.; Bellu, A.R.; Winter, E.M.; Vrolijk, J.; Plantinga, J.; Maas, S.; Brinker, M.; Mahtab, E.A.; Gittenberger-de Groot, A.C.; et al. Epicardium-derived cells enhance proliferation, cellular maturation and alignment of cardiomyocytes. J. Mol. Cell Cardiol. 2010, 49, 606–616. [Google Scholar] [CrossRef]
  60. Giacomelli, E.; Meraviglia, V.; Campostrini, G.; Cochrane, A.; Cao, X.; van Helden, R.W.J.; Krotenberg Garcia, A.; Mircea, M.; Kostidis, S.; Davis, R.P.; et al. Human-iPSC-Derived Cardiac Stromal Cells Enhance Maturation in 3D Cardiac Microtissues and Reveal Non-cardiomyocyte Contributions to Heart Disease. Cell Stem Cell 2020, 26, 862–879 e811. [Google Scholar] [CrossRef]
  61. Linask, K.K. N-cadherin localization in early heart development and polar expression of Na+,K(+)-ATPase, and integrin during pericardial coelom formation and epithelialization of the differentiating myocardium. Dev. Biol. 1992, 151, 213–224. [Google Scholar] [CrossRef]
  62. Navaratnam, V.; Kaufman, M.H.; Skepper, J.N.; Barton, S.; Guttridge, K.M. Differentiation of the myocardial rudiment of mouse embryos: An ultrastructural study including freeze-fracture replication. J. Anat. 1986, 146, 65–85. [Google Scholar]
  63. Linask, K.K.; Knudsen, K.A.; Gui, Y.H. N-cadherin-catenin interaction: Necessary component of cardiac cell compartmentalization during early vertebrate heart development. Dev. Biol. 1997, 185, 148–164. [Google Scholar] [CrossRef]
  64. Imanaka-Yoshida, K.; Knudsen, K.A.; Linask, K.K. N-cadherin is required for the differentiation and initial myofibrillogenesis of chick cardiomyocytes. Cell Motil. Cytoskelet. 1998, 39, 52–62. [Google Scholar] [CrossRef]
  65. Radice, G.L.; Rayburn, H.; Matsunami, H.; Knudsen, K.A.; Takeichi, M.; Hynes, R.O. Developmental defects in mouse embryos lacking N-cadherin. Dev. Biol. 1997, 181, 64–78. [Google Scholar] [CrossRef] [PubMed]
  66. Vreeker, A.; van Stuijvenberg, L.; Hund, T.J.; Mohler, P.J.; Nikkels, P.G.; van Veen, T.A. Assembly of the cardiac intercalated disk during pre- and postnatal development of the human heart. PLoS ONE 2014, 9, e94722. [Google Scholar] [CrossRef] [PubMed]
  67. Kostetskii, I.; Li, J.; Xiong, Y.; Zhou, R.; Ferrari, V.A.; Patel, V.V.; Molkentin, J.D.; Radice, G.L. Induced deletion of the N-cadherin gene in the heart leads to dissolution of the intercalated disc structure. Circ. Res. 2005, 96, 346–354. [Google Scholar] [CrossRef]
  68. Piven, O.O.; Kostetskii, I.E.; Macewicz, L.L.; Kolomiets, Y.M.; Radice, G.L.; Lukash, L.L. Requirement for N-cadherin-catenin complex in heart development. Exp. Biol. Med. 2011, 236, 816–822. [Google Scholar] [CrossRef]
  69. Gallicano, G.I.; Kouklis, P.; Bauer, C.; Yin, M.; Vasioukhin, V.; Degenstein, L.; Fuchs, E. Desmoplakin is required early in development for assembly of desmosomes and cytoskeletal linkage. J. Cell Biol. 1998, 143, 2009–2022. [Google Scholar] [CrossRef]
  70. Ha, J.; Kim, B.S.; Min, B.; Nam, J.; Lee, J.G.; Lee, M.; Yoon, B.H.; Choi, Y.H.; Im, I.; Park, J.S.; et al. Intermediate cells of in vitro cellular reprogramming and in vivo tissue regeneration require desmoplakin. Sci. Adv. 2022, 8, eabk1239. [Google Scholar] [CrossRef]
  71. Heuser, A.; Plovie, E.R.; Ellinor, P.T.; Grossmann, K.S.; Shin, J.T.; Wichter, T.; Basson, C.T.; Lerman, B.B.; Sasse-Klaassen, S.; Thierfelder, L.; et al. Mutant desmocollin-2 causes arrhythmogenic right ventricular cardiomyopathy. Am. J. Hum. Genet. 2006, 79, 1081–1088. [Google Scholar] [CrossRef] [PubMed]
  72. Moazzen, H.; Venger, K.; Kant, S.; Leube, R.E.; Krusche, C.A. Desmoglein 2 regulates cardiogenesis by restricting hematopoiesis in the developing murine heart. Sci. Rep. 2021, 11, 21687. [Google Scholar] [CrossRef] [PubMed]
  73. Martin, E.D.; Moriarty, M.A.; Byrnes, L.; Grealy, M. Plakoglobin has both structural and signalling roles in zebrafish development. Dev. Biol. 2009, 327, 83–96. [Google Scholar] [CrossRef] [PubMed]
  74. Bierkamp, C.; McLaughlin, K.J.; Schwarz, H.; Huber, O.; Kemler, R. Embryonic heart and skin defects in mice lacking plakoglobin. Dev. Biol. 1996, 180, 780–785. [Google Scholar] [CrossRef]
  75. Schinner, C.; Xu, L.; Franz, H.; Zimmermann, A.; Wanuske, M.T.; Rathod, M.; Hanns, P.; Geier, F.; Pelczar, P.; Liang, Y.; et al. Defective Desmosomal Adhesion Causes Arrhythmogenic Cardiomyopathy by Involving an Integrin-alphaVbeta6/TGF-beta Signaling Cascade. Circulation 2022, 146, 1610–1626. [Google Scholar] [CrossRef]
  76. Ruiz, P.; Brinkmann, V.; Ledermann, B.; Behrend, M.; Grund, C.; Thalhammer, C.; Vogel, F.; Birchmeier, C.; Gunthert, U.; Franke, W.W.; et al. Targeted mutation of plakoglobin in mice reveals essential functions of desmosomes in the embryonic heart. J. Cell Biol. 1996, 135, 215–225. [Google Scholar] [CrossRef] [PubMed]
  77. Martin, E.D.; Grealy, M. Plakoglobin expression and localization in zebrafish embryo development. Biochem. Soc. Trans. 2004, 32, 797–798. [Google Scholar] [CrossRef] [PubMed]
  78. Moriarty, M.A.; Ryan, R.; Lalor, P.; Dockery, P.; Byrnes, L.; Grealy, M. Loss of plakophilin 2 disrupts heart development in zebrafish. Int. J. Dev. Biol. 2012, 56, 711–718. [Google Scholar] [CrossRef]
  79. Moriarty, M.A.; Martin, E.D.; Byrnes, L.; Grealy, M. Molecular cloning and developmental expression of plakophilin 2 in zebrafish. Biochem. Biophys. Res. Commun. 2008, 367, 124–129. [Google Scholar] [CrossRef]
  80. Sanchez-Fernandez, C.; Rodriguez-Outeirino, L.; Matias-Valiente, L.; Ramirez de Acuna, F.; Hernandez-Torres, F.; Lozano-Velasco, E.; Dominguez, J.N.; Franco, D.; Aranega, A.E. Regulation of Epicardial Cell Fate during Cardiac Development and Disease: An Overview. Int. J. Mol. Sci. 2022, 23, 3220. [Google Scholar] [CrossRef]
  81. Bannerman, D.; Pascual-Gil, S.; Floryan, M.; Radisic, M. Bioengineering strategies to control epithelial-to-mesenchymal transition for studies of cardiac development and disease. APL Bioeng. 2021, 5, 021504. [Google Scholar] [CrossRef] [PubMed]
  82. Kant, S.; Freytag, B.; Herzog, A.; Reich, A.; Merkel, R.; Hoffmann, B.; Krusche, C.A.; Leube, R.E. Desmoglein 2 mutation provokes skeletal muscle actin expression and accumulation at intercalated discs in murine hearts. J. Cell Sci. 2019, 132. [Google Scholar] [CrossRef] [PubMed]
  83. Oxford, E.M.; Musa, H.; Maass, K.; Coombs, W.; Taffet, S.M.; Delmar, M. Connexin43 remodeling caused by inhibition of plakophilin-2 expression in cardiac cells. Circ. Res. 2007, 101, 703–711. [Google Scholar] [CrossRef]
  84. Dubash, A.D.; Kam, C.Y.; Aguado, B.A.; Patel, D.M.; Delmar, M.; Shea, L.D.; Green, K.J. Plakophilin-2 loss promotes TGF-beta1/p38 MAPK-dependent fibrotic gene expression in cardiomyocytes. J. Cell Biol. 2016, 212, 425–438. [Google Scholar] [CrossRef] [PubMed]
  85. Kam, C.Y.; Dubash, A.D.; Magistrati, E.; Polo, S.; Satchell, K.J.F.; Sheikh, F.; Lampe, P.D.; Green, K.J. Desmoplakin maintains gap junctions by inhibiting Ras/MAPK and lysosomal degradation of connexin-43. J. Cell Biol. 2018, 217, 3219–3235. [Google Scholar] [CrossRef] [PubMed]
  86. Agullo-Pascual, E.; Reid, D.A.; Keegan, S.; Sidhu, M.; Fenyo, D.; Rothenberg, E.; Delmar, M. Super-resolution fluorescence microscopy of the cardiac connexome reveals plakophilin-2 inside the connexin43 plaque. Cardiovasc. Res. 2013, 100, 231–240. [Google Scholar] [CrossRef] [PubMed]
  87. Rhee, D.Y.; Zhao, X.Q.; Francis, R.J.; Huang, G.Y.; Mably, J.D.; Lo, C.W. Connexin 43 regulates epicardial cell polarity and migration in coronary vascular development. Development 2009, 136, 3185–3193. [Google Scholar] [CrossRef] [PubMed]
  88. Li, W.E.; Waldo, K.; Linask, K.L.; Chen, T.; Wessels, A.; Parmacek, M.S.; Kirby, M.L.; Lo, C.W. An essential role for connexin43 gap junctions in mouse coronary artery development. Development 2002, 129, 2031–2042. [Google Scholar] [CrossRef]
  89. Dai, P.; Nakagami, T.; Tanaka, H.; Hitomi, T.; Takamatsu, T. Cx43 mediates TGF-beta signaling through competitive Smads binding to microtubules. Mol. Biol. Cell 2007, 18, 2264–2273. [Google Scholar] [CrossRef]
  90. Fukuda, S.; Akiyama, M.; Harada, H.; Nakahama, K.I. Effect of gap junction-mediated intercellular communication on TGF-beta induced epithelial-to-mesenchymal transition. Biochem. Biophys. Res. Commun. 2019, 508, 928–933. [Google Scholar] [CrossRef]
  91. Lim, M.C.; Maubach, G.; Zhuo, L. TGF-beta1 down-regulates connexin 43 expression and gap junction intercellular communication in rat hepatic stellate cells. Eur. J. Cell Biol. 2009, 88, 719–730. [Google Scholar] [CrossRef] [PubMed]
  92. de Boer, T.P.; van Veen, T.A.; Bierhuizen, M.F.; Kok, B.; Rook, M.B.; Boonen, K.J.; Vos, M.A.; Doevendans, P.A.; de Bakker, J.M.; van der Heyden, M.A. Connexin43 repression following epithelium-to-mesenchyme transition in embryonal carcinoma cells requires Snail1 transcription factor. Differentiation 2007, 75, 208–218. [Google Scholar] [CrossRef] [PubMed]
  93. Lyon, H.; Yin, N.; Rupenthal, I.D.; Green, C.R.; Mugisho, O.O. Blocking connexin43 hemichannels prevents TGF-beta2 upregulation and epithelial-mesenchymal transition in retinal pigment epithelial cells. Cell Biol. Int. 2022, 46, 323–330. [Google Scholar] [CrossRef]
  94. Yang, Y.; Liu, W.; Wei, J.; Cui, Y.; Zhang, D.; Xie, J. Transforming growth factor-beta1-induced N-cadherin drives cell-cell communication through connexin43 in osteoblast lineage. Int. J. Oral Sci. 2021, 13, 15. [Google Scholar] [CrossRef]
  95. Kahata, K.; Dadras, M.S.; Moustakas, A. TGF-beta Family Signaling in Epithelial Differentiation and Epithelial-Mesenchymal Transition. Cold Spring Harb. Perspect. Biol. 2018, 10, a022194. [Google Scholar] [CrossRef] [PubMed]
  96. Kwee, L.; Baldwin, H.S.; Shen, H.M.; Stewart, C.L.; Buck, C.; Buck, C.A.; Labow, M.A. Defective development of the embryonic and extraembryonic circulatory systems in vascular cell adhesion molecule (VCAM-1) deficient mice. Development 1995, 121, 489–503. [Google Scholar] [CrossRef]
  97. Dokic, D.; Dettman, R.W. VCAM-1 inhibits TGFbeta stimulated epithelial-mesenchymal transformation by modulating Rho activity and stabilizing intercellular adhesion in epicardial mesothelial cells. Dev. Biol. 2006, 299, 489–504. [Google Scholar] [CrossRef]
  98. Bakin, A.V.; Tomlinson, A.K.; Bhowmick, N.A.; Moses, H.L.; Arteaga, C.L. Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated epithelial to mesenchymal transition and cell migration. J. Biol. Chem. 2000, 275, 36803–36810. [Google Scholar] [CrossRef]
  99. Zhou, B.; Liu, Y.; Kahn, M.; Ann, D.K.; Han, A.; Wang, H.; Nguyen, C.; Flodby, P.; Zhong, Q.; Krishnaveni, M.S.; et al. Interactions between beta-catenin and transforming growth factor-beta signaling pathways mediate epithelial-mesenchymal transition and are dependent on the transcriptional co-activator cAMP-response element-binding protein (CREB)-binding protein (CBP). J. Biol. Chem. 2012, 287, 7026–7038. [Google Scholar] [CrossRef]
  100. Cho, H.J.; Yoo, J. Rho activation is required for transforming growth factor-beta-induced epithelial-mesenchymal transition in lens epithelial cells. Cell Biol. Int. 2007, 31, 1225–1230. [Google Scholar] [CrossRef]
  101. Valcourt, U.; Kowanetz, M.; Niimi, H.; Heldin, C.H.; Moustakas, A. TGF-beta and the Smad signaling pathway support transcriptomic reprogramming during epithelial-mesenchymal cell transition. Mol. Biol. Cell 2005, 16, 1987–2002. [Google Scholar] [CrossRef]
  102. Zavadil, J.; Cermak, L.; Soto-Nieves, N.; Bottinger, E.P. Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO J. 2004, 23, 1155–1165. [Google Scholar] [CrossRef]
  103. Timmerman, L.A.; Grego-Bessa, J.; Raya, A.; Bertran, E.; Perez-Pomares, J.M.; Diez, J.; Aranda, S.; Palomo, S.; McCormick, F.; Izpisua-Belmonte, J.C.; et al. Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 2004, 18, 99–115. [Google Scholar] [CrossRef]
  104. del Monte, G.; Casanova, J.C.; Guadix, J.A.; MacGrogan, D.; Burch, J.B.; Perez-Pomares, J.M.; de la Pompa, J.L. Differential Notch signaling in the epicardium is required for cardiac inflow development and coronary vessel morphogenesis. Circ. Res. 2011, 108, 824–836. [Google Scholar] [CrossRef]
  105. Dimitrova, Y.; Gruber, A.J.; Mittal, N.; Ghosh, S.; Dimitriades, B.; Mathow, D.; Grandy, W.A.; Christofori, G.; Zavolan, M. TFAP2A is a component of the ZEB1/2 network that regulates TGFB1-induced epithelial to mesenchymal transition. Biol. Direct. 2017, 12, 8. [Google Scholar] [CrossRef]
  106. Xiong, Y.; Feng, Y.; Zhao, J.; Lei, J.; Qiao, T.; Zhou, Y.; Lu, Q.; Jiang, T.; Jia, L.; Han, Y. TFAP2A potentiates lung adenocarcinoma metastasis by a novel miR-16 family/TFAP2A/PSG9/TGF-beta signaling pathway. Cell Death Dis. 2021, 12, 352. [Google Scholar] [CrossRef]
  107. Chen, X.; Li, Y.; Luo, J.; Hou, N. Molecular Mechanism of Hippo-YAP1/TAZ Pathway in Heart Development, Disease, and Regeneration. Front. Physiol. 2020, 11, 389. [Google Scholar] [CrossRef]
  108. Meng, Z.; Moroishi, T.; Guan, K.L. Mechanisms of Hippo pathway regulation. Genes Dev. 2016, 30, 1–17. [Google Scholar] [CrossRef]
  109. Hu, Y.; Pu, W.T. Hippo activation in arrhythmogenic cardiomyopathy. Circ. Res. 2014, 114, 402–405. [Google Scholar] [CrossRef]
  110. Chen, S.N.; Gurha, P.; Lombardi, R.; Ruggiero, A.; Willerson, J.T.; Marian, A.J. The hippo pathway is activated and is a causal mechanism for adipogenesis in arrhythmogenic cardiomyopathy. Circ. Res. 2014, 114, 454–468. [Google Scholar] [CrossRef]
  111. Yang, Y.; Ren, J.; Sun, Y.; Xue, Y.; Zhang, Z.; Gong, A.; Wang, B.; Zhong, Z.; Cui, Z.; Xi, Z.; et al. A connexin43/YAP axis regulates astroglial-mesenchymal transition in hemoglobin induced astrocyte activation. Cell Death Differ. 2018, 25, 1870–1884. [Google Scholar] [CrossRef]
  112. Narimatsu, M.; Samavarchi-Tehrani, P.; Varelas, X.; Wrana, J.L. Distinct polarity cues direct Taz/Yap and TGFbeta receptor localization to differentially control TGFbeta-induced Smad signaling. Dev. Cell 2015, 32, 652–656. [Google Scholar] [CrossRef]
  113. Noguchi, S.; Saito, A.; Nagase, T. YAP/TAZ Signaling as a Molecular Link between Fibrosis and Cancer. Int. J. Mol. Sci. 2018, 19, 3674. [Google Scholar] [CrossRef]
  114. Kant, S.; Holthofer, B.; Magin, T.M.; Krusche, C.A.; Leube, R.E. Desmoglein 2-Dependent Arrhythmogenic Cardiomyopathy Is Caused by a Loss of Adhesive Function. Circ. Cardiovasc. Genet. 2015, 8, 553–563. [Google Scholar] [CrossRef]
Figure 1. Desmosomes in the heart. (A) Scheme of the molecular desmosome structure. PM: plasma membrane. (B) Fluorescence microscopy detecting desmoplakin (Dsp; red) and nuclei (blue) in embryonic (left) and adult murine heart (right). CM: compact myocardium, Epi: Epicardium. Scale bars: 20 μm. (C) Electron micrographs of cardiomyocytes in embryonic (left) and adult murine hearts (right). Asterisks point to sarcomeres, arrows to actin-anchoring adherens junctions, and arrowheads to desmin-anchoring desmosomes. TM: trabecular myocardium; CM: compact myocardium; m, mitochondrion. Scale bar: 1 μm.
Figure 1. Desmosomes in the heart. (A) Scheme of the molecular desmosome structure. PM: plasma membrane. (B) Fluorescence microscopy detecting desmoplakin (Dsp; red) and nuclei (blue) in embryonic (left) and adult murine heart (right). CM: compact myocardium, Epi: Epicardium. Scale bars: 20 μm. (C) Electron micrographs of cardiomyocytes in embryonic (left) and adult murine hearts (right). Asterisks point to sarcomeres, arrows to actin-anchoring adherens junctions, and arrowheads to desmin-anchoring desmosomes. TM: trabecular myocardium; CM: compact myocardium; m, mitochondrion. Scale bar: 1 μm.
Cells 12 02122 g001
Figure 2. The impact of desmosomal mutation (Dsg2ΔE4–6) on the developing and mature murine heart. The scheme at left indicates that desmosome deficiency leads to remodeling of the embryonic heart, resulting in an expansion of hematopoietic stem cells and erythrocytes. The image below shows the dorsal side of an E11.5 heart with an excessive expansion of erythrocytes that populate the left ventricle and spread out to the pericardial side of the heart (arrow). The scheme at right shows an adult heart, which responds to desmoglein 2 mutation by cardiac remodeling resulting in the expansion of fibrotic cells. The photograph below depicts these changes in the right ventricle (arrow). OFT: outflow tract; RV: right ventricle; LV: left ventricle; RA: right atrium; EPDCs: Epicardial derived cells.
Figure 2. The impact of desmosomal mutation (Dsg2ΔE4–6) on the developing and mature murine heart. The scheme at left indicates that desmosome deficiency leads to remodeling of the embryonic heart, resulting in an expansion of hematopoietic stem cells and erythrocytes. The image below shows the dorsal side of an E11.5 heart with an excessive expansion of erythrocytes that populate the left ventricle and spread out to the pericardial side of the heart (arrow). The scheme at right shows an adult heart, which responds to desmoglein 2 mutation by cardiac remodeling resulting in the expansion of fibrotic cells. The photograph below depicts these changes in the right ventricle (arrow). OFT: outflow tract; RV: right ventricle; LV: left ventricle; RA: right atrium; EPDCs: Epicardial derived cells.
Cells 12 02122 g002
Table 1. Desmosomal protein involvement in cardiogenesis.
Table 1. Desmosomal protein involvement in cardiogenesis.
Cardiogenesis Phenotype
MutationOrganismEdemaPericardial BloodHypoplastic MyocardDefective ContractionDisrupted Myocardial PatterningDefective Intercellular AdhesionRupturePerturbed Endocardial DifferentiationReduced Desmosomal PlaqueRefs.
Dsc2morpholinoZebrafish× × ×[71]
Dsg2ΔE4–E6Mouse××× ×××× [72]
Dsg2W2AMouse×× [75]
Pkp2morpholinoZebrafish ×× [73]
Pkp2−/−Mouse××× × [17]
Jup−/−Mouse××× × × ×[74,76]
Dsp−/− extraembryonal rescueMouse ×××× [16]
Table 2. Interaction of desmosomal proteins with EMT pathways.
Table 2. Interaction of desmosomal proteins with EMT pathways.
MutationCell/Animal ModelPathwayRefs.
Dsg2RNAiHuman pluripotent stem cells (hPSCs)Inhibition of E-cadherin and elevation of Slug[15]
Dsg2ΔE4–E6
Dsg2ΔE4–E5 myocardial induction
Adult murine heartTGFβ and SRF signaling[82]
Dsg2W2AAdult murine heartIntegrin-αVβ6/TGF-β signaling[75]
Pkp2RNAiNeonatal rat ventricular myocytes (NRVMs)
Epcardial-mesenchymal cells (EPDCs)
Reduction and redistribution of Cx43[83]
Pkp2RNAiNeonatal rat ventricular myocytes (NRVMs)TGF-β1/p38 MAPK kinase signaling[84]
Pkp2c.del2013cHuman-induced pluripotent stem cell- derived (hiPSC)-epicardial cellsActivation of TFAP2A[22]
DspRNAiNeonatal rat ventricular myocytes (NRVMs)
Murine HL-1 atrial cardiomyocytes
ERK1/2-MAPK signaling
Phosphorylation and degradation of Cx43
[85]
DspW/F epicardial inductionAdult Mouse HeartFGF2 and TGF-β1 signaling[21]
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

Moazzen, H.; Bolaji, M.D.; Leube, R.E. Desmosomes in Cell Fate Determination: From Cardiogenesis to Cardiomyopathy. Cells 2023, 12, 2122. https://doi.org/10.3390/cells12172122

AMA Style

Moazzen H, Bolaji MD, Leube RE. Desmosomes in Cell Fate Determination: From Cardiogenesis to Cardiomyopathy. Cells. 2023; 12(17):2122. https://doi.org/10.3390/cells12172122

Chicago/Turabian Style

Moazzen, Hoda, Mistura Dolapo Bolaji, and Rudolf E. Leube. 2023. "Desmosomes in Cell Fate Determination: From Cardiogenesis to Cardiomyopathy" Cells 12, no. 17: 2122. https://doi.org/10.3390/cells12172122

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