Skip to main content

The benign nature and rare occurrence of cardiac myxoma as a possible consequence of the limited cardiac proliferative/ regenerative potential: a systematic review

Abstract

Background

Cardiac Myxoma is a primary tumor of heart. Its origins, rarity of the occurrence of primary cardiac tumors and how it may be related to limited cardiac regenerative potential, are not yet entirely known. This study investigates the key cardiac genes/ transcription factors (TFs) and signaling pathways to understand these important questions.

Methods

Databases including PubMed, MEDLINE, and Google Scholar were searched for published articles without any date restrictions, involving cardiac myxoma, cardiac genes/TFs/signaling pathways and their roles in cardiogenesis, proliferation, differentiation, key interactions and tumorigenesis, with focus on cardiomyocytes.

Results

The cardiac genetic landscape is governed by a very tight control between proliferation and differentiation-related genes/TFs/pathways. Cardiac myxoma originates possibly as a consequence of dysregulations in the gene expression of differentiation regulators including Tbx5, GATA4, HAND1/2, MYOCD, HOPX, BMPs. Such dysregulations switch the expression of cardiomyocytes into progenitor-like state in cardiac myxoma development by dysregulating Isl1, Baf60 complex, Wnt, FGF, Notch, Mef2c and others.

The Nkx2–5 and MSX2 contribute predominantly to both proliferation and differentiation of Cardiac Progenitor Cells (CPCs), may possibly serve roles based on the microenvironment and the direction of cell circuitry in cardiac tumorigenesis. The Nkx2–5 in cardiac myxoma may serve to limit progression of tumorigenesis as it has massive control over the proliferation of CPCs. The cardiac cell type-specific genetic programming plays governing role in controlling the tumorigenesis and regenerative potential.

Conclusion

The cardiomyocytes have very limited proliferative and regenerative potential. They survive for long periods of time and tightly maintain the gene expression of differentiation genes such as Tbx5, GATA4 that interact with tumor suppressors (TS) and exert TS like effect. The total effect such gene expression exerts is responsible for the rare occurrence and benign nature of primary cardiac tumors. This prevents the progression of tumorigenesis. But this also limits the regenerative and proliferative potential of cardiomyocytes. Cardiac Myxoma develops as a consequence of dysregulations in these key genes which revert the cells towards progenitor-like state, hallmark of CM. The CM development in carney complex also signifies the role of TS in cardiac cells.

Peer Review reports

Background

Primary tumors of the heart are exceedingly rare, with nearly 90% of them classified as benign [1]. This study focuses on a particular subtype of benign cardiac tumors known as cardiac myxomas (CM), primarily because they are the most prevalent among primary benign cardiac tumors [1]. Cardiac myxomas, despite their benign nature, hold significant relevance in cardiac research and clinical practice. CM are benign tumors of the heart. They are multipotent with mesenchymal stem cell-like nature. To comprehend the significance of CM and their potential implications for cardiomyocyte biology and cardiac regeneration, it is crucial to first understand the general concepts surrounding cardiac regeneration. The adult human heart has limited regenerative capacity, a characteristic that poses significant challenges in addressing cardiac diseases [2]. This limited regenerative potential of cardiomyocytes has sparked interest in understanding the mechanisms governing cardiac cell fate and the factors that might influence their regenerative and proliferative abilities [3].

Histopathological features of CM often reveal areas of hypercellularity, necrosis, and atypia [1, 3, 4]. It is within these intricate patterns that the transformation of cardiomyocytes into cardiac progenitor-like cells, a distinctive hallmark of cardiac myxoma, becomes evident [5, 6]. These areas of hypercellularity suggest the presence of cells in various stages of differentiation, reminiscent of cardiac progenitors [7, 8]. Furthermore, the presence of atypia hints at the dynamic reprogramming of cardiac cells, as they shift from their terminally differentiated state toward a more primitive, progenitor-like phenotype. The histopathological landscape of CM serves as a visual representation of the intriguing process by which cardiomyocytes seem to undergo transformation into progenitor-like cells, hallmark of CM [9, 10].

Current challenges and gaps

The precise etiology of CM remains elusive, and despite some studies hinting at the possible role of transcription factor Nkx2–5 in CM development, our understanding of CM’s origin and the factors involved is far from comprehensive [11,12,13,14,15].

The possible role of limited regenerative and proliferative potential of cardiomyocytes in the development of primary cardiac tumors such as CM is not yet fully understood [4, 5], and how this is related to the benign nature of CM [6, 8].

Objectives of the study

CM are benign tumors of the heart. They are multipotent with mesenchymal stem cell-like nature [9, 10, 16,17,18]. This study intends to contribute to a deeper understanding of cardiomyocyte biology and the factors that possibly influence their resistance to neoplastic transformations [19]. It investigates how the resistance to malignant transformation of CM may possibly be related to the limited proliferative and regenerative potential of cardiomyocytes [7].

Our aim is to explore the relationship between CM and the limited regenerative potential of cardiomyocytes. By investigating key cardiac transcription factors, genes, signaling pathways, and other mechanisms, we seek to shed light on the development of CM and its potential implications for cardiac regeneration [20,21,22,23,24].

Methods

Article screening and inclusion

The process of article screening and inclusion was meticulously conducted to ensure that only relevant and high-quality literature was incorporated into this study. The aim was to identify and evaluate articles that provided insight into the roles of cardiac genes, transcription factors (TFs), and signaling pathways in cardiogenesis, cardiomyocyte development, proliferation, differentiation, tumorigenesis, and their connection to Cardiac Myxoma (CM). The literature search and data collection commenced in January 2019 and concluded in February 2022. During the revision process, additional literature searches were conducted and referenced up to October 2022. The study’s timeline facilitated a significant focus on genes, TFs, and pathways that are fundamental to the regulation of cardiac development, lineage commitment, and the maintenance of cardiac identity. The extensive duration enabled a thorough search across various databases and resources, ensuring that no relevant information was overlooked. Understanding the roles of these pivotal cardiac genes and TFs is crucial for unraveling the complexities of CM development. Their participation in balancing proliferation and differentiation processes is particularly intriguing and necessitated a wide search horizon to encompass the evolving body of knowledge in this field. This endeavor involved a comprehensive exploration of key cardiac genes, transcription factors (TFs), and signaling pathways, each of which plays multifaceted roles in cardiogenesis, cardiomyocyte development, proliferation, differentiation, tumor suppression or tumorigenesis.

  1. 1.

    Database selection and search strategy: The search process commenced with a thorough selection of databases renowned for their comprehensive coverage in the field of biomedical research. Three primary databases were: PubMed, MEDLINE, and Google Scholar. These platforms were selected due to their accessibility and comprehensive indexing, allowing us to obtain a broad spectrum of literature.

  2. 2.

    Multi-step screening process: A multi-step screening process was systematically implemented to identify articles aligning with our predefined inclusion and exclusion criteria. This process aimed at meticulously filtering through a large pool of potentially relevant studies to ensure that only the most pertinent articles were considered for inclusion in our study.

Title and abstract screening

Initially, articles were screened based on their titles and abstracts. This preliminary step served as an effective means of identifying articles that exhibited direct relevance to the research objectives of this study. Articles that clearly did not pertain to cardiac genes, TFs, signaling pathways, or cardiomyocyte biology were eliminated.

Full-text review

Articles that passed the title and abstract screening phase underwent a comprehensive full-text review. During this in-depth assessment, we scrutinized each article to determine its relevance to the study’s focus areas. Those that did not offer valuable insights into the roles of cardiac genes, TFs, and signaling pathways in the context of cardiogenesis, cardiomyocyte development, proliferation, differentiation, tumorigenesis, or CM were excluded.

  1. 3.

    Data extraction: Following the final selection of articles, relevant data was extracted from each article, encompassing key findings and outcomes that were significant to our study objectives. This process aimed to ensure that the data extracted was relevant and provided valuable insights into the complex interplay of cardiac genes, TFs, and signaling pathways.

Inclusion criteria, selection of genes, transcription factors, and signaling pathways

To be deemed eligible for inclusion in our study, articles had to be directly related to key cardiac transcription factors/genes and signaling pathways involved in cardiogenesis, with a specific emphasis on the developmental biology of cardiomyocytes and cardiomyocyte differentiation. Articles that did not meet these specific criteria were excluded. The choice of specific cardiac genes, TFs, and signaling pathways for investigation was guided by their well-established roles in cardiogenesis, cardiomyocyte development, proliferation, and differentiation. Furthermore, these factors were also examined in the context of tumor suppression, tumorigenesis, and their relevance to CM. While the factors we selected are indeed significant contributors to cardiac biology and pathology, it is essential to acknowledge that other factors may also play a role in the processes under investigation. However, due to the defined scope of our study, these factors were beyond the current study’s scope.

The following genes/TFs and signaling pathways were investigated for their role in cardiogenesis/cardiomyocyte development, proliferation, differentiation, tumor suppression, tumorigenesis and in CM: Isl1, Brg1/Baf60 – Smarcd3 complex, Nkx2–5, GATA4, Tbx5, Mef2c, HAND1/2, MYOCD, MSX2, HOPX, Wnt-signaling pathway, Notch, FGF, BMPs.

These key cardiac genes/TFs play pivotal roles in cardiac development by guiding lineage commitment, balancing proliferation and differentiation, regulating essential processes, and maintaining cardiac identity.

This study adheres to relevant PRISMA guidelines (Preferred Reporting Items for Systematic Reviews and Meta-Analyses).

Rationale for screening and inclusion

It is deeply rooted in the pivotal significance of certain cardiac genes, transcription factors (TFs), and signaling pathways in unraveling the complex process of cardiac tumorigenesis, with a primary focus on cardiac myxomas (CM). The selection of these specific factors is driven by their unique and critical roles, which have the potential to shed light on CM development. These include Isl1, a key controller of cardiomyocyte cell fate, highly expressed in multipotent cardiac progenitor cells (CPCs), which not only specifies cardiac lineage and differentiation but also exhibits interactions with Nkx2–5 and Estrogen Receptor Alpha, suggesting its potential involvement in CM. The Brg1/Baf60 – Smarcd3 Complex acts as a crucial transcriptional regulator, inducing CPC proliferation, and its defects are linked to differentiation anomalies, potentially collaborating with the Wnt signaling pathway in promoting tumorigenesis. Nkx2–5, among the earliest cardiac-specific patterning genes, plays a central role in inducing cardiac programming in CPCs, enhancing differentiation, and interacting with Tbx5 and GATA4, especially with its upregulated expression in CM, possibly contributing to CM heterogeneity. GATA4, a master regulator of genes pivotal for cardiogenesis, significantly influences cardiac morphogenesis, survival, and differentiation, with decreased expression leading to cardiomyocyte reversion to a progenitor-like state. Inclusion of Tbx5 is essential due to its ability to boost the expression of other cardiogenic TFs, thereby guiding CPC differentiation while suppressing non-cardiac gene expression and potentially contributing to CM heterogeneity. The Mef2c gene is incorporated as it contributes to CPC proliferation and forms complexes with key cardiac TFs, participating in cardiac morphogenesis and enhancing differentiation, potentially collaborating with the Wnt pathway and Isl1 in generating CPC-like states. The HAND1/2, with regulatory roles in cardiogenesis, including the enhancement of both proliferation with Nkx2–5 and differentiation with GATA4, is of particular interest as it may act as a tumor suppressor and possibly becomes downregulated in CM development, making it crucial to understand the multifaceted roles of these TFs in both normal cardiac development and CM tumorigenesis. The MYOCD gene is another crucial factor under investigation, as it regulates CPC growth arrest and governs CPC stemness. Understanding the role of MYOCD can offer insights into the factors that maintain the unique characteristics of CM. MSX2, included for its interactions with HAND1/2 in regulating gene expression and its role in enhancing CPC proliferation, is of particular interest due to its potential involvement in promoting progenitor-like states in advanced CM. HOPX, expressed as CPCs commit to the cardiomyocyte fate, is essential for enhancing cardiomyocyte differentiation and acting as a tumor suppressor. The investigation of HOPX provides insights into how its downregulation may contribute to CM development. The inclusion of the Wnt Signaling Pathway is vital as it is involved in CPC renewal and maintenance, processes that are essential in understanding the regenerative potential of cardiac cells. Its role in enhancing CPC stemness and contributing to cardiomyocyte dedifferentiation in CM development is central to this research. The FGF Signaling Pathway is included as it drives stem cell differentiation into CPCs and forms complexes that regulate differentiation and proliferation. The potential for its dysregulation to lead to the reversion of cardiomyocytes toward progenitor-like states in CM development highlights its significance in this study. BMPs, through their role in downregulating progenitor genes in CPCs and enhancing cardiomyocyte differentiation, are vital in understanding the control of cardiac cell fate and its impact on CM development. The Notch Signaling Pathway, participating in cardiac morphogenesis and regulating cardiomyocyte proliferation and differentiation, is included to uncover its potential collaboration with Isl1 and Mef2c in CM development. The comprehensive exploration of these key genes, TFs, and signaling pathways is essential to provide valuable insights into their roles in cardiogenesis, proliferation, differentiation, and their potential contributions to the development of cardiac myxomas.

Assessment of article quality and potential biases

During the article screening and inclusion process, the quality of the selected articles and the assessment of potential biases were pivotal aspects to ensure the rigor and reliability of the research findings.

  1. 1.

    Quality assessment: The first step in quality assessment involved evaluating the methodological rigor of the selected articles. This entailed a careful examination of the study design, data collection methods, and analyses conducted in those studies. The quality of evidence was considered when determining the significance of the study’s findings. Articles that demonstrated sound methodology, such as well-designed studies/experiments, controlled variables, and appropriate scientifically sound data, were considered of higher quality. The fact that the selected articles had undergone a peer-review process was also a significant indicator of quality. Peer-reviewed articles are subject to scrutiny by experts in the field, ensuring the validity and credibility of the research. The quality of evidence presented in the selected articles was also a focus of the assessment. High-quality evidence often comes from properly conducted reviews or well-designed studies with rigorous data collection methods and robust analyses.

  2. 2.

    Potential biases assessment:

Publication bias: The potential for publication bias was addressed. This bias can occur when only studies with positive or significant results are published, leading to an overestimation of effects. To minimize this bias, studies that provided a balanced representation of both positive and negative results were actively sought. A comprehensive search strategy, including databases like Google Scholar, was adopted to include a wide range of published articles.

Selection bias: To assess selection bias, predefined and transparent inclusion criteria was applied to minimize subjectivity. Articles were selected based on their relevance to this study’s objectives, and this process adhered to predefined criteria. This approach reduced the risk of subjectivity in article selection.

Reporting bias: Reporting bias occurs when studies selectively report certain outcomes while omitting others. Articles were checked for inconsistencies or missing data to make sure that such studies do not mislead the findings of this study. To identify and address reporting bias, multiple detailed reviews of the methodologies and results were conducted for all the selected articles.

By ensuring that high-quality, peer-reviewed studies were included and potential biases were assessed, this study aimed to provide a robust foundation for results and conclusions presented in the study. This enhanced the reliability and credibility of the study, making it a valuable contribution to the field of cardiac genetics and its role in tumorigenesis.

Language and publication restrictions

We restricted our selection to publications in the English language. There were no limitations imposed on the date of publication. Unpublished studies were not included in our analysis.

Results

A total of 2610 articles were identified using database searching, and 2277 were recorded after duplicates removal. One thousand seven hundred eighty-five (1785) were excluded after screening of title/abstract, 215 articles were further excluded from consideration based on a more detailed review of the full texts. These exclusions were primarily due to factors such as non-conformity with the study focus, insufficient methodological rigor, or data that did not align with our research questions. and 3 articles were excluded during data extraction. Finally, 274 articles were included (79 were review articles, 1 was clinical trial, 105 were in vivo studies and 89 were in vitro studies). Ultimately, 2 duplicate references were also deleted during the final checks. Figure 1 illustrates the landscape of genes/transcription factors/signaling pathways involved in cardiac development and their potential roles in Cardiac Myxoma. The flow of citations is represented in Fig. 2.

Fig. 1
figure 1

Landscape of genes/tfs/signaling pathways involved in cardiac development and their possible roles in CM

Fig. 2
figure 2

PRISMA flow diagram: This figure only highlights the methodology of the study in relation to its limitations. This figure represents graphically the flow of citations in the study

Significance of cardiac genes/TFs in investigating cardiac tumorigenesis: an overview of the study

  1. 1.

    Isl1: Controls cardiomyocyte cell fate, strongly expressed in multipotent cardiac progenitor cells (CPCs). Specifies cardiac lineage and differentiation. Interactions with Nkx2–5 and Estrogen Receptor Alpha suggest potential involvement.

  2. 2.

    Brg1/Baf60 – Smarcd3 Complex: Acts as a transcriptional regulator. Induces CPC proliferation; defects in this complex lead to differentiation defects. May collaborate with the Wnt signaling pathway to promote tumorigenesis.

  3. 3.

    Nkx2–5: Among the earliest cardiac-specific patterning genes, induces cardiac programming in CPCs. Enhances differentiation; interacts with Tbx5 and GATA4. Upregulated expression in CM; potential contribution to CM heterogeneity.

  4. 4.

    GATA4: Regulates genes crucial for cardiogenesis. Influences morphogenesis, survival, and differentiation. Reduced expression leads to cardiomyocyte reversion to a progenitor-like state.

  5. 5.

    Tbx5: Boosts expression of other cardiogenic TFs. Enhances CPC differentiation; suppresses non-cardiac gene expression. May contribute to CM heterogeneity.

  6. 6.

    Mef2c: Contributes to CPC proliferation; forms complexes with key cardiac TFs. Involved in cardiac morphogenesis; enhances differentiation. May collaborate with Wnt and Isl1 in generating CPC-like states.

  7. 7.

    HAND1/2: Regulates cardiogenesis; enhances proliferation with Nkx2–5 and differentiation with GATA4. May act as a tumor suppressor; possibly downregulated in CM development.

  8. 8.

    MYOCD: Regulates CPC growth arrest. Governs CPC stemness. May contribute to CM’s benign nature and rarity.

  9. 9.

    MSX2: Interacts with HAND1/2 to regulate gene expression. Enhances CPC proliferation. May promote progenitor-like states in advanced CM.

  10. 10.

    HOPX: Expressed as CPCs commit to cardiomyocyte fate. Enhances cardiomyocyte differentiation; acts as a tumor suppressor. Downregulation may contribute to CM development.

  11. 11.

    Wnt Signaling Pathway: Involved in CPC renewal and maintenance. Enhances CPC stemness; contributes to cardiomyocyte dedifferentiation in CM development.

  12. 12.

    FGF Signaling Pathway: Drives stem cell differentiation into CPCs. Forms complexes regulating differentiation and proliferation; dysregulation may reverse cardiomyocytes toward progenitor-like states in CM development.

  13. 13.

    BMPs: Downregulates progenitor genes in CPCs. Enhances cardiomyocyte differentiation. Downregulation may contribute to CM.

  14. 14.

    Notch Signaling Pathway: Participates in cardiac morphogenesis. Regulates cardiomyocyte proliferation and differentiation. May collaborate with Isl1 and Mef2c in CM development.

Based on the objectives of the study, the comprehensive exploration of key genes, TFs, and signaling pathways is given below. It is capable of offering valuable insights into their roles in cardiogenesis, proliferation, differentiation, and their potential contributions to the development of cardiac myxomas.

Table 1 presents an overview of cardiac genes, transcription factors, and signaling pathways, shedding light on their pivotal roles in cardiogenesis, proliferation/ differentiation, and their possible role in involvement in Cardiac Myxoma (CM) development.

Table 1 Cardiac genes/transcription factors/signaling pathways and their roles in cardiogenesis, proliferation/differentiation, and in CM development

Table 2 shows a classification of the cited references, offering insights into the topics covered by each study, their categorization in terms of histopathology, human samples, or clinical details, and their relevance to this study.

Table 2 Classification of cited references

Key cardiac transcription factors/genes

In this section, this study investigates cardiac myxoma through the lens of developmental biology [152, 153]. The CPCs are multi-lineage cells with major expression of Nkx2–5 and Isl1 [154, 155]. By increasing the gene expressions of BMP signaling pathway and downregulating Wnt pathway, the CPCs begin to differentiate into cardiomyocytes [156]. The Isl1 and Nkx2–5 TFs act via activating cascade of downstream cardiac genes in time specific manner [58].

It is important to note that Isl1 is a pioneering Transcription factor (PTF) of cardiomyocyte cell fate. As Isl1 expression begins to decline, the HOPX becomes upregulated [157]. The Nkx2–5 has very strong interactions with HOPX as it acts as a downstream regulator of HOPX and governs its gene expression. The HOPX is expressed in cardiomyoblast and is very important in the process of differentiation as it is also expressed in pre-cardiac mesoderm. HOPX positively interacts with BMPs, SMADs and negatively with Wnt-signaling and Axin2 signaling pathway. The Wnt-pathway and Axin2 oppose differentiation of CPCs [158].

In CPCs, the Nkx2–5 expression continuously increases over the duration of differentiation. When HOPX is defective, Wnt-pathway becomes upregulated and this downregulates Nkx2–5. Normally, the BMP-SMAD complex is activated by HOPX. This downregulates WNT-signaling pathway and promotes differentiation of CPCs towards cardiomyocyte development. This BMP-SMAD complex increases MSX1 expression to promote differentiation and downregulates Axin2 [159].

Islet1

Role in cardiogenesis

Islet1 (Isl1) plays the role of a PTF in epigenetic control of cardiomyocyte cell fate [25]. This also governs epigenetic programming and shapes chromatin landscape. It works with additional regulatory factors to specify cell lineage and cardiac differentiation [26]. Isl1 governs a regulatory network of genes that is involved in unfolding cardiac lineage [27]. It is transiently expressed in CPCs including atrial area and is involved in their proliferation, survival and migration [28]. When Isl1 is defective, cardiac development gets disrupted. Isl1 expression is greater before progenitor cells differentiate into heart tube [29].

Proliferation-related roles

Isl1 is one of the earliest genes expressed in the cardiac progenitors. Isl1 interacts strongly with Tbx1 and both are strongly expressed in multipotent CPCs [30, 31]. It also has major interaction with Wnt-signaling pathway. Isl1 also works with FGF10 to promote proliferation of progenitor cells [32]. Isl1 expression also plays important roles in the neural crest cells, and in heart, in which the progenitor cells expressing Isl1 are capable of differentiating into cardiomyocytes, endothelial and smooth muscle lineage [33]. The CPCs also maintain expression of Isl1 in postnatal state. The Isl1 expression is essential in renewal of cardiac progenitors. Prior to differentiation, the gene expression of Isl1 contributes to proliferation of CPCs [34, 35].

Key interactions with tumor suppressors/ differentiation-related genes

Isl1 sets in motion the gene expression of Brg1- Baf60 which contributes to commit CPCs towards cardiomyocyte fate. The cardiac differentiation-related genes downregulate the expression of genes involved in the maintenance of progenitor-like state in CPCs. Nkx2–5 promotes the process of differentiation, and downregulates the gene expression of Isl1 [36]. GATA4 activates Isl1 enhancer. The BMPs downregulate the gene expression of Isl1 and Tbx1 to increase myocardial differentiation [37].

Contributions to combinatorial code/ cell type specific genetic-programming

The induction of cell type specific genetic-programming needs PTFs such as Isl1 which works with other special TFs to form combinations which determine the final cell type and regulate the process of cardiomyocyte development [38, 39].

Presence in other tumors

Upregulated expression of Isl1 is present in many cancers including pheochromocytoma, pancreatic, gastrointestinal, lung tumors, bile duct carcinoma, prostate and breast cancers. Isl1 expression is also present in insulinoma cells, bladder cancer, Non-Hodgkin lymphoma, glioma, melanoma and others [160,161,162,163]. Isl1 is a novel regulator of cyclins and c-myc gene. This also emphasizes the role of Isl1 in tumor development [164].

Possible role in cardiac myxoma

Despite being very similar to multipotent CPCs, CM are c-kit positive but very rarely Isl1 positive [40]. As the Nkx2–5 has been found to be involved in CM development, its role in interacting with Isl1 is very significant. Possibly, the influence of Nkx2–5 over the genetic landscape in cardiac myxoma prevents the progression of cardiac myxoma cells towards malignant state [41, 42].

Isl1 and GATA4 also interact with Estrogen Receptor alpha. This may be a contributing factor in the development of more cardiac myxoma cases in female patients [43,44,45].

Summary: unraveling the multifaceted roles of Isl1

Isl1 is a pivotal player in cardiogenesis, orchestrating cardiomyocyte cell fate by shaping the epigenetic landscape and interacting with regulatory factors. It forms a network of genes that guide cardiac lineage development, with transient expression in cardiac progenitor cells, influencing proliferation, survival, and migration. Malfunctions in Isl1 disrupt cardiac development. It interacts strongly with Tbx1, the Wnt pathway, and FGF10 to promote progenitor cell proliferation and differentiation into various cardiac cell types. Isl1 also influences cardiomyocyte commitment by partnering with Brg1-Baf60. Its interactions with differentiation-related genes such as Nkx2–5, GATA4, and BMPs, drive myocardial differentiation. Isl1 collaborates with other transcription factors to determine final cell types, and its upregulated expression is found in various cancers. Despite its presence in multipotent cardiac progenitor cells, Isl1 is rarely expressed in cardiac myxomas, possibly influenced by Nkx2–5, GATA4, and Estrogen Receptor alpha interactions.

Brg1/Baf60 – Smarcd3 complex

Role in cardiogenesis

It is massively expressed in early stage of cardiac development [46]. Its defects exhibit cardiac morphogenetic defects as it acts as transcriptional regulator. It promotes progenitors towards cardiomyocytes, whereas its over-expression has been found to accelerate the activation of cardiac lineage-related target genes [47, 48].

Proliferation-related roles

When this complex is under the influence of Wnt-signaling, it contributes to epithelial-mesenchymal transition (EMT). The Baf60 is capable of inducing proliferation in progenitor cells, but with SMARCD3 complex its function becomes so much different and it contributes to cardiac differentiation. In neural progenitors, it interacts with notch to promote proliferation [49, 50].

Key interactions with tumor suppressors/ differentiation-related genes

It mediates interactions with core cardiac TFs including Tbx5, Nkx2–5 and GATA4. Specifically, it promotes the binding of GATA4 and Tbx5 to cardiac specific genes, thus inducing downstream regulatory networks. The process of cardiac differentiation becomes defective when there are defects in this complex [51]. SMARCD3 is also considered to play a TS role [52, 53].

Contributions to combinatorial code/ cell type specific genetic-programming

This complex when combined with GATA4 and Tbx5, is capable of switching on the cardiac gene expression in non-cardiac regions. It is capable of driving mesenchymal cells to develop into cardiomyocytes. This complex has major interactions with GATA4 to turn on cardiac-specific gene expression [54] Then it combines with Tbx5 to repress the gene expression of non-cardiac genes. This complex has considerable control over cardiac differentiation and may have fundamental influence over cardiac regeneration potential, as it is one of the key parts of cardiac-specific cell programming [55].

Presence in other tumors

Brg1/Baf60 – Smarcd3 complex plays multiple roles in different cancers depending on the microenvironment and also influenced of predominant signaling pathways such as Wnt, TGF- beta and MAPK. In colorectal cancer, it works with Wnt-pathway to promote metastasis. On the contrary, it acts as a possible TS in breast cancer [56, 57, 165, 166].

Possible role in cardiac myxoma

There is not much data about the role of this complex in CM. The role that this gene complex plays is variable and is also dependent on the gene expression of other key regulatory genes/TFs and signaling pathways. Wnt-signaling is involved in the proliferation of progenitor cells, but in the presence of GATA4 and Tbx5 it promotes the differentiation of cardiomyocytes. Baf60 complex works by interacting with genes/TFs involved in governing the gene expression of CPCs. Similarly, in CM development the expression of Baf60 complex may promote tumorigenesis because of the dysregulated expression of genes/TFs involved in progenitor-like state, that is hallmark of CM.

Summary: unraveling the multifaceted roles of Brg1/Baf60-Smarcd3 complex

The Brg1/Baf60-Smarcd3 complex plays a pivotal role in cardiogenesis by acting as a transcriptional regulator expressed early in cardiac development. It promotes progenitors’ transition to cardiomyocytes, and its over-expression accelerates the activation of cardiac lineage-related genes. When influenced by Wnt-signaling, it contributes to epithelial-mesenchymal transition (EMT) and cardiac differentiation, interacting with key regulators like Tbx5, Nkx2–5, and GATA4. This complex can reprogram non-cardiac regions into cardiac gene expression, affecting cardiac-specific cell programming. In various cancers, its roles vary depending on microenvironment and signaling pathways. While little is known about its role in cardiac myxoma, its interactions with regulatory genes and signaling pathways may influence tumorigenesis by affecting progenitor-like states, a hallmark of CM.

Nkx2–5

Role in cardiogenesis

The TF Nkx2–5 is among the very first cardiac specific patterning genes in cardiac development. The site of its expression, the heart forming region plays key roles in cardiac specification, differentiation and proliferation [56]. It is one of the four key regulators of cardiac cell type. It is expressed in precursor cardiac cells and leads to proper cardiac development. It decides atrial and ventricular fate and defects in the gene can lead to congenital heart defects [57].

Proliferation-related roles

This gene is of key significance as it results in a cascade of downstream signaling and induces the cardiac programming in pluripotent mesenchymal stem cells. It is dependent on JAK-STAT pathway. The TF Nkx2–5 controls cardiomyocyte differentiation by working with Mef2c, which is a key enhancer of Nkx2–5 [58]. The most significant aspect of this TF related to the etiology of cardiac myxomas is that Nkx2–5 is first expressed in CPCs and its gene expression is downregulated temporarily during cardiomyocyte differentiation [167]. However, a constant low level of Nkx2–5 gene expression persists throughout life. It is involved in the induction of initial but not late phases of cardiomyocyte development [59]. Moreover, it interacts with notch signaling pathway to promote proliferation of CPCs. The final determination of cardiovascular lineages is regulated by Nkx2–5 in the earliest specified multipotent cardiac progenitors. Early multipotent cardiovascular progenitor cells expressing Nkx2–5 give rise to endothelial lineages, smooth muscles cells and cardiomyocytes [60].

Key interactions with tumor suppressors/ differentiation-related genes

Nkx2–5 has major interactions with GATA4 and Tbx5. It establishes a positive feedback loop with GATA4 and interacts with Tbx5 to enhance the differentiation of CPCs into cardiomyocytes. It directs cardiac looping by working with MEF2c, Hand1 and Hand2 [61]. Working with Nkx2–7, Nkx2–5 is involved in maintenance of cardiomyocyte cellular identity. The expression of Nkx2–5 is very significant during cardiomyocyte differentiation, as it acts as a repressor of FGF10 and Isl1 to enhance differentiation [62]. Nkx2–5 enhances cardiac phenotype by antagonizing TBX1 which is involved in the proliferation of CPCs [63]. Nkx2–5 interacts with BMP signaling pathway to enhance differentiation. As Nkx2–5 is a cardiac-specific patterning TF, its expression represses non-cardiac genes while inducing the gene expression of cardiogenic genes. The presence of Nkx2–5 expression sets controls in cardiac cells that give them cell type-specific features such as the permanent nature of cardiac cell type. TF Nkx2–5, working as master regulator of cardiac development, induces a cascade of regulatory and developmental genes [64]. This sets in motion the cell type-specific combinatorial code that governs cardiomyogenesis. In post-natal cardiomyocytes, it is needed for proper functioning. NPCEDRG is a novel tumor suppressive gene and has potential binding sites for Nkx2–5, thus inducing cell differentiation, controlling cell growth and regulating the cell cycle. Nkx2–5 influences and regulates gene activity of HOPX to modulate cardiac gene expression. HOPX has a potential tumor suppressive activity [65].

Contributions to combinatorial code/ cell type specific genetic-programming

Nkx2–5 sets in motion a cascade of combinatorial genetic interactions that govern the genetic programming of cardiogenesis. The Nkx2–5 based early patterning sets stage for BMP and Notch based gene expression. The defects in Nkx2–5 expression down-regulate BMPs and Notch signaling pathway, resulting in the disruption of cardiogenesis [66]. TF Nkx2–5 auto-regulates itself and is further mainly regulated by GATA4 and SMAD proteins. Its expression is also dependent on Isl1. In the induction of the TF Nkx2–5, the expression of BMP2/4 is required and the activity of Wnt-pathway is inhibited. The Wnt-signaling negatively impacts the TF Nkx2–5 gene expression, hence must be inhibited. The BMPs induces the FGF8 signaling to promote the development of cardiac proteins. Nkx2–5 is involved in the upregulation of HAND1 and HAND2, resulting in the differentiation and proliferation of cardiac cells [67]. The TF Nkx2–5 also has major interactions with p53, FGF16 and FGF10. Through its interactions, it controls proliferation and differentiation [68].

Presence in other tumors

The role of Nkx2–5 varies in different microenvironments; it may promote both proliferation and differentiation in different situations. Nkx2–5 is dysregulated in acute lymphoblastic leukemia (ALL), hepatocellular carcinoma (HCC), and T cell neoplasias, methylated in prostate adenocarcinoma, hyper-methylated in salivary gland adenoid cystic carcinoma [168]. Some other significant roles of Nkx2–5 in other tumors include its interactions with Mef2c in ALL, with Notch3 in T cell leukemias, dysregulated Nkx2–5 expression in sarcomas and hypermethylation of Nkx2–5 in breast, prostate and colon cancer [169]. It is expressed in papillary thyroid carcinoma (PTC) and reduces the expression of thyroid differentiation markers. There is age-related Nkx2–5 methylation in normal prostate tissues and may predispose to prostate adenocarcinoma [170, 171]. In ALL, Nkx2–5 has direct interactions with GATA genes and Mef2c oncogenic expression is influenced by Nkx2–5. The Mef2c expression inhibits apoptosis promoting NR4A1/NUR77 expression. Nkx2–5 is not expressed in hematopoietic stem cells, but in ALL it contributes to oncogenesis and interacts with BCL11 [172, 173] [174]. ,Importantly, Nkx2–5 deletions cause thyroid hypoplasia and this signifies its role in survival and proliferation as well as its various roles in different microenvironments.

Possible role in cardiac myxoma

Some studies have hinted towards the possible role of Nkx2–5 in this tumor development [69]. Nkx2.5/Csx, GATA-4, MEF2, and eHAND are key involved genes in CMs. Defects in Nkx2–5 cause abnormalities in atrial growth and development [70]. Nkx2–5, Oct-4, Isl1, and c-kit are upregulated and this produces cardiac progenitor stem cell-like state.

This study postulates that deviation of cardiomyocyes from cell type-specific well-differentiated state results in turning back of the cells into progenitor-like cardiac stem cells. The hallmark of this process is the upregulation of Nkx2–5 gene expression. Nkx2–5 has major interactions with p53 TS gene that also prevents this tumor from becoming malignant. Cardiomyocytes have very limited proliferation potential in adult life [71]. This nature of cardiomyocytes is governed by the cell type-specific programming that also restricts the proliferative potential of this cell type after completion of cardiogenesis. Nkx2–5 exerts vast control over proliferation. It has been found that it has wide range of functions depending on where it is expressed, as it enhances the gene expression of Mesenchymal Stem Cells (MSCs) in transplant patients and controls CPC proliferation [72].

The heterogeneity that exists in CM may be a consequence of the multitude of roles that Nkx2–5 and other key genes/TFs and signaling pathways play in different microenvironments and in different cell types. Their dysregulations result in the deviation of cells away from cell type-specific gene expression. As the cardiac-specific combinatorial code based functioning of TFs gets dysregulated, the direction of lineages deviates from one to multiple cell types suggesting a significant role of these key genes/TFs and signaling pathways in the developmental process.

Summary: unraveling the multifaceted roles of Nkx2–5

Nkx2–5, a pivotal player in cardiogenesis, orchestrates the early patterning of cardiac development, deciding atrial and ventricular fate. It controls cardiomyocyte differentiation through the JAK-STAT pathway and interacts with Mef2c to induce downstream signaling. Nkx2–5 promotes proliferation in cardiac progenitor cells (CPCs) by interacting with the Notch signaling pathway. In various tumors, Nkx2–5’s role varies, contributing to both proliferation and differentiation. This transcription factor’s presence in cardiac myxomas may relate to reprogramming cardiomyocytes into progenitor-like stem cells. Nkx2–5’s control over proliferation is vital in governing the limited proliferative potential of cardiomyocytes. Dysregulation of Nkx2–5 and other key genes/TFs and signaling pathways contributes to the heterogeneity in cardiac myxomas and their deviation from well-differentiated states, emphasizing their significance in cardiac tumor development.

GATA4

Role in cardiogenesis

GATA4 is a very important regulator of genes in the process of development. It plays a key role in the process of myocardial differentiation. The GATA4 also plays an essential role in testicular development. The key interactions include Nkx2–5, TBX5, SRF, HAND2, HDAC2, Erbb3, FOG-1 and FOG-2 [73, 74].

Proliferation-related roles

GATA4 plays a significant role in morphogenesis and promotes cardiomyocyte survival. When GATA4 is deleted or defective, Erb and Erk expression is down-regulated. They both normally play key role in EMT. GATA4 down-regulates the c-myc gene expression to promote differentiation process in cardiomyocytes during development. It also regulates hypertrophic growth of heart. Although GATA4 interacts with p53 and p21, it also works with Bcl2. This GATA4-Bcl2 interaction promotes cardiomyocyte survival [75, 76].

Key interactions with tumor suppressors/ differentiation-related genes

It is expressed in both embryonic and adult cardiomyocytes. It regulates the gene expression of many downstream cardiac genes. It also maintains the cardiac function in adult heart. GATA4 is an important regulator of terminal differentiation program in cardiomyocytes. It antagonizes c-myc to limit the replication potential. Multiple studies have suggested that damage to GATA4 also damages the Tbx5. This damage also contributes to congenital heart defects [77]. GATA4 plays a very significant role in differentiation process also by governing genes associated with cell-to-cell adhesion, cytoskeleton organization and extracellular matrix dynamics; this promotes them to become more differentiated and less proliferative [78]. It interacts with p53 and p21, which have TS effects. It is important to note that GATA4 interacts with CD40L and this way it is capable of inducing senescence. GATA4 also acts as a switch to activate NF-κB signaling [79].

Contributions to combinatorial code/ cell type specific genetic-programming

GATA4 works with other key cardiac TFs including Nkx2–5 and Tbx5. GATA4 is considered to be a key regulator of cardiac phenotype. It has upstream interactions with BMP, FGF and Wnt signaling pathways [175]. The significance of GATA4 can also be estimated from the fact that when ectopically its expression is induced together with Tbx5 and SMARCD3, this is capable of inducing genetic programming of cardiomyogenesis in non-cardiac regions of embryo. GATA4 regulates Mef2c expression and acts also as Isl1 enhancer. Note that GATA4 which is primarily involved in cardiomyocyte differentiation interacts with Mef2c and Isl1 both of which are involved in regulating progenitor and proliferation-related genes in CPCs [176]. Both GATA4-Tbx5 and Mef2c-Tbx5 work by triggering the gene expression of subsequent downstream cardiomyocyte-specific genes. GATA4 and Tbx5 are considered key regulators of cardiac gene regulatory networks. Nkx2–5 – GATA4 complex also plays role in cardiac hypertrophy in response to stretch. This complex interaction also governs the release of Atrial and Brain Natriuretic Peptides [177].

Presence in other tumors

In lung cancer, it plays the role of TS, as it down-regulates the Wnt7b and TGF-beta. The presence of SMAD4 and GATA4 is considered to be related to poor-prognosis in esophageal adenocarcinoma. Similarly, GATA4 is also upregulated in pancreatic cancer and other cancers. Different models have shown that upregulation will increase the process of differentiation [178]. However, it fails to halt or reduce proliferation in tumor microenvironments [179, 180]. In ALL, GATA4 has been associated with increased proliferation and inhibition of apoptosis. The predominant effect of specific genes and signaling pathways that are governing the landscape of a tumor may undermine the specific function of many differentiation-related genes [181, 182].

Possible role in cardiac myxoma

Primitive cardiomyocyte TFs have been detected in CM including GATA4, Mef2c, Nkx2–5 and eHAND [80]; they are slightly or even intensely positive in cardiac myxoma samples. In many samples, GATA4 gene expression was dysregulated. Decline or disruptions in gene expression of key regulatory differentiation genes such as GATA4 may have drastic impact on the overall genetic composition of differentiated cardiomyocytes. Such alterations can disrupt the delicate cell type-specific balance of expression among different types of genes/signaling pathways. This may contribute to switch the cells more towards a progenitor-like state, that is a hallmark of CM [81].

Summary: unraveling the multifaceted roles of GATA4

GATA4, a critical regulator in cardiogenesis, governs myocardial differentiation and is essential in testicular development. It interacts with various factors, including Nkx2–5, TBX5, SRF, HAND2, HDAC2, Erbb3, FOG-1, and FOG-2. GATA4 promotes cardiomyocyte survival, morphogenesis, and hypertrophic growth while down-regulating c-myc expression. It interacts with multiple tumor suppressors like p53 and p21. GATA4’s role extends to cardiac phenotype regulation, influencing BMP, FGF, and Wnt signaling pathways. It can induce genetic programming of cardiomyogenesis in non-cardiac regions. In tumors, GATA4 may have variable effects, acting as a tumor suppressor in lung cancer but upregulated in pancreatic cancer. In cardiac myxoma, alterations in GATA4 expression may shift cells toward a progenitor-like state, disrupting cell type-specific gene balance.

Tbx5

Role in cardiogenesis

Tbx5 is one of the key regulators of cardiogenesis. It is involved in promoting differentiation of CPCs into cardiomyocytes. It interacts with NKX2–5, GATA4 and BAF remodeling complex. Studies in which Tbx5 was deleted by CRISPR/Cas9 editing, showed that the cells maintained stem cell-like pluripotent state [82, 83]. Tbx5 is a key player in switching CPCs towards developmental gene expression by inducing differentiation into cardiomyocytes. Mutations in this key TF contribute to Atrial Septal Defect (ASD). It is essential for the development of heart and limbs. It is expressed in the embryonic, adult heart and in the endocardium of left ventricle [84,85,86].

Proliferation-related roles

In the ventricle, Tbx5 expression originates from the FHF but atrial gene expression originates from Mef2c in the SHF. Mef2c plays very important role in the proliferation of CPCs [87]. Tbx5 works with SHH in the formation of atrial septum. The TF Tbx5 has a very strong relationship with Nkx2–5, and Tbx5 – Nkx2–5 complex contributes to the process of cardiomyocyte differentiation. This complex also prevents activation of non-cardiac genes [88, 89].

Key interactions with tumor suppressors/ differentiation-related genes

Tbx5 is mutated in Holt-Oram syndrome. Tbx5 promotes other cardiogenic TFs. It is strongly interconnected with GATA4 and damage to GATA4 also damages Tbx5. The TF Tbx5 is so significant for the process of differentiation of cardiomyocytes that when it is defective, this contributes to the apoptosis [90, 91]. Tbx5 interacts with Nkx2–5, GATA4 and BAF60c to drive expression of cardiac genes. Tbx5 also interacts with repressor genes such as NuRD complex, SALL4 and others to downregulate the expression of non-cardiac genes. Moreover, Tbx5 induces the expression of downstream genes related to cardiomyocyte differentiation including NPPA and GJA5. Just like Tbx5-Nkx2–5 complex, Tbx5 also forms a complex with GATA5 and Mef2c to contribute to the process of cardiomyocyte differentiation. These partnerships by Tbx5 play cell type-specific key roles in the process of development [92,93,94].

Contributions to combinatorial code/ cell type specific genetic-programming

Tbx5 works with Nkx2–5 to promote cardiac differentiation. Tbx5 shifts the gene expression profile more towards cardiogenesis and it also plays key role in the beating of cardiomyocytes. In the entire process of cardiac development, the gene expression of Tbx5 is maintained, whereas it also persists in the adult heart. The key interactions of Tbx5 include Nkx2–5, GATA4, Baf60c, and Mef2c in cardiomyocyte development. It also interacts and regulates the gene expression of a cascade of downstream genes involved in cardiac differentiation. It inhibits the gene expression of neural and other non-cardiac cell types in cardiogenesis through Tbx5-NuRD interaction [183].

Presence in other tumors

Tbx5 inhibits cell proliferation in osteosarcoma. It is a critical regulator of oncogenesis. It has been found to suppress proliferation in Non-Small Cell Lung Cancer (NSCLC), acting as a TS. Even in normal embryonic developmental processes, its over-expression induces apoptosis and halts cell development. Tbx5 is epigenetically inhibited in colorectal cancer [184,185,186].

Possible role in cardiac myxoma

In the normal heart, the atrial expression of Tbx5 is far greater than the ventricular and Tbx5-Nkx2–5 forms a complex. This is very important as dysregulated expression of Nkx2–5 is considered to play a very significant role in the development of CM. Tbx5 forms key complexes that have a major effect in cell fate of cardiomyocytes, Tbx5 is involved in activation and maintenance of cardiac lineage genes as well. It prevents off-target binding of TFs in cardiac development. Hence, alterations in its gene expression may have profound consequences [95, 96]. It is not expressed in CM; this may be a defining feature in CM development as Tbx5 is one of the principal regulators of cardiomyocyte differentiation. Any dysregulation in Tbx5 can trigger a cascade of destruction by altering the direction of cell type towards mesenchymal progenitor-like state. In the development and maintenance of cardiomyocytes, Tbx5 suppresses the expression of genes involved in non-cardiac cell types. Hence, the dysregulations in Tbx5 may be a major contributor in the emergence of heterogeneity in CM.

Summary: unraveling the multifaceted roles of Tbx5

Tbx5 is a vital regulator in cardiogenesis, inducing CPC differentiation into cardiomyocytes through interactions with Nkx2–5, GATA4, and the BAF60c complex. Mutations can lead to Atrial Septal Defect (ASD), impacting heart and limb development. Tbx5 collaborates with Mef2c in CPC proliferation and prevents activation of non-cardiac genes. It interacts with GATA4, Nkx2–5, BAF60c, and Mef2c to drive cardiac gene expression, playing essential roles in cardiomyocyte development. Tbx5, together with Nkx2–5, shifts gene expression toward cardiogenesis and is involved in cardiomyocyte beating. Dysregulated Tbx5 expression is associated with cardiac myxoma development, potentially disrupting cell fate and gene expression, contributing to heterogeneity. In tumors, Tbx5 inhibits proliferation in osteosarcoma, acts as a tumor suppressor in lung cancer, and is epigenetically inhibited in colorectal cancer.

Mef2c

Role in cardiogenesis, contributions to the combinatorial code/cell type programming and key interactions

Mef2c works with Nkx2–5 in controlling the differentiation of CPCs. GATA4 works also by interacting with both Mef2c and Isl1, and they both have major roles in proliferation of progenitor cells. The Mef2c forms complexes with both key differentiation-related genes (GATA4 and Tbx5) of cardiomyocytes. Mef2c interacts with NF-κB and downregulates its signaling in multiple cell types in endothelial cells. The role of Mef2c is significant because of its individual effect on proliferation and also with the complexes it forms [97, 98]. Mef2c contributes to activation of the TF HAND1 [99, 100].

Proliferation-related roles

Mef2c is involved in cardiac morphogenesis, myogenesis, vascular development and neurogenesis. It contributes to maintaining differentiated state in muscle cells by working with other regulatory complexes. In hematopoiesis, ERK expression proportionally controls Mef2c expression. Mef2c plays oncogenic role in many cancers. One of the very important interactions of Mef2c includes its interactions with Tbx5 and GATA4. These interactions are of immense significance as Mef2c also plays key role in the proliferation of CPCs. The complexes that Mef2c forms with Tbx5 and GATA4, they contribute to switch the CPCs towards differentiated fate while sustaining the process of proliferation in cardiac development [101].

Presence in other tumors

Mef2c plays oncogenic role in ALL, Acute Myeloid Leukemia (AML), colon adenocarcinoma, Diffuse Large B Cell Lymphoma (DLBCL), and T-cell lymphomas. It also plays oncogenic role in prostate cancer and interacts with dysregulated notch signaling pathway. In hepatic cancer cells, it increases proliferative signaling. Mef2c acts as an essential transcription factor in AML oncogenesis. It interacts with Sox2 during the process of oncogenesis in cancer stem cells [187, 188]. CDKN1B deletions frequently coincide with the expression of Mef2c in ALL. Mef2c also plays oncogenic role in Chronic Myelogenous Leukemia (CML) and imatinib abrogates its expression. Common cascade pathways (p38 MAPKs-Mef2c) that can result in proliferation, differentiation and apoptosis work with genes IL1R and TGFBR in many breast cancer subtypes. Mef2c and Wnt signaling pathway both regulate SIX1 in Hodgkin Lymphoma. Mef2c exerts direct control over Socs2 [189]. The normal response of increased Mef2c expression is upregulation of Socs2. The Mef2c exerts oncogenic effects on Socs2 in different leukemias such as AML and ALL. Mef2c is also upregulated in Rhabdomyosarcomas [190,191,192]. Another important role of Mef2c is also seen in pancreatic cancer. YY1 acts as tumor suppressor, suppresses invasion and metastasis of pancreatic cancer cells by downregulating MMP10 which is upregulated by Mef2c.

Possible role in cardiac myxoma

Multiple studies have detected Mef2c gene expression in CM samples. As Mef2c works in the form of complexes with other key regulatory genes/pathways including GATA4, Isl1, Wnt-pathway, its role is also governed by microenvironment. It is capable of playing oncogenic role [102]. When key differentiation-related genes such as GATA4 become dysregulated, this may have drastic impact on the functioning of Mef2c which can ultimately go on to serve like an oncogene in CM landscape [103]. In such conditions, it may switch to work with Wnt and Isl1 resulting in the emergence of CPC-like state that is hallmark of CM [104, 105].

Summary: unraveling the multifaceted roles of Mef2c

Mef2c collaborates with Nkx2–5, GATA4, Tbx5, and Isl1 in controlling CPC differentiation and proliferation during cardiogenesis. It also forms essential complexes with key differentiation-related genes. Mef2c is involved in cardiac, muscle, vascular, and neurogenesis development and has interactions with NF-κB. In cancer, Mef2c plays oncogenic roles in various types, including ALL, AML, colon adenocarcinoma, lymphomas, prostate, and hepatic cancers. It interacts with different genes and pathways in these malignancies. Mef2c expression is detected in cardiac myxoma (CM) samples, where its role may be influenced by microenvironment and the dysregulation of key differentiation genes. This could contribute to a progenitor-like state, a hallmark of CM.

HAND1/2

Role in cardiogenesis, contributions to the combinatorial code/cell type programming and key interactions

HAND1/2 is expressed in the adult heart and is downregulated in cardiomyopathies, it modulates cardiac hypertrophy and is also involved in heart, vascular, gastrointestinal tract, limb and neuronal development. Mef2c contributes to the activation of HAND1. HAND1 plays a key role in neural crest development. It also interacts with BMP4 which contributes further to the differentiation of cardiomyocytes [106, 107]. It has major interactions with Nkx2–5 and GATA4. It encourages proliferation with Nkx2–5 and when it interacts with GATA4, it affects differentiation of cardiomyocytes. It is important to remember that it also has a TS effect [108, 109].

Presence in other tumors

HAND2 also acts as TS. It is downregulated in many tumors such as NSCLC and other cancers including ovarian, breast, gastric, colorectal, cervical, endometrial, prostate and esophageal squamous cell cancer [193]. But in the micro-environment of HCC it promotes tumor development. In the normal liver, the gene expression of HAND2 is undetectable. But in some samples of HCC, it has been found downregulated. In HCC, HAND2 interacts with BMP signaling cascade. Due to limitations of data on this role of HAND2, it is not possible to draw concrete conclusions about the role of HAND2 in HCC [194]. HAND2 negatively regulates TGFbeta, ROCK2 and JAK-STAT pathway [195, 196].

Possible role in cardiac myxoma

Detected in many but not all cases of CM. It is considered to be involved in the development of CM [110]. HAND1/2 acts as TS. Thus, it may have a possible contributing role in limiting the regenerative potential of cardiomyocytes and may have a contributing role in the benign nature of CM. This may also prevent the emergence of primary malignant tumors in cardiomyocytes.

Summary: unraveling the multifaceted roles of HAND1/2

HAND1/2 is expressed in the heart tissues, modulating cardiac hypertrophy. It also contributes to the vascular, gastrointestinal tract, limb, and neuronal development. It interacts with Mef2c and BMP4, promoting cardiomyocyte differentiation. HAND1/2 plays pivotal roles by interacting with Nkx2–5 and GATA4: it encourages proliferation alongside Nkx2–5 and promotes differentiation with GATA4, while also acting as a tumor suppressor (TS). In other cancers, HAND2 acts as a TS, downregulated in numerous cancer types, including NSCLC, ovarian, breast, gastric, colorectal, cervical, endometrial, prostate, and esophageal squamous cell cancer. However, in hepatocellular carcinoma (HCC), it may promote tumor development, interacting with the BMP signaling cascade. In cardiac myxoma, HAND1/2 is detected in many cases, potentially limiting cardiomyocyte regenerative potential, contributing to the benign nature of CM, and preventing primary malignant tumors in cardiomyocytes.

MYOCD

Role in cardiogenesis, contributions to the combinatorial code/cell type programming and key interactions

Mostly MYOCD works with p16 against the TGF-beta signaling, it induces growth arrest and also inhibits cellular proliferation by inhibiting NF-κB signaling. This is important because MYOCD-SRF axis forms a major complex with Mef2c to exert control on cardiac progenitors. This is involved in cardiomyocyte survival and maintenance of heart function. When MYOCD is defective, pro-apoptotic factors take over the control of cardiomyocytes. MYOCD is also involved in maintaining cardiac structural organization [111,112,113]. It interacts with Nkx2–5 to enhance proliferation. But proliferation is downregulated when SMAD3 gene expression is present. MYOCD also interacts with NFAT, HNRNPA1, SRF and Mef2c to enhance proliferation.

Role in proliferation, differentiation and in some other tumors

It inhibits stemness in NSCLC as it is an essential TS. It is downregulated in lung squamous cell carcinoma and lung adenocarcinoma. It inhibits stemness by inhibiting TGF-beta receptor signaling. The SRF-MYOCD axis is driver of well-differentiated leiomyosarcoma [197]. But MYOCD functions are also governed by the interactive complexes it forms with key regulatory genes. When MYOCD forms an interactive loop with SMAD3/4, it derives TGF-beta based Epithelial mesenchymal transition (EMT) [198, 199]. MYOCD, which also has TS effect, is repressed through proliferative signaling by FOXO3A and KLF4/KLF5. The TS P53 also has a dose dependent regulatory repressor effect on MYOCD. GSK3-beta can inhibit MYOCD-dependent cardiac gene expression. The activators of MYOCD include p300. MYOCD is also inhibited by its ERK1/2 based phosphorylation [200,201,202,203].

Possible role in cardiac myxoma

There are no proper data on the role of MYOCD in CM. But it may have a possible significant role in the process of cardiac tumorigenesis. Based on its interactions with key TFs and its role in inhibiting the stemness-related progenitor genes and signaling pathways, MYOCD may have a profound role in preventing the occurrence of primary tumors in cardiac tissue. As it works together with Nkx2–5 which is expressed in CM cells, MYOCD may have a role in maintaining the benign nature of cardiac myxoma and in preventing the occurrence of malignant tumors in cardiac tissue.

Summary: unveiling the multifaceted roles of MYOCD

MYOCD primarily collaborates with p16 to counteract TGF-beta signaling, inducing growth arrest and inhibiting cellular proliferation. Through the MYOCD-SRF axis, it forms a significant complex with Mef2c, impacting the regulation of cardiac progenitors, enhancing cardiomyocyte survival, and maintaining heart function. Defects in MYOCD may lead to the dominance of pro-apoptotic factors, disrupting cardiomyocyte regulation and cardiac structural organization. Concerning proliferation, MYOCD interacts with Nkx2–5 to enhance it but downregulates when SMAD3 is present. Inhibitory interactions with NFAT, HNRNPA1, SRF, and Mef2c also contribute to proliferation. MYOCD acts as a tumor suppressor by inhibiting stemness in non-small cell lung carcinoma (NSCLC), downregulated in lung squamous cell carcinoma and lung adenocarcinoma. Although the role of MYOCD in cardiac myxoma is not well-documented, it may play a crucial part in preventing primary tumors in cardiac tissue. Its interactions with key TFs and its influence on inhibiting stemness-related genes and signaling pathways could contribute to maintaining the benign nature of cardiac myxoma and preventing malignant tumors in cardiac tissue.

MSX2

Role in cardiogenesis, contributions to the combinatorial code/cell type programming and key interactions

In cardiogenesis, MSX2 interacts with HAND1/2 and they regulate the gene expression of each other. MSX2 regulates survival of SHF precursors by protecting them against apoptosis. It also makes sure that there are no excessive proliferations of cardiac cells, cardiac neural crest cells and endothelial cells. It acts more as a regulator by interacting with both proliferation-related genes and differentiation related genes. MSX1/2 are required for EMT of atrioventricular cushions and patterning of atrioventricular myocardium [114,115,116].

Role in proliferation, differentiation and in some other tumors

MSX2 functions to maintain a balance between survival and apoptosis. Its upregulation enhances malignant phenotype [204, 205]. It also acts as transcriptional repressor. It induces EMT in pancreatic cancer [206]. MSX2 working with RAS promotes cell growth. MSX2 is downstream target of RAS. The MSX2 expression is upregulated in diabetes and colorectal cancer [207, 208]. The MSX2 interacts with SOX2 to control cancer stem cell-like characterization in oral squamous cell carcinoma (SCC). MSX2 represses tumor stem cell phenotypes by SOX2 dysregulations in SCC [209]. The in vitro expression of MSX2 has been found to inactivate AKT pathway to promote cell cycle arrest and apoptosis [210].

Possible role in cardiac myxoma

There are no proper data on the role of MSX2 in CM. As its function is dependent on its interactions and cross-talk, it also varies with microenvironment. Hence, in CM its role is more likely to be dependent on tumor microenvironment. Such as in advanced CM, it may possibly contribute to tumorigenesis by promoting progenitor-like state.

Summary: unraveling the multifaceted roles of MSX2

MSX2 regulates gene expression with HAND1/2 in cardiogenesis, ensuring survival of SHF precursors and preventing excessive proliferation. It balances survival and apoptosis in proliferation, with upregulation enhancing malignancy. In diabetes and colorectal cancer, MSX2 is upregulated. MSX2 interacts with SOX2 in oral SCC to control cancer stem cell-like traits. In cardiac myxoma, MSX2’s role depends on the tumor microenvironment, potentially promoting a progenitor-like state in advanced cases.

HOPX

Role in cardiogenesis, contributions to the combinatorial code/cell type programming and key interactions

HOPX is expressed when CPCs become committed to cardiomyocyte fate. The niche signals help regulate the committed state. It interacts with activated SMADS to repress Wnt-signaling pathway [117]. It switches the cells more towards differentiated fate of cardiomyocytes by promoting local BMP signals to inhibit Wnt-signaling pathway, thus promoting cardiomyogenesis [118, 119].

Role in proliferation, differentiation and in some other tumors

HOPX inhibits Wnt-signaling; this causes HOPX to trigger stem cell quiescence and also explains the role of HOPX as TS by acting as RAS inhibitor. The downregulation of HOPX expression contributes to colorectal, head, neck and other cancers. It plays a critical role in cell type homeostasis [211,212,213].

Possible role in cardiac myxoma

There are no proper data on the role of HOPX in CM. The dysregulations in HOPX may possibly serve to contribute towards CM development. The downregulation in its gene expression may alter the genetic landscape of cardiomyocytes as HOPX plays key roles in differentiation and also acts as a TS. HOPX dysregulations may lead to switching the gene expression in the direction of progenitor-like state, as it is present in CPCs.

Summary: unraveling the multifaceted roles of HOPX

HOPX is expressed during CPC commitment to cardiomyocytes, interacting with activated SMADS to repress Wnt signaling, promoting cardiomyogenesis. It inhibits Wnt signaling and serves as a tumor suppressor (TS) by inhibiting RAS. Downregulation of HOPX contributes to various cancers. In cardiac myxoma (CM), HOPX’s role remains unclear, but its dysregulation may influence CM development by altering the genetic landscape, potentially pushing gene expression toward a progenitor-like state present in CPCs.

Key cardiac signaling pathways

Wnt signaling pathway

Role in cardiogenesis and key interactions

Wnt plays a very important role in cardiac development also by contributing to planar cell polarity in cardiogenesis. The Wnt-signaling is also involved in adult heart remodeling. It also contributes to cardiac hypertrophy and increases ANP gene expression. Reduced Wnt levels have been linked to premature myocardial infarction. Wnt3a is involved in cardiac progenitor renewal. This pathway is involved in cardiogenesis and cardiac disease development [120, 121]. Wnt-signaling pathway promotes fibrosis in cardiac repair. This is a very important factor in defining the limitations of cardiac regeneration. The Secreted frizzled-related protein (SFRP) based downregulation of Wnt/beta-catenin is cardio-protective as it inhibits fibrosis and inflammation. This impact of SFRP gene expression causes EMT in post myocardial infarction state. The Wnt/beta-catenin pathway promotes proliferation in CPCs and its inhibition promotes differentiation [122, 123].

Role in proliferation, differentiation and in tumorigenesis

This pathway contributes to stemness in hematopoietic stem cells. In cancers, abnormal Wnt-signaling contributes to the maintenance of cancer stem cells. Wnt/beta-catenin is upregulated in ALL and Chronic lymphocytic leukemia (CLL). It interacts with Notch signaling too in cancer microenvironment. The APC TS also plays important role in regulating this signaling pathway. Inhibiting Wnt-pathway increases apoptosis in CLL [214,215,216]. In melanoma, it promotes tumor growth through abnormal Wnt5a. It is also upregulated in breast cancers and its upregulation silences its repressors [217, 218]. The loss of PTEN TS and c-myc amplifications are linked to abnormal Wnt-signaling. In tumorigenesis, this pathway derives tumor development [219].

Wnt/beta-catenin pathway has massive influence over other key genes such as TSs including Numb and it is capable of repressing the numb gene expression. This results in the maintenance of cancer stem cells. This is also one of the mechanisms for immune evasion by cancer stem cells. This pathway is also involved in EMT and is upregulated in colorectal cancer, prostate, pancreatic and many other cancers [220, 221].

Possible role in cardiac myxoma

When Wnt-signaling pathway is disrupted, it contributes to upregulation of the gene expression of progenitor-like signatures [124]. Wnt/beta-catenin maintains telomeres through Telomerase Reverse Transcriptase (TERT) gene. When this signaling pathway combines with NF-κB signaling pathway, it contributes to dedifferentiation into stem cell-like state [125]. As the Wnt-signaling also plays important role in early stages of cardiogenesis, hence this dedifferentiation-related role may have possible implications in CM development.

Summary: unraveling the multifaceted roles of Wnt signaling pathway

The Wnt signaling pathway is vital in cardiogenesis, influencing planar cell polarity and adult heart remodeling. It plays a role in cardiac hypertrophy, progenitor renewal, and fibrosis. Dysregulation of Wnt signaling is linked to myocardial infarction. In proliferation, it impacts stemness in hematopoietic stem cells and cancer stem cells, contributing to tumorigenesis in various cancers. The pathway interacts with Notch signaling and regulates TS genes. In the context of cardiac myxoma (CM), disrupted Wnt signaling may lead to gene expression patterns resembling progenitor-like states, potentially influencing CM development. Wnt signaling is involved in both cardiac development and tumorigenesis, making it significant in understanding CM.

FGF signaling pathway

Role in cardiogenesis, proliferation and key interactions

The FGF Signaling Pathway is involved in the differentiation of stem cells to SHF progenitors and is also involved in the maintenance of pluripotency. These effects are based on interactions and complexes which FGF signaling pathway forms in order to exert effect on cell fate [126]. FGF2 inhibits TGF-beta1 and promotes cardio-protection. It is also involved in epicardial EMT, coronary vasculogenesis and angiogenesis through FGF1. The FGF Signaling Pathway interacts with the IGF1/2, VEGF, BMPS, TGF-Beta, Wnt and Notch signaling pathway. FGF10 and FGF8 contribute to the proliferation of SHF progenitor cells [127]. The FGF-MAPK axis promotes CPCs multi-potency. FGFs also have major interaction with PI3K/AKT pathway [128, 129]. In cardiogenesis, FGF2-Wnt complex exerts influence over human pluripotent stem cells to shift them into CPCs by suppressing GSK3-beta [130,131,132].

Role in differentiation and in tumorigenesis

FGF2-BMP2 complex promotes cardiomyocyte differentiation. Isl1-Tbx1 positively interacts with FGF10, which contributes to differentiation of CPCs. Nkx2–5 negatively regulates FGF10, which is involved in promoting cardiomyocyte differentiation [222]. In cardiomyocyte differentiation, GATA4 interacts with FGF16 and suppresses proliferation potential. It also provides postnatal cardio-protection. The FGF16 negatively regulates FGF2-RAS-MAPK complex [223, 224].

In postnatal adult cardiomyocytes, FGF Signaling plays very important role in modulating proliferation; FGF1 is involved in homeostasis and remodeling [225]. FGFs have multifunctional roles ranging from proliferation, homeostasis to differentiation. FGF acts as blocker of premature CPCs differentiation. The FGF-BMP crosstalk plays key regulatory role in governing cardiomyocyte differentiation [226, 227]. The FGF Signaling Pathway is downregulated by BMP4-MSX1 complex which promotes differentiation of neural crest cells. The FGF Pathway interacts with Nkx2–5 to produce more profoundly the ventricular characteristics in the developing heart [228, 229].

Possible role in cardiac myxoma

The FGF Signaling Pathway may have significant role in CM development as loss of FGF causes gradual accumulation of atrial cells [133]. It is important to note that most CMs originate in the atria. The loss of FGF has such immense impact that it causes ectopic atrial gene expression in ventricles. One of the most important impacts of the sustained FGF signaling is that it acts to suppress cardiomyocyte plasticity. This may also point to the origins of CM [134].

Summary: unraveling the multifaceted roles of FGF signaling pathway

The FGF Signaling Pathway plays a crucial role in cardiogenesis by influencing stem cell differentiation to SHF progenitors, maintaining pluripotency, and promoting cardiomyocyte differentiation. It interacts with various signaling pathways, including IGF1/2, VEGF, BMPs, TGF-Beta, Wnt, and Notch. FGF2-Wnt complex shifts pluripotent stem cells to CPCs, suppressing GSK3-beta. FGFs, such as FGF10 and FGF8, drive SHF progenitor cell proliferation. In postnatal cardiomyocytes, FGF signaling modulates proliferation and homeostasis. FGF1 maintains adult cardiomyocyte homeostasis and remodeling. Dysregulation of the FGF pathway may contribute to CM development by promoting ectopic atrial gene expression in ventricles and suppressing cardiomyocyte plasticity. Most CMs originate in the atria, highlighting the pathway’s significance.

BMPs

Role in cardiogenesis and key interactions

BMPs downregulate the expression of progenitor genes in CPCs. It enhances differentiation of cardiomyocyte. They induce some progenitor genes as well. The BMP signaling pathway downregulates Isl1, Tbx1, FGF10 and switches the gene expression towards cardiomyocyte differentiation [135]. When BMP-signaling is defective, the gene expression of HAND2 and Nkx2–5 remains unchanged [136, 137]. BMPs interact with Nkx2–5, HAND2, Tbx2 and Tbx20 to promote cardiomyocyte differentiation. It is also involved in epicardial EMT which is regulated by both TGF-beta and BMPs. SMADs negatively regulate TGF-beta [138, 139].

Role in proliferation, differentiation and in tumorigenesis

BMPs have dual role in tumorigenesis. They are capable of acting both as TSs and promotors of tumor development. This is based on microenvironment and overall profile of governing key regulatory genes. For example, the absence of BMPs causes the progression of colorectal carcinoma. In Barrett’s esophagus, the BMP-signaling pathway is upregulated. BMP4 also contribute to neural development. BMPs interact with K-RAS and are upregulated in NSCLC [230]. BMPs are also involved in adult tissue homeostasis. In cardiogenesis, BMP2 causes the differentiation of CPCs. Similarly, BMP10 reduces the cardiomyocyte proliferation potential [231]. The gene expression of BMP2 in cardiac cushions causes EMT myocardial patterning. The role of BMPs is influenced by the microenvironment [232]. The BMP-signaling pathway also acts on the progenitor genes. It promotes the gene expression of Oct-4 and Nestin. They are among the key genes involved in stem cells [233]. Another fascinating feature of BMPs includes their interactions with TSs such as p53, p21, SMADs and cause repression of TGF-beta. When p53 is mutated, Wnt-signaling pathway is upregulated. As a result of Wnt-pathway upregulation, the interconnected loop of BMP signaling becomes dysregulated [234, 235]. BMPs have been found to act as TSs in Renal Cell Carcinoma, Glioblastoma, esophageal adenocarcinoma, prostate adenocarcinoma, diffuse gastric adenocarcinoma and others [236, 237].

The role of BMPs in HCC is different and of immense significance as these cells have vast regenerative potential; they contribute towards G1 to S transitions through cyclins [238,239,240].

Possible role in cardiac myxoma

BMPs may possibly have a very significant role in CM development as it is involved in cardiomyocyte differentiation during the process of cardiogenesis. It is also involved in limiting the cardiac regenerative potential. Further studies should be conducted to evaluate the role of BMPs in CM development.

Summary: unraveling the multifaceted roles of BMPs

BMPs are vital in cardiogenesis, promoting cardiomyocyte differentiation by downregulating progenitor genes in CPCs. They interact with Nkx2–5, HAND2, Tbx2, and Tbx20 to facilitate cardiomyocyte differentiation and regulate epicardial EMT. In tumorigenesis, BMPs exhibit a dual role, acting as both tumor suppressors and promoters based on microenvironment and gene interactions. BMPs interact with key genes like p53, p21, and SMADs to repress TGF-beta and influence cell behavior. Their role in HCC is especially significant due to their impact on G1 to S transitions. In cardiac myxoma, BMPs may play a substantial role in CM development by influencing cardiomyocyte differentiation and limiting regenerative potential, but further research is needed to fully understand this role.

Notch signaling pathway

Role in cardiogenesis and key interactions

It is involved in cardiomyocyte proliferation, differentiation, cell fate specification and patterning [140]. Its specific role depends on its interactions with other key regulatory genes such as with BMP2 to promote cardiomyocyte differentiation [141]. Similarly, it interacts with activins and PI3K/AKT pathway to promote mesenchymal state in CPCs. The notch pathway interacts with p21, c-myc, snail1/2, TGF-beta and in EMT it interacts with Dll4, Jag1, BMP2, Alk3/6 and other key regulatory genes [142, 143]. ,In EMT, it also works through important interactions with snail1/2-TGFbeta. In SHF, Notch regulates BMP4 and FGF8 gene expression [144,145,146].

Role in proliferation, differentiation and in tumorigenesis

The notch pathway in cancer contributes to the stemness of cancer stem cells [241]. It interacts with proto-oncogenes and inflammatory pathways. It also has strong cross-talk with FGF and Wnt-signaling pathways [242]. Notch has key interactions with many TSs such as PTEN, P53, P21 and others. The network of these key cross-talks governs the direction of cell fate, and dysregulations in such key regulators contribute to the disease development including cancers [243, 244].

Possible role in cardiac myxoma

Notch may have possible implications in CM origins because of interactions with key cardiac TFs such as Isl1 and Mef2c. Notch expression increases postnatal cardiac survival and contributes to the proliferation of CPCs in cardiac development. The Notch signaling pathway governs cardiac tissue renewal by maintaining CPCs in a committed state. Further studies should be conducted to evaluate the role of Notch in CM development [1, 15, 19].

Summary: unraveling the multifaceted roles of notch signaling pathway

The Notch signaling pathway is crucial in cardiogenesis, influencing cardiomyocyte proliferation, differentiation, and cell fate. Its interactions with BMP2, activins, PI3K/AKT, p21, c-myc, and TGF-beta determine specific roles in CPCs and EMT. In SHF, it regulates BMP4 and FGF8 gene expression. In cancer, Notch contributes to stemness, interacting with proto-oncogenes, inflammatory pathways, FGF, and Wnt signaling, alongside multiple tumor suppressors like PTEN and P53. Dysregulations in this network impact disease development. Notch may have implications in CM due to interactions with cardiac TFs like Isl1 and Mef2c, supporting postnatal cardiac survival, CPC proliferation, and cardiac tissue renewal. Further research is needed to explore Notch’s role in CM development.

Occurrence of Cardiac Myxoma in Carney complex, pointing towards the significance of results in this study

The mutation in PRKAR1A causes myxomas and carney complex, and multiple myxomas are a feature of Carney complex [148].

PRKAR1A acts also as a TS. The mutated PRKAR1A also causes other tumors including thyroid tumors because of increase in gene expression of RET/PTC2 signaling, multiple endocrine neoplasias and myxomas [149]. Mutations in PRKAR1A lead to the onset of dysregulated c-AMP protein kinase A signaling. CMs occur in 20–40% of Carney Complex patients and can occur in many chambers [147]. The nature of mutation in PRKAR1A also points towards the origin of CM as postulated in this study. This signifies how important the role of differentiation-related genes/TSs is in the maintenance of cell type-specific gene expression in cardiomyocytes. It also signifies how the defects in such key regulatory genes such as PRKAR1A can result in switching of cardiac cells towards a mesenchymal-like progenitor state present in CM [150, 151]. The CM development in carney complex also signifies the role of TSs and differentiation-related genes/TFs in maintenance, homeostasis of cardiomyocytes and also in tumorigenesis.

Discussion

Transformation of cardiomyocytes into progenitor-like state, hallmark of CM

Cardiac cells undergoing a transformation into a progenitor-like state, a distinctive hallmark of this benign cardiac tumor. This transformation is among the most significant possible ways of CM tumorigenesis. This reversion is attributed to dysregulations in key cardiac genes, transcription factors (TFs), and signaling pathways involved in the control of cardiomyocyte differentiation and maintenance of cardiac cell fate. CM develops when genes/TFs/signaling pathways with proliferation-related roles are upregulated and differentiation regulators are downregulated [7, 8]. The resulting tumor microenvironment also switches many factors towards tumorigenesis [5, 6].

The cardiac genetic landscape meticulously balances between genes, transcription factors (TFs), and pathways governing proliferation and differentiation. Dysregulations in the gene expression of key differentiation regulators, including Tbx5, GATA4, HAND1/2, MYOCD, HOPX, and BMPs, serve as the catalysts for this remarkable reprogramming. These regulatory actors orchestrate a symphony that drives the once-differentiated cardiomyocytes to abandon their mature identities and embrace a more primitive, progenitor-like phenotype [262, 263]. Dysregulation in factors like Isl1, Baf60 complex, Wnt, FGF, Notch, and Mef2c plays a pivotal role in this complex process. The Nkx2–5 and MSX2 contribute predominantly to both proliferation and differentiation of Cardiac Progenitor Cells (CPCs), are capable of serving influential roles in the landscape of CM development [264, 265]. This highlights the intricate balance between differentiation, proliferation, and regulation of cardiac cell fate. The dysregulations in the interconnected networks of genes/TFs/signaling pathways emphasizes the re-differentiation of cardiomyocytes in CM development [152, 266].

The cross-talk and interactions in regulating the fate of cardiomyocytes

Cardiac development is a multistep developmental process. It is governed by CPCs that differentiate into cardiomyocytes. The entire process at every stage is governed by sequential unfolding of combinatorial code/ cell type-specific genetic programming which directs the cell type-specific genetic program to unfold [245]. The cascades of TFs/genes involved are governed also by cross-talk, the interaction between signaling pathways. These cross-talk based interactions cause the emergence of specific gene expression effect. The cardiac development process defines the nature and developmental architecture of cardiomyocytes [246]. This study has investigated cardiogenesis to trace the possible origins of CM and the relationship of limited cardiac regenerative potential with the rare occurrence/benign nature of tumorigenesis in cardiac tissues. The PTFs trigger the epigenetic programming in CPCs [247]. This sets in motion the unfolding of successive lineage specification. Further unfolding of the lineage, reshapes the genetic landscape towards cardiomyocyte development by establishing the commitment of cells towards differentiation. The committed CPCs emerge as a consequence of this effect. When PTFs combine with specific TFs, this causes initiation of cell program via unfolding regulatory networks. This process directs the development of CPCs towards cardiomyocytes [248].

The governing role of proliferation, differentiation and tumor suppressor genes in regulating the cell fate

The entire process involving unfolding of progenitor genes, cell type-specific genes, and associated functioning of regulatory genes, is governed very tightly through cross-talks and interconnectedness of genes/TFs with key regulatory genes. The differentiation-related genes and TSs switch the cell circuitry more towards differentiation pathways. There exists specific combinatorial code/ genetic programming for each cell type; Nkx2–5, Mef2c, GATA4, Tbx5 are among the key regulators of cardiomyocyte programming [249]. The combinatorial code/ cell type-specific genetic programming works also via cross-talk and tightly regulated relationships. The cross-talk of TFs with proliferation genes, differentiation/ TS genes, fate-specific genes, and others, govern the development and maintenance of cell type [250].

When the gene expression of key genes including Tbx5 that govern the programming of cell type becomes dysregulated, this can alter the homeostasis of cell type. This can lead to switching the gene expression of cardiomyocytes towards disease development such as dedifferentiation into the cardiac progenitor-like state, resulting in the emergence of CM [251].

Carney complex also signifies how differentiation/ TS genes govern cell fate and their defects can cause CM. The role of TSs in limiting cardiac regenerative and proliferative potential is also reflected by the TS-like effect exerted by the landscape of differentiation genes. The key TF Nkx2–5, which is a major TF in cardiogenesis works in close association with TSs by interacting with p53 and p21. This interaction modulates the activity of this key TF. The p53 is able to interact with both wild-type Nkx2–5 and mutant Nkx2–5 in cardiac tissue. The p21, which is a CDK inhibitor, works to regulate Mef2c expression. The cell type-specific interactions of key genes may vary in different microenvironments [252].

Stemness and TS/ differentiation genes controlling the cell fate, and their dysregulations resulting in disease development

The stemness and differentiated state are the two extremes of the state of cell. Here we are focusing primarily on the cardiomyocytes. As cardiomyocytes are differentiated cells with very limited regenerative potential and they have to survive for very long durations in life, they must maintain themselves in the differentiated state. For that purpose, they maintain a sufficient level of gene expression of differentiation-related genes and TSs [253]. As other cell types, including neuronal cells, cardiomyocytes don’t go into proliferative phase the way skin or hepatic cells do, hence with age the cell circuitry shifts more towards the enhanced expression of differentiation-related genes and TSs. This overall exerts an increased TS-like effect. These differentiation-related genes interact with TSs and exert massive influence over other genes including those involved in proliferation. Hence, with aging there are increased degeneration-related effects in these cell types such as neurodegeneration in neurons and sigmoid shape of heart with decline in cardiac function. In order for these cell types to remain in arrested cell cycle state, the TSs exert massive influence on the genomic landscape of these cell types. But due to aging, this effect becomes more pronounced and contributes also to degenerative diseases [254].

Cell types with long survivals and limited regeneration capacity including neurons and cardiomyoctes, maintain profound gene expression of differentiation-related and TS genes. With increasing age, this predisposes these cell types towards the risk of degeneration such as Alzheimer’s disease in neurons, sigmoid shape heart with decline in cardiac function and others [255]. Due to the accumulation of dysregulations in gene expression of the key regulators that are involved in the maintenance of differentiated state, the cells switch towards stem cell-like progenitor state, that is also the hallmark of CM. This is possibly because of the same reason that an inverse relationship exists between Alzheimer’s disease and cancer in terms of molecular mechanisms and cellular pathways [256], further signifying the importance of differentiation and proliferation genes in disease development. It is already well established that TSs such as p21, p27, p53, and others promote and induce differentiation. The p53 expression also induces differentiation in pluripotent stem cells by suppressing stemness transcription factors including Oct-4, nanog and others. Similarly, p53 and p63 downregulate Oct-4 and promote the process of differentiation.

The interconnectedness in the cell circuitry is so profound that mitogens/ proliferation-related genes also work through cross-talk and interconnectedness. For example, the c-Myc gene cooperates with BCL2, BCR /ABL and interacts with TSs too, including p53 gene. Other examples include the Ras TF, as it also drives proliferation like c-Myc; similarly FGFR3 works through tyrosine kinase and act as proto-oncogene. The Ras/Raf pathway and PI3K/AKT pathway interact with c-Myc, and the c-Myc works with p53. Similarly, cyclins /CDKs interact with EGFR resulting in massive increase in the gene expression of Ras/Raf and PI3K/AKT/mTOR pathway. These interactions and cross-talks also play key roles in pluripotent stem cells, development, post-natal cells, and disease development, including tumorigenesis [257].

Deviation of cardiomyocyes from cell type-specific well-differentiated state increases the risk of disease development, including the turning back of the cells into progenitor-like cardiac stem cells. Some studies have pointed to the dysregulated expression of Nkx2–5 in tumor development. As Nkx2–5 has major interactions with p53, this also prevents the tumor from becoming malignant. Such interactions have been described in detail in respective sections [258]. Cardiomyocytes have very limited proliferation potential in adult life. This nature of this cell type is governed by the cell type-specific programming that also restricts the proliferative potential of this cell type after completion of development in postnatal period [259].

Heterogeneity in cardiac myxoma

The key genes/TFs and signaling pathways also play important roles in other cells types. Due to dysregulations and deviation in the cell type-specific genetic programing based gene expression in CM development, many key regulators of cell type go in the direction of gene expression related to the other cell types. This results in the presence of tumor heterogeneity in CM. For example, Isl1 also plays a role in other cell types. It interacts with FGFR in ganglion cells during embryogenesis, essential regulators of pancreatic morphogenesis and differentiation, and facilitates neuronal differentiation [260]. Retinoic acid promotes the Isl1 expression in pancreatic endocrine differentiation. The Isl1/2 defects contribute to the developmental defects in motor neurons. Shh induces Isl1 expression in neural development. Other major interactions of Isl1 include: Ngn2, NeuroD4, NeuroM, Pax6 and Nkx2.2. Isl1 gene expression, that is key component of CPCs, is also detected in pancreas, brain, lung, thymus, ovary [261].

The heterogeneity that exists in CM, may be because of the multitude of roles that Nkx2–5, Isl1, GATA4, Tbx5 and other key genes/TFs and signaling pathways play in different microenvironments and in different cell types. The dysregulations in them result in the deviation of cells away from cell type-specific gene expression [262]. As the cardiac specific combinatorial code-based functioning of TFs get dysregulated, this deviates the direction of lineages from one specific to multiple cell types having significant role of these key genes/TFs and signaling pathways in their development [263].

Interconnected landscape of key regulatory genes governing the cell type-specific roles

Increased TS/ differentiation-related gene expression limits the expression of proto-oncogenes; this results in declining the risk of tumorigenesis but simultaneously limits the regenerative capacity. The key cardiac genes such as Nkx2–5 have major interactions with TSs. The genes/signaling pathways work in the form of association and combinations. Such combinations include the proliferating genes working with differentiation-related genes with key regulatory interactions with the TSs. This combination-based genetic functioning is very important part of cell type-specific combinatorial code/ genetic programming [264]. The heterogeneity in CM is because of the dysregulations in such key genes/TFs and signaling pathways. It is because of these disruptions in the interconnected working of key regulatory genes that disrupts the cell type-specific combinatorial functioning. This leads every key gene/TF to swing towards other cell lineage directions in which they also participate in development. The example of interconnected working is also seen in the TF Nkx2–5, which works in the interconnected network of Nkx2–5 – mef2c –p53, and includes many other interactions too. The TF Nkx2–5 contributes to proliferation through JAK-STAT pathway. This Nkx2–5 complex doesn’t work alone, it has positive feedback loop with GATA4 and interacts with Tbx5. This sets in motion the unfolding of cell fate specific machinery. It also down-regulates FGF10 and Isl1 for the promotion of differentiation. This also represses the expression of non-cardiac genes [265]. The presence of dysregulated Nkx 2–5 is present in cancers such as ALL, because there it is working in a very different microenvironment and with different regulatory genes such as Mef2c-Nkx2–5 complex, which plays oncogenic role. When the Nkx2–5 combines with TSs, it promotes differentiation, cell cycle regulation, and apoptosis. Its function is dependent on microenvironment, key interactions and cross-talk with regulatory genes [152, 266].

The targets for gene editing and epigenome editing in the development of future cardiac therapies

The decline in the gene expression of cardiac cell type-specific genes dysregulates the genetic programing of cardiac tissues involved in cell type-specific homeostasis. The multiple roles of every gene/TF begin to manifest themselves causing the emergence of heterogeneity [267, 268]. As the control exerted by the differentiating genes begin to decline, then the genetic landscape of CPCs begin to manifest. This switches the gene expression of cardiac tissues towards progenitor-like state, hallmark of the CM. The multipotent nature of CPCs begins to manifest itself in CM development. In adult cardiomyocytes, there remains a persistent expression of cardiac lineage-specific genes which maintains the cardiac tissue homeostasis [269]. This same persistent expression of the differentiation-related genes with TS effect also prevents the CM from becoming malignant. This effect possibly maintains the benign nature of primary benign cardiac tumors including CM, and overall contributes to the rare occurrence of primary cardiac tumors. The overall increased exertion of tumor suppressive-effect in this way also causes cardiomyocytes to have very limited regenerative potential because it antagonizes the expression of proliferation-related genes [270].

This study also provides the targets for gene-editing tools such as CRISPR gene editing or epigenome editing to correct or regulate the genes/signaling pathways which become dysregulated in CM development [271]. This study may serve as a map for genetic and epigenetic targets for the development of new therapeutic approaches towards reviving cardiac regenerative potential, targeting CM development and the development of cardiac organoids [272].

Conclusions

Cardiomyocytes are one of the cell types that have very limited regenerative potential and survive for a very long duration of time. For this purpose, they continuously need to maintain themselves in a well-differentiated state. Based on the process of cardiomyocyte development in cardiogenesis, the emergence of CM is possibly governed by the dysregulations in key cardiac genes/TFs and signaling pathways. The dysregulations in differentiation related genes/TFs including Tbx5, GATA4, MYOCD, HAND1/2, HOPX, MSX2, BMPs have profound effect on controlling the gene expression of cardiomyocytes. They also contribute to limiting the regenerative potential of cardiomyocytes. The defective or dysregulated gene expression of such key differentiation-related genes causes the switching of cardiomyocytes towards progenitor-like state by causing upregulation of progenitor and proliferation-related genes. The key signaling pathways including Wnt, BMPs, Notch, FGF signaling pathways also play key regulatory roles in cardiac tissues. In cardiac development, their roles are very tightly regulated. And they work through cross-talk and interactions with cardiac-programming genes and regulators. The dysregulations in Wnt, Notch, FGF are capable of contributing to the process of tumorigenesis. Similarly, BMPs have more profound role as TSs in many cancers and are key contributors to the process of differentiation. The possible reason that cardiomyocytes are unable to easily change into primary malignant tumors, it is because they are very strongly regulated in differentiated state through a loop of multiple interconnected differentiation and TS genes. The massive influence exerted by these genes also causes the limitations in the regenerative abilities of cardiomyocytes. And many PTFs of CPCs including Isl1, Nkx2–5 expressed in CPCs, they also function to maintain cardiac fate, or final cell type. The Mef2c and Baf60 also don’t function independently; they are also very tightly regulated and work in the form of complexes with key TFs of cardiomyocytes. Another example of control over stemness is the presence of MYOCD gene expression. It exerts control over stemness-related genes and prevents them from changing into a total stem cell-like state. The benign nature and rare occurrence of CM is a possible consequence of the limited cardiac proliferative/ regenerative potential. More research is needed in this area; this can be done by developing models of cardiac organoids focused on cardiogenesis, and gene-editing them to transform into CM.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Abbreviations

BMPs:

Bone Morphogenetic Proteins

CDKs:

Cyclin-dependent kinases

CM:

Cardiac Myxoma

CPCs:

Cardiac Progenitor Cells

EGFR:

Epidermal growth factor receptor

EMT:

Epithelial mesenchymal transition

FHF:

first heart field

FGF:

fibroblast growth factor

GATA4:

GATA Binding Protein 4

GSK3:

Glycogen synthase kinase 3

HAND1/2:

Heart and neural crest derivatives expressed protein ½

HOPX:

homeodomain-only protein homeobox

ISL1:

ISL LIM Homeobox 1

JAK:

Janus kinase

Mef2c:

myocyte enhancer factor 2C

MYOCD gene:

Myocardin

Myc:

MYC proto-oncogene

MSX2:

Msh Homeobox 2

MAPK:

Mitogen-activated protein kinase

mTOR:

Mechanistic target of rapamycin

NF-κB:

Nuclear factor kappa-light-chain-enhancer of activated B cells

Nkx2-5:

Homeobox protein Nkx-2.5

Notch:

Signaling pathway

Oct2:

Octamer transcription factor 2

PI3K:

Phosphoinositide-3 kinase

PRKAR1A:

cAMP-dependent protein kinase type I-alpha regulatory subunit

PTEN:

Phosphatase and tensin homolog

STAT:

Signal transducer and activator of transcription

SHF:

second heart field

Tbx5:

T-box transcription factor 5

TFs:

Transcription Factors

TGF-β:

Transforming Growth Factor-β

TSs:

Tumor Suppressors

Wnt:

Signaling pathway

References

  1. Singhal P, Luk A, Rao V, Butany J. Molecular basis of cardiac myxomas. Int J Mol Sci. 2014;15(1):1315–37. https://doi.org/10.3390/ijms15011315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Nugraha B, Buono MF, von Boehmer L, Hoerstrup SP, Emmert MY. Human cardiac organoids for disease modeling. Clin Pharmacol Ther. 2019;105(1):79–85. https://doi.org/10.1002/cpt.1286.

    Article  PubMed  Google Scholar 

  3. Mazzola M, Di Pasquale E. Toward cardiac regeneration: combination of pluripotent stem cell-based therapies and bioengineering strategies. Front Bioeng Biotechnol. 2020;8:455. https://doi.org/10.3389/fbioe.2020.00455.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Verjans R, van Bilsen M, Schroen B. Reviewing the limitations of adult mammalian cardiac regeneration: noncoding RNAs as regulators of Cardiomyogenesis. Biomolecules. 2020;10(2):262. https://doi.org/10.3390/biom10020262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hashimoto H, Olson EN, Bassel-Duby R. Therapeutic approaches for cardiac regeneration and repair. Nat Rev Cardiol. 2018;15(10):585–600. https://doi.org/10.1038/s41569-018-0036-6.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Puente BN, Kimura W, Muralidhar SA, Moon J, Amatruda JF, Phelps KL, et al. The oxygen-rich postnatal environment induces cardiomyocyte cell-cycle arrest through DNA damage response. Cell. 2014;157(3):565–79. https://doi.org/10.1016/j.cell.2014.03.032.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Breckwoldt K, Weinberger F, Eschenhagen T. Heart regeneration. Biochim Biophys Acta. 2016;1863(7 Pt B):1749–59. https://doi.org/10.1016/j.bbamcr.2015.11.010.

    Article  CAS  PubMed  Google Scholar 

  8. Zhang Y, Mignone J, MacLellan WR. Cardiac regeneration and stem cells. Physiol Rev. 2015;95(4):1189–204. https://doi.org/10.1152/physrev.00021.2014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Doevendans PA, van Bilsen M. Transcription factors and the cardiac gene programme. Int J Biochem Cell Biol. 1996;28(4):387–403. https://doi.org/10.1016/1357-2725(95)00145-x.

    Article  CAS  PubMed  Google Scholar 

  10. Hong JH, Zhang HG. Transcription factors involved in the development and prognosis of cardiac remodeling. Front Pharmacol. 2022;(13):828549. https://doi.org/10.3389/fphar.2022.828549.

  11. Zhu L, Liu K, Feng Q, Liao Y. Cardiac organoids: a 3D Technology for Modeling Heart Development and Disease. Stem Cell Rev Rep. 2022; https://doi.org/10.1007/s12015-022-10385-1.

  12. Filippo Buono M, von Boehmer L, Strang J, Hoerstrup SP, Emmert MY, Nugraha B. Human cardiac organoids for modeling genetic cardiomyopathy. Cells. 2020;9(7):1733. https://doi.org/10.3390/cells9071733.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Drakhlis L, Devadas SB, Zweigerdt R. Generation of heart-forming organoids from human pluripotent stem cells. Nat Protoc. 2021;16(12):5652–72. https://doi.org/10.1038/s41596-021-00629-8.

    Article  CAS  PubMed  Google Scholar 

  14. Petris AO, Alexandrescu DM, Costache II. Cardiac tumors. Rev Med Chir Soc Med Nat Iasi. 2014;118(2):289–92.

    CAS  PubMed  Google Scholar 

  15. Rahouma M, Arisha MJ, Elmously A, El-Sayed Ahmed MM, Spadaccio C, Mehta K, et al. Cardiac tumors prevalence and mortality: a systematic review and meta-analysis. Int J Surg. 2020;76:178–89. https://doi.org/10.1016/j.ijsu.2020.02.039.

    Article  PubMed  Google Scholar 

  16. Hori Y, Tanimoto Y, Takahashi S, Furukawa T, Koshiba-Takeuchi K, Takeuchi JK. Important cardiac transcription factor genes are accompanied by bidirectional long non-coding RNAs. BMC Genomics. 2018;19(1):967. https://doi.org/10.1186/s12864-018-5233-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Voges HK, Mills RJ, Elliott DA, Parton RG, Porrello ER, Hudson JE. Development of a human cardiac organoid injury model reveals innate regenerative potential. Development. 2017;144(6):1118–27. https://doi.org/10.1242/dev.143966.

    Article  CAS  PubMed  Google Scholar 

  18. Scalise M, Marino F, Salerno L, Cianflone E, Molinaro C, Salerno N, et al. From spheroids to organoids: the next generation of model Systems of Human Cardiac Regeneration in a dish. Int J Mol Sci. 2021;22(24):13180. https://doi.org/10.3390/ijms222413180.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Leja MJ, Shah DJ, Reardon MJ. Primary cardiac tumors. Tex Heart Inst J. 2011;38(3):261–2.

    PubMed  PubMed Central  Google Scholar 

  20. Nakada Y, Kimura W, Sadek HA. Defining the limit of embryonic heart regeneration. Circulation. 2015;132(2):77–8. https://doi.org/10.1161/CIRCULATIONAHA.115.017070.

    Article  PubMed  Google Scholar 

  21. Ghiroldi A, Piccoli M, Cirillo F, Monasky MM, Ciconte G, Pappone C, et al. Cell-based therapies for cardiac regeneration: a comprehensive review of past and ongoing strategies. Int J Mol Sci. 2018;19(10):3194. https://doi.org/10.3390/ijms19103194.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Caspi O, Gepstein L. Insights from the third dimension: cardiac organoids help identify regenerative pathways. Cell Stem Cell. 2019;24(6):833–4. https://doi.org/10.1016/j.stem.2019.05.012.

    Article  CAS  PubMed  Google Scholar 

  23. Varzideh F, Pahlavan S, Ansari H, Halvaei M, Kostin S, Feiz MS, et al. Human cardiomyocytes undergo enhanced maturation in embryonic stem cell-derived organoid transplants. Biomaterials. 2019;192:537–50. https://doi.org/10.1016/j.biomaterials.2018.11.033.

    Article  CAS  PubMed  Google Scholar 

  24. Cho J, Lee H, Rah W, Chang HJ, Yoon YS. From engineered heart tissue to cardiac organoid. Theranostics. 2022;12(6):2758–72. https://doi.org/10.7150/thno.67661.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gao R, Liang X, Cheedipudi S, Cordero J, Jiang X, Zhang Q, et al. Pioneering function of Isl1 in the epigenetic control of cardiomyocyte cell fate. Cell Res. 2019;29(6):486–501. https://doi.org/10.1038/s41422-019-0168-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cai CL, Liang X, Shi Y, Chu PH, Pfaff SL, Chen J, et al. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev Cell. 2003;5(6):877–89. https://doi.org/10.1016/s1534-5807(03)00363-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Pandur P, Sirbu IO, Kühl SJ, Philipp M, Kühl M. Islet1-expressing cardiac progenitor cells: a comparison across species. Dev Genes Evol. 2013;223(1–2):117–29. https://doi.org/10.1007/s00427-012-0400-1.

    Article  PubMed  Google Scholar 

  28. Hatzistergos KE, Durante MA, Valasaki K, Wanschel ACBA, Harbour JW, Hare JM. A novel cardiomyogenic role for Isl1+ neural crest cells in the inflow tract. Sci Adv. 2020;6(49):eaba9950. https://doi.org/10.1126/sciadv.aba9950.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Quaranta R, Fell J, Rühle F, Rao J, Piccini I, Araúzo-Bravo MJ, et al. Revised roles of ISL1 in a hES cell-based model of human heart chamber specification. Elife. 2018;7:e31706. https://doi.org/10.7554/eLife.31706.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Golzio C, Havis E, Daubas P, Nuel G, Babarit C, Munnich A, et al. ISL1 directly regulates FGF10 transcription during human cardiac outflow formation. PLoS One. 2012;7(1):e30677. https://doi.org/10.1371/journal.pone.0030677.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Khatami M, Heidari MM, Kazeminasab F, Zare BR. Identification of a novel non-sense mutation in TBX5 gene in pediatric patients with congenital heart defects. J Cardiovasc Thorac Res. 2018;10(1):41–5. https://doi.org/10.15171/jcvtr.2018.07.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Genead R, Danielsson C, Andersson AB, Corbascio M, Franco-Cereceda A, Sylvén C, et al. Islet-1 cells are cardiac progenitors present during the entire lifespan: from the embryonic stage to adulthood. Stem Cells Dev. 2010;19(10):1601–15. https://doi.org/10.1089/scd.2009.0483.

    Article  CAS  PubMed  Google Scholar 

  33. Barreto S, Hamel L, Schiatti T, Yang Y, George V. Cardiac progenitor cells from stem cells: learning from genetics and biomaterials. Cells. 2019;8(12):1536. https://doi.org/10.3390/cells8121536.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Laugwitz KL, Moretti A, Lam J, Gruber P, Chen Y, Woodard S, et al. Postnatal isl1+ cardioblasts enter fully differentiated cardiomyocyte lineages. Nature. 2005;433(7026):647–53. https://doi.org/10.1038/nature03215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Bartulos O, Zhuang ZW, Huang Y, Mikush N, Suh C, Bregasi A, et al. ISL1 cardiovascular progenitor cells for cardiac repair after myocardial infarction. JCI Insight. 2016;1(10):e80920. https://doi.org/10.1172/jci.insight.80920.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Zhou B, von Gise A, Ma Q, Rivera-Feliciano J, Pu WT. Nkx2-5- and Isl1-expressing cardiac progenitors contribute to proepicardium. Biochem Biophys Res Commun. 2008;375(3):450–3. https://doi.org/10.1016/j.bbrc.2008.08.044.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Dodou E, Verzi MP, Anderson JP, Xu SM, Black BL. Mef2c is a direct transcriptional target of ISL1 and GATA factors in the anterior heart field during mouse embryonic development. Development. 2004;131(16):3931–42. https://doi.org/10.1242/dev.01256.

    Article  CAS  PubMed  Google Scholar 

  38. Ma Q, Zhou B, Pu WT. Reassessment of Isl1 and Nkx2-5 cardiac fate maps using a Gata4-based reporter of Cre activity. Dev Biol. 2008;323(1):98–104. https://doi.org/10.1016/j.ydbio.2008.08.013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kelly RG, Buckingham ME, Moorman AF. Heart fields and cardiac morphogenesis. Cold Spring Harb Perspect Med. 2014;4(10):a015750. https://doi.org/10.1101/cshperspect.a015750.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Scalise M, Torella M, Marino F, Ravo M, Giurato G, Vicinanza C, et al. Atrial myxomas arise from multipotent cardiac stem cells. Eur Heart J. 2020;41(45):4332–45. https://doi.org/10.1093/eurheartj/ehaa156.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Dorn T, Goedel A, Lam JT, Haas J, Tian Q, Herrmann F, et al. Direct nkx2-5 transcriptional repression of isl1 controls cardiomyocyte subtype identity. Stem Cells. 2015;33(4):1113–29. https://doi.org/10.1002/stem.1923.

    Article  CAS  PubMed  Google Scholar 

  42. Colombo S, de Sena-Tomás C, George V, Werdich AA, Kapur S, MacRae CA, et al. Nkx genes establish second heart field cardiomyocyte progenitors at the arterial pole and pattern the venous pole through Isl1 repression. Development. 2018;145(3):dev161497. https://doi.org/10.1242/dev.161497.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gay F, Anglade I, Gong Z, Salbert G. The LIM/homeodomain protein islet-1 modulates estrogen receptor functions. Mol Endocrinol. 2000;14(10):1627–48. https://doi.org/10.1210/mend.14.10.0538.

    Article  CAS  PubMed  Google Scholar 

  44. Davis AM, Seney ML, Walker HJ, Tobet SA. Differential colocalization of Islet-1 and estrogen receptor alpha in the murine preoptic area and hypothalamus during development. Endocrinology. 2004;145(1):360–6. https://doi.org/10.1210/en.2003-0996.

    Article  CAS  PubMed  Google Scholar 

  45. Miranda-Carboni GA, Guemes M, Bailey S, Anaya E, Corselli M, Peault B, et al. GATA4 regulates estrogen receptor-alpha-mediated osteoblast transcription. Mol Endocrinol. 2011;25(7):1126–36. https://doi.org/10.1210/me.2010-0463.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ye Y, Chen X, Zhang W. Mammalian SWI/SNF chromatin remodeling complexes in embryonic stem cells: regulating the balance between pluripotency and differentiation. Front Cell Dev Biol. 2021;8:626383. https://doi.org/10.3389/fcell.2020.626383.

    Article  PubMed  PubMed Central  Google Scholar 

  47. George RM, Firulli AB. Epigenetics and heart development. Front Cell Dev Biol. 2021;9:637996. https://doi.org/10.3389/fcell.2021.637996.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Clapier CR, Cairns BR. The biology of chromatin remodeling complexes. Annu Rev Biochem. 2009;78:273–304. https://doi.org/10.1146/annurev.biochem.77.062706.153223.

    Article  CAS  PubMed  Google Scholar 

  49. Jordan NV, Prat A, Abell AN, Zawistowski JS, Sciaky N, Karginova OA, et al. SWI/SNF chromatin-remodeling factor Smarcd3/Baf60c controls epithelial-mesenchymal transition by inducing Wnt5a signaling. Mol Cell Biol. 2013;33(15):3011–25. https://doi.org/10.1128/MCB.01443-12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Javaid S, Zhang J, Anderssen E, Black JC, Wittner BS, Tajima K, et al. Dynamic chromatin modification sustains epithelial-mesenchymal transition following inducible expression of Snail-1. Cell Rep. 2013;5(6):1679–89. https://doi.org/10.1016/j.celrep.2013.11.034.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Oh J, Sohn DH, Ko M, Chung H, Jeon SH, Seong RH. BAF60a interacts with p53 to recruit the SWI/SNF complex. J Biol Chem. 2008;283(18):11924–34. https://doi.org/10.1074/jbc.M705401200.

    Article  CAS  PubMed  Google Scholar 

  52. Cai W, Albini S, Wei K, Willems E, Guzzo RM, Tsuda M, et al. Coordinate Nodal and BMP inhibition directs Baf60c-dependent cardiomyocyte commitment. Genes Dev. 2013;27(21):2332–44. https://doi.org/10.1101/gad.225144.113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Chen L, Fulcoli FG, Ferrentino R, Martucciello S, Illingworth EA, Baldini A. Transcriptional control in cardiac progenitors: Tbx1 interacts with the BAF chromatin remodeling complex and regulates Wnt5a. PLoS Genet. 2012;8(3):e1002571. https://doi.org/10.1371/journal.pgen.1002571.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hota SK, Johnson JR, Verschueren E, Thomas R, Blotnick AM, Zhu Y, et al. Dynamic BAF chromatin remodeling complex subunit inclusion promotes temporally distinct gene expression programs in cardiogenesis. Development. 2019;146(19):dev174086. https://doi.org/10.1242/dev.174086.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hatcher CJ, Kim MS, Mah CS, Goldstein MM, Wong B, Mikawa T, et al. TBX5 transcription factor regulates cell proliferation during cardiogenesis. Dev Biol. 2001;230(2):177–88. https://doi.org/10.1006/dbio.2000.0134.

    Article  CAS  PubMed  Google Scholar 

  56. Akazawa H, Komuro I. Cardiac transcription factor Csx/Nkx2-5: its role in cardiac development and diseases. Pharmacol Ther. 2005;107(2):252–68. https://doi.org/10.1016/j.pharmthera.2005.03.005.

    Article  CAS  PubMed  Google Scholar 

  57. Bouveret R, Waardenberg AJ, Schonrock N, Ramialison M, Doan T, de Jong D, et al. NKX2-5 mutations causative for congenital heart disease retain functionality and are directed to hundreds of targets. Elife. 2015;4:e06942. https://doi.org/10.7554/eLife.06942.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Clark CD, Lee KH. Second heart field-specific expression of Nkx2-5 requires promoter proximal interaction with Srf. Mech Dev. 2020;162:103615. https://doi.org/10.1016/j.mod.2020.103615.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Cambier L, Plate M, Sucov HM, Pashmforoush M. Nkx2-5 regulates cardiac growth through modulation of Wnt signaling by R-spondin3. Development. 2014;141(15):2959–71. https://doi.org/10.1242/dev.103416.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Liu Z, Li T, Liu Y, Jia Z, Li Y, Zhang C, et al. WNT signaling promotes Nkx2.5 expression and early cardiomyogenesis via downregulation of Hdac1. Biochim Biophys Acta. 2009;1793(2):300–11. https://doi.org/10.1016/j.bbamcr.2008.08.013.

    Article  CAS  PubMed  Google Scholar 

  61. Li H, Wang J, Huang K, Zhang T, Gao L, Yang S, et al. Front Oncol. 2021;11:648045. https://doi.org/10.3389/fonc.2021.648045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Komuro I, Izumo S. Csx: a murine homeobox-containing gene specifically expressed in the developing heart. Proc Natl Acad Sci U S A. 1993;90(17):8145–9. https://doi.org/10.1073/pnas.90.17.8145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chen L, Fulcoli FG, Tang S, Baldini A. Tbx1 regulates proliferation and differentiation of multipotent heart progenitors. Circ Res. 2009;105(9):842–51. https://doi.org/10.1161/CIRCRESAHA.109.200295.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hou D, Guan Y, Liu J, Xiao Z, Ouyang Y, Yu Y, et al. Cloning and characterization of the NPCEDRG gene promoter. Mol Cell Biochem. 2011;346(1–2):1–10. https://doi.org/10.1007/s11010-010-0584-5.

    Article  CAS  PubMed  Google Scholar 

  65. Behrens AN, Iacovino M, Lohr JL, Ren Y, Zierold C, Harvey RP, et al. Nkx2-5 mediates differential cardiac differentiation through interaction with Hoxa10. Stem Cells Dev. 2013;22(15):2211–20. https://doi.org/10.1089/scd.2012.0611.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Jamali M, Karamboulas C, Rogerson PJ, Skerjanc IS. BMP signaling regulates Nkx2-5 activity during cardiomyogenesis. FEBS Lett. 2001;509(1):126–30. https://doi.org/10.1016/s0014-5793(01)03151-9.

    Article  CAS  PubMed  Google Scholar 

  67. Wang J, Jin Y, Cattini PA. Expression of the cardiac maintenance and survival factor FGF-16 gene is regulated by Csx/Nkx2.5 and is an early target of doxorubicin cardiotoxicity. DNA Cell Biol. 2017;36(2):117–26. https://doi.org/10.1089/dna.2016.3507.

    Article  CAS  PubMed  Google Scholar 

  68. Watanabe Y, Zaffran S, Kuroiwa A, Higuchi H, Ogura T, Harvey RP, et al. Fibroblast growth factor 10 gene regulation in the second heart field by Tbx1, Nkx2-5, and Islet1 reveals a genetic switch for down-regulation in the myocardium. Proc Natl Acad Sci U S A. 2012;109(45):18273–80. https://doi.org/10.1073/pnas.1215360109.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Kodama H, Hirotani T, Suzuki Y, Ogawa S, Yamazaki K. Cardiomyogenic differentiation in cardiac myxoma expressing lineage-specific transcription factors. Am J Pathol. 2002;161(2):381–9. https://doi.org/10.1016/S0002-9440(10)64193-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Serpooshan V, Liu YH, Buikema JW, Galdos FX, Chirikian O, Paige S, et al. Nkx2.5+ Cardiomyoblasts contribute to Cardiomyogenesis in the neonatal heart. Sci Rep. 2017;7(1):12590. https://doi.org/10.1038/s41598-017-12869-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Yannarelli G, Pacienza N, Montanari S, Santa-Cruz D, Viswanathan S, Keating A. OCT4 expression mediates partial cardiomyocyte reprogramming of mesenchymal stromal cells. PLoS One. 2017;12(12):e0189131. https://doi.org/10.1371/journal.pone.0189131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Nagura S, Otaka S, Koike C, Okabe M, Yoshida T, Fathy M, et al. Effect of exogenous Oct4 overexpression on cardiomyocyte differentiation of human amniotic mesenchymal cells. Cell Reprogram. 2013;15(5):471–80. https://doi.org/10.1089/cell.2013.0002.

    Article  CAS  PubMed  Google Scholar 

  73. Sam J, Mercer EJ, Torregroza I, Banks KM, Evans T. Specificity, redundancy and dosage thresholds among gata4/5/6 genes during zebrafish cardiogenesis. Biol Open. 2020;9(6):bio053611. https://doi.org/10.1242/bio.053611.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Laemmle LL, Cohen JB, Glorioso JC. Constitutive expression of GATA4 dramatically increases the cardiogenic potential of D3 mouse embryonic stem cells. Open. Biotechnol J. 2016;10:248–57. https://doi.org/10.2174/1874070701610010248.

    Article  CAS  Google Scholar 

  75. Yamak A, Latinkic BV, Dali R, Temsah R, Nemer M. Cyclin D2 is a GATA4 cofactor in cardiogenesis. Proc Natl Acad Sci U S A. 2014;111(4):1415–20. https://doi.org/10.1073/pnas.1312993111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Liu J, Cheng H, Xiang M, Zhou L, Wu B, Moskowitz IP, et al. Gata4 regulates hedgehog signaling and Gata6 expression for outflow tract development. PLoS Genet. 2019;15(5):e1007711. https://doi.org/10.1371/journal.pgen.1007711.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Lu F, Zhou Q, Liu L, Zeng G, Ci W, Liu W, et al. A tumor suppressor enhancing module orchestrated by GATA4 denotes a therapeutic opportunity for GATA4 deficient HCC patients. Theranostics. 2020;10(2):484–97. https://doi.org/10.7150/thno.38060.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Agnihotri S, Wolf A, Munoz DM, Smith CJ, Gajadhar A, Restrepo A, et al. A GATA4-regulated tumor suppressor network represses formation of malignant human astrocytomas. J Exp Med. 2011;208(4):689–702. https://doi.org/10.1084/jem.20102099.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Hellebrekers DM, Lentjes MH, van den Bosch SM, Melotte V, Wouters KA, Daenen KL, et al. GATA4 and GATA5 are potential tumor suppressors and biomarkers in colorectal cancer. Clin Cancer Res. 2009;15(12):3990–7. https://doi.org/10.1158/1078-0432.CCR-09-0055.

    Article  CAS  PubMed  Google Scholar 

  80. Ma S, Xu Q, Shi R, Zhang X, Chen X. The omitted symptoms challenge the diagnosis of right atrial myxoma: a case report. BMC Cardiovasc Disord. 2020;20(1):149. https://doi.org/10.1186/s12872-020-01413-4.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Amano J, Kono T, Wada Y, Zhang T, Koide N, Fujimori M, et al. Cardiac myxoma: its origin and tumor characteristics. Ann Thorac Cardiovasc Surg. 2003;9(4):215–21.

    PubMed  Google Scholar 

  82. Zhao T, Bai R, Wu F, Lu WJ, Zhang J. Generation of a TBX5 homozygous knockout embryonic stem cell line (WAe009-A-45) by CRISPR/Cas9 genome editing. Stem Cell Res. 2021;51:102156. https://doi.org/10.1016/j.scr.2021.102156.

    Article  CAS  PubMed  Google Scholar 

  83. Lahm H, Heinrich P, Zierler E, Dzilic E, Neb I, Luzius T, et al. Generation of a CRISPR/Cas edited human induced pluripotent stem cell line DHMi005-A-1 carrying a patient-specific disease-causing point mutation in the TBX5 gene. Stem Cell Res. 2022;60:102691. https://doi.org/10.1016/j.scr.2022.102691.

    Article  CAS  PubMed  Google Scholar 

  84. Anderson RH, Webb S, Brown NA, Lamers W, Moorman A. Development of the heart: (2) Septation of the atriums and ventricles. Heart. 2003;89(8):949–58. https://doi.org/10.1136/heart.89.8.949.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Boogerd CJ, Dooijes D, Ilgun A, Mathijssen IB, Hordijk R, van de Laar IM, et al. Functional analysis of novel TBX5 T-box mutations associated with Holt-Oram syndrome. Cardiovasc Res. 2010;88(1):130–9. https://doi.org/10.1093/cvr/cvq178.

    Article  CAS  PubMed  Google Scholar 

  86. Goetz SC, Brown DD, Conlon FL. TBX5 is required for embryonic cardiac cell cycle progression. Development. 2006;133(13):2575–84. https://doi.org/10.1242/dev.02420.

    Article  CAS  PubMed  Google Scholar 

  87. Ghosh TK, Song FF, Packham EA, Buxton S, Robinson TE, Ronksley J, et al. Physical interaction between TBX5 and MEF2C is required for early heart development. Mol Cell Biol. 2009;29(8):2205–18. https://doi.org/10.1128/MCB.01923-08.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Ma R, Yang Y, Tu Q, Hu K. Overexpression of T-box transcription factor 5 (TBX5) inhibits proliferation and invasion in non-small cell lung carcinoma cells. Oncol Res. 2017;25(9):1495–504. https://doi.org/10.3727/096504017X14883287513729.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Misra C, Chang SW, Basu M, Huang N, Garg V. Disruption of myocardial Gata4 and Tbx5 results in defects in cardiomyocyte proliferation and atrioventricular septation. Hum Mol Genet. 2014;23(19):5025–35. https://doi.org/10.1093/hmg/ddu215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Mori AD, Bruneau BG. TBX5 mutations and congenital heart disease: Holt-Oram syndrome revealed. Curr Opin Cardiol. 2004;19(3):211–5. https://doi.org/10.1097/00001573-200405000-00004.

    Article  PubMed  Google Scholar 

  91. Fan C, Duhagon MA, Oberti C, Chen S, Hiroi Y, Komuro I, et al. Novel TBX5 mutations and molecular mechanism for Holt-Oram syndrome. J Med Genet. 2003;40(3):e29. https://doi.org/10.1136/jmg.40.3.e29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Fijnvandraat AC, Lekanne Deprez RH, Christoffels VM, Ruijter JM, Moorman AF. TBX5 overexpression stimulates differentiation of chamber myocardium in P19C16 embryonic carcinoma cells. J Muscle Res Cell Motil. 2003;24(2–3):211–8. https://doi.org/10.1023/a:1026063409656.

    Article  CAS  PubMed  Google Scholar 

  93. Snyder M, Huang XY, Zhang JJ. Stat3 directly controls the expression of Tbx5, Nkx2.5, and GATA4 and is essential for cardiomyocyte differentiation of P19CL6 cells. J Biol Chem. 2010;285(31):23639–46. https://doi.org/10.1074/jbc.M110.101063.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Jia Y, Chang Y, Guo Z, Li H. Transcription factor Tbx5 promotes cardiomyogenic differentiation of cardiac fibroblasts treated with 5-azacytidine. J Cell Biochem. 2019;120(10):16503–15. https://doi.org/10.1002/jcb.28885.

    Article  CAS  PubMed  Google Scholar 

  95. Li Q, Guo ZK, Chang YQ, Yu X, Li CX, Li H. Gata4, Tbx5 and Baf60c induce differentiation of adipose tissue-derived mesenchymal stem cells into beating cardiomyocytes. Int J Biochem Cell Biol. 2015;66:30–6. https://doi.org/10.1016/j.biocel.2015.06.008.

    Article  CAS  PubMed  Google Scholar 

  96. Stirnimann CU, Ptchelkine D, Grimm C, Müller CW. Structural basis of TBX5-DNA recognition: the T-box domain in its DNA-bound and -unbound form. J Mol Biol. 2010;400(1):71–81. https://doi.org/10.1016/j.jmb.2010.04.052.

    Article  CAS  PubMed  Google Scholar 

  97. Peng C, Zhang WH, Pan B, Gao WQ, Tian J. Temporal regulation of transcription factor Mef2c by histone acetylases during cardiogenesis. Zhongguo Dang Dai Er Ke Za Zhi. 2014;16(4):418–23.

    CAS  PubMed  Google Scholar 

  98. Materna SC, Sinha T, Barnes RM, Lammerts van Bueren K, Black BL. Cardiovascular development and survival require Mef2c function in the myocardial but not the endothelial lineage. Dev Biol. 2019;445(2):170–7. https://doi.org/10.1016/j.ydbio.2018.12.002.

    Article  CAS  PubMed  Google Scholar 

  99. Vincentz JW, Barnes RM, Firulli BA, Conway SJ, Firulli AB. Cooperative interaction of Nkx2.5 and Mef2c transcription factors during heart development. Dev Dyn. 2008;237(12):3809–19. https://doi.org/10.1002/dvdy.21803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Torgersen JS, Takle H, Andersen Ø. Differential spatial expression of mef2 paralogs during cardiac development in Atlantic cod (Gadus morhua). Comp Biochem Physiol B Biochem Mol Biol. 2011;158(2):181–7. https://doi.org/10.1016/j.cbpb.2010.11.006.

    Article  CAS  PubMed  Google Scholar 

  101. Lei X, Zhao J, Sagendorf JM, Rajashekar N, Xu J, Dantas Machado AC, et al. Crystal structures of ternary complexes of MEF2 and NKX2-5 bound to DNA reveal a disease related protein-protein interaction Interface. J Mol Biol. 2020;432(19):5499–508. https://doi.org/10.1016/j.jmb.2020.07.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Nenna A, Loreni F, Giacinto O, Chello C, Nappi P, Chello M, et al. miRNAs in cardiac Myxoma: new pathologic findings for potential therapeutic opportunities. Int J Mol Sci. 2022;23(6):3309. https://doi.org/10.3390/ijms23063309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Izarra A, Moscoso I, Cañón S, Carreiro C, Fondevila D, Martín-Caballero J, et al. miRNA-1 and miRNA-133a are involved in early commitment of pluripotent stem cells and demonstrate antagonistic roles in the regulation of cardiac differentiation. J Tissue Eng Regen Med. 2017;11(3):787–99. https://doi.org/10.1002/term.1977.

    Article  CAS  PubMed  Google Scholar 

  104. Kula-Alwar D, Marber MS, Hughes SM, Hinits Y. Mef2c factors are required for early but not late addition of cardiomyocytes to the ventricle. Dev Biol. 2021;470:95–107. https://doi.org/10.1016/j.ydbio.2020.11.008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Pane LS, Zhang Z, Ferrentino R, Huynh T, Cutillo L, Baldini A. Tbx1 is a negative modulator of Mef2c. Hum Mol Genet. 2012;21(11):2485–96. https://doi.org/10.1093/hmg/dds063.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Okubo C, Narita M, Inagaki A, Nishikawa M, Hotta A, Yamanaka S, et al. Expression dynamics of HAND1/2 in in vitro human cardiomyocyte differentiation. Stem Cell Rep. 2021;16(8):1906–22. https://doi.org/10.1016/j.stemcr.2021.06.014.

    Article  CAS  Google Scholar 

  107. Vincentz JW, Barnes RM, Firulli AB. Hand factors as regulators of cardiac morphogenesis and implications for congenital heart defects. Birth Defects Res A Clin Mol Teratol. 2011;91(6):485–94. https://doi.org/10.1002/bdra.20796.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Witman N, Zhou C, Grote Beverborg N, Sahara M, Chien KR. Cardiac progenitors and paracrine mediators in cardiogenesis and heart regeneration. Semin Cell Dev Biol. 2020;100:29–51. https://doi.org/10.1016/j.semcdb.2019.10.011.

    Article  PubMed  Google Scholar 

  109. Koninckx R, Daniëls A, Windmolders S, Carlotti F, Mees U, Steels P, et al. Mesenchymal stem cells or cardiac progenitors for cardiac repair? A comparative study. Cell Mol Life Sci. 2011;68(12):2141–56. https://doi.org/10.1007/s00018-010-0560-y.

    Article  CAS  PubMed  Google Scholar 

  110. Orlandi A, Ciucci A, Ferlosio A, Genta R, Spagnoli LG, Gabbiani G. Cardiac myxoma cells exhibit embryonic endocardial stem cell features. J Pathol. 2006;209(2):231–9. https://doi.org/10.1002/path.1959.

    Article  CAS  PubMed  Google Scholar 

  111. Taubenschmid J, Weitzer G. Mechanisms of cardiogenesis in cardiovascular progenitor cells. Int Rev Cell Mol Biol. 2012;293:195–267. https://doi.org/10.1016/B978-0-12-394304-0.00012-9.

    Article  CAS  PubMed  Google Scholar 

  112. Sun Q, Taurin S, Sethakorn N, Long X, Imamura M, Wang DZ, et al. Myocardin-dependent activation of the CArG box-rich smooth muscle gamma-actin gene: preferential utilization of a single CArG element through functional association with the NKX3.1 homeodomain protein. J Biol Chem. 2009;284(47):32582–90. https://doi.org/10.1074/jbc.M109.033910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Wang N, Xu Y, Qin T, Wang FP, Ma LL, Luo XG, et al. Myocardin-related transcription factor-a is a key regulator in retinoic acid-induced neural-like differentiation of adult bone marrow-derived mesenchymal stem cells. Gene. 2013;523(2):178–86. https://doi.org/10.1016/j.gene.2013.03.043.

    Article  CAS  PubMed  Google Scholar 

  114. Zhang L, Wang H, Liu C, Wu Q, Su P, Wu D, et al. MSX2 initiates and accelerates mesenchymal stem/stromal cell specification of hPSCs by regulating TWIST1 and PRAME. Stem Cell Rep. 2018;11(2):497–513. https://doi.org/10.1016/j.stemcr.2018.06.019.

    Article  CAS  Google Scholar 

  115. Chen YH, Ishii M, Sucov HM, Maxson RE Jr. Msx1 and Msx2 are required for endothelial-mesenchymal transformation of the atrioventricular cushions and patterning of the atrioventricular myocardium. BMC Dev Biol. 2008;8:75. https://doi.org/10.1186/1471-213X-8-75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Wu Q, Zhang L, Su P, Lei X, Liu X, Wang H, et al. MSX2 mediates entry of human pluripotent stem cells into mesendoderm by simultaneously suppressing SOX2 and activating NODAL signaling. Cell Res. 2015;25(12):1314–32. https://doi.org/10.1038/cr.2015.118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Jain R, Li D, Gupta M, Manderfield LJ, Ifkovits JL, Wang Q, et al. HEART DEVELOPMENT. Integration of bmp and Wnt signaling by Hopx specifies commitment of cardiomyoblasts. Science. 2015;348(6242):aaa6071. https://doi.org/10.1126/science.aaa6071.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Chen F, Kook H, Milewski R, Gitler AD, Lu MM, Li J, et al. Hop is an unusual homeobox gene that modulates cardiac development. Cell. 2002;110(6):713–23. https://doi.org/10.1016/s0092-8674(02)00932-7.

    Article  CAS  PubMed  Google Scholar 

  119. Schneider MD, Baker AH, Riley P. Hopx and the Cardiomyocyte parentage. Mol Ther. 2015;23(9):1420–2. https://doi.org/10.1038/mt.2015.140.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Li D, Sun J, Zhong TP. Wnt signaling in heart development and regeneration. Curr Cardiol Rep. 2022; https://doi.org/10.1007/s11886-022-01756-8.

  121. Pahnke A, Conant G, Huyer LD, Zhao Y, Feric N, Radisic M. The role of Wnt regulation in heart development, cardiac repair and disease: a tissue engineering perspective. Biochem Biophys Res Commun. 2016;473(3):698–703.

    Article  CAS  PubMed  Google Scholar 

  122. Tian Y, Cohen ED, Morrisey EE. The importance of Wnt signaling in cardiovascular development. Pediatr Cardiol. 2010;31(3):342–8. https://doi.org/10.1007/s00246-009-9606-z.

    Article  PubMed  Google Scholar 

  123. Flaherty MP, Kamerzell TJ, Dawn B. Wnt signaling and cardiac differentiation. Prog Mol Biol Transl Sci. 2012;111:153–74. https://doi.org/10.1016/B978-0-12-398459-3.00007-1.

    Article  CAS  PubMed  Google Scholar 

  124. Cohen ED, Miller MF, Wang Z, Moon RT, Morrisey EE. Wnt5a and Wnt11 are essential for second heart field progenitor development. Development. 2012;139(11):1931–40. https://doi.org/10.1242/dev.069377.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Du Q, Geller DA. Cross-regulation between Wnt and NF-κB signaling pathways. For Immunopathol Dis Therap. 2010;1(3):155–81. https://doi.org/10.1615/ForumImmunDisTher.v1.i3.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Park EJ, Watanabe Y, Smyth G, Miyagawa-Tomita S, Meyers E, Klingensmith J, et al. An FGF autocrine loop initiated in second heart field mesoderm regulates morphogenesis at the arterial pole of the heart. Development. 2008;135(21):3599–610. https://doi.org/10.1242/dev.025437.

    Article  CAS  PubMed  Google Scholar 

  127. Zhang J, Tao R, Campbell KF, Carvalho JL, Ruiz EC, Kim GC, et al. Functional cardiac fibroblasts derived from human pluripotent stem cells via second heart field progenitors. Nat Commun. 2019;10(1):2238. https://doi.org/10.1038/s41467-019-09831-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Khosravi F, Ahmadvand N, Bellusci S, Sauer H. The multifunctional contribution of FGF signaling to cardiac development, homeostasis, disease and repair. Front Cell Dev Biol. 2021;9:672935. https://doi.org/10.3389/fcell.2021.672935.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Bosada FM, Devasthali V, Jones KA, Stankunas K. Wnt/β-catenin signaling enables developmental transitions during valvulogenesis. Development. 2016;143(6):1041–54. https://doi.org/10.1242/dev.130575.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Rosenblatt-Velin N, Lepore MG, Cartoni C, Beermann F, Pedrazzini T. FGF-2 controls the differentiation of resident cardiac precursors into functional cardiomyocytes. J Clin Invest. 2005;115(7):1724–33. https://doi.org/10.1172/JCI23418.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Tahara N, Akiyama R, Wang J, Kawakami H, Bessho Y, Kawakami Y. The FGF-AKT pathway is necessary for cardiomyocyte survival for heart regeneration in zebrafish. Dev Biol. 2021;472:30–7. https://doi.org/10.1016/j.ydbio.2020.12.019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Kiyonari H, Kaneko M, Abe S, Aizawa S. Three inhibitors of FGF receptor, ERK, and GSK3 establishes germline-competent embryonic stem cells of C57BL/6N mouse strain with high efficiency and stability. Genesis. 2010;48(5):317–27. https://doi.org/10.1002/dvg.20614.

    Article  CAS  PubMed  Google Scholar 

  133. Fujisawa H, Koide N, Kono T, Takayama K, Tsukioka K, Wada Y, et al. Expression of basic fibroblast growth factor and its receptor-1 in cardiac myxoma. J Cardiovasc Surg. 2002;43(5):589–94.

    CAS  Google Scholar 

  134. Liao S, Bodmer JR, Azhar M, Newman G, Coffin JD, Doetschman T, et al. The influence of FGF2 high molecular weight (HMW) isoforms in the development of cardiac ischemia-reperfusion injury. J Mol Cell Cardiol. 2010;48(6):1245–54. https://doi.org/10.1016/j.yjmcc.2010.01.014.

    Article  CAS  PubMed  Google Scholar 

  135. Marques SR, Yelon D. Differential requirement for BMP signaling in atrial and ventricular lineages establishes cardiac chamber proportionality. Dev Biol. 2009;328(2):472–82. https://doi.org/10.1016/j.ydbio.2009.02.010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. van Wijk B, Moorman AF, van den Hoff MJ. Role of bone morphogenetic proteins in cardiac differentiation. Cardiovasc Res. 2007;74(2):244–55. https://doi.org/10.1016/j.cardiores.2006.11.022.

    Article  CAS  PubMed  Google Scholar 

  137. Bin Z, Sheng LG, Gang ZC, Hong J, Jun C, Bo Y, et al. Efficient cardiomyocyte differentiation of embryonic stem cells by bone morphogenetic protein-2 combined with visceral endoderm-like cells. Cell Biol Int. 2006;30(10):769–76. https://doi.org/10.1016/j.cellbi.2006.05.011.

    Article  CAS  PubMed  Google Scholar 

  138. Pashmforoush M, Lu JT, Chen H, Amand TS, Kondo R, Pradervand S, et al. Nkx2-5 pathways and congenital heart disease; loss of ventricular myocyte lineage specification leads to progressive cardiomyopathy and complete heart block. Cell. 2004;117(3):373–86. https://doi.org/10.1016/s0092-8674(04)00405-2.

    Article  CAS  PubMed  Google Scholar 

  139. Lien CL, McAnally J, Richardson JA, Olson EN. Cardiac-specific activity of an Nkx2-5 enhancer requires an evolutionarily conserved Smad binding site. Dev Biol. 2002;244(2):257–66. https://doi.org/10.1006/dbio.2002.0603.

    Article  CAS  PubMed  Google Scholar 

  140. Gude N, Joyo E, Toko H, Quijada P, Villanueva M, Hariharan N, et al. Notch activation enhances lineage commitment and protective signaling in cardiac progenitor cells. Basic Res Cardiol. 2015;110(3):29. https://doi.org/10.1007/s00395-015-0488-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Gude N, Sussman M. Notch signaling and cardiac repair. J Mol Cell Cardiol. 2012;52(6):1226–32. https://doi.org/10.1016/j.yjmcc.2012.03.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Bray SJ. Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol. 2006;7(9):678–89. https://doi.org/10.1038/nrm2009.

    Article  CAS  PubMed  Google Scholar 

  143. Miao L, Lu Y, Nusrat A, Abdelnasser HY, Datta S, Zhou B, et al. The spatiotemporal expression of Notch1 and numb and their functional interaction during cardiac morphogenesis. Cells. 2021;10(9):2192. https://doi.org/10.3390/cells10092192.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM, Díez J, et al. Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 2004;18(1):99–115. https://doi.org/10.1101/gad.276304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Papoutsi T, Luna-Zurita L, Prados B, Zaffran S, de la Pompa JL. Bmp2 and notch cooperate to pattern the embryonic endocardium. Development. 2018;145(13):dev163378. https://doi.org/10.1242/dev.163378.

    Article  CAS  PubMed  Google Scholar 

  146. de la Pompa JL, Epstein JA. Coordinating tissue interactions: notch signaling in cardiac development and disease. Dev Cell. 2012;22(2):244–54. https://doi.org/10.1016/j.devcel.2012.01.014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Pitsava G, Zhu C, Sundaram R, Mills JL, Stratakis CA. Predicting the risk of cardiac myxoma in Carney complex. Genet Med. 2021;23(1):80–5. https://doi.org/10.1038/s41436-020-00956-3.

    Article  CAS  PubMed  Google Scholar 

  148. Stratakis CA, Kirschner LS, Carney JA. Clinical and molecular features of the Carney complex: diagnostic criteria and recommendations for patient evaluation. J Clin Endocrinol Metab. 2001;86(9):4041–6. https://doi.org/10.1210/jcem.86.9.7903.

    Article  CAS  PubMed  Google Scholar 

  149. Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, et al. Mutations of the gene encoding the protein kinase a type I-alpha regulatory subunit in patients with the Carney complex. Nat Genet. 2000;26(1):89–92. https://doi.org/10.1038/79238.

    Article  CAS  PubMed  Google Scholar 

  150. Bertherat J, Horvath A, Groussin L, Grabar S, Boikos S, Cazabat L, et al. Mutations in regulatory subunit type 1A of cyclic adenosine 5′-monophosphate-dependent protein kinase (PRKAR1A): phenotype analysis in 353 patients and 80 different genotypes. J Clin Endocrinol Metab. 2009;94(6):2085–91. https://doi.org/10.1210/jc.2008-2333.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Briassoulis G, Kuburovic V, Xekouki P, Patronas N, Keil MF, Lyssikatos C, et al. Recurrent left atrial myxomas in Carney complex: a genetic cause of multiple strokes that can be prevented. J Stroke Cerebrovasc Dis. 2012;21(8):914.e1–8. https://doi.org/10.1016/j.jstrokecerebrovasdis.2012.01.006.

    Article  PubMed  Google Scholar 

  152. Hashimoto H, Wang Z, Garry GA, Malladi VS, Botten GA, Ye W, et al. Cardiac reprogramming factors synergistically activate genome-wide cardiogenic stage-specific enhancers. Cell Stem Cell. 2019;25(1):69–86.e5. https://doi.org/10.1016/j.stem.2019.03.022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Lozano-Velasco E, Garcia-Padilla C, Del Mar Muñoz-Gallardo M, Martinez-Amaro FJ, Caño-Carrillo S, Castillo-Casas JM, et al. Post-transcriptional regulation of molecular determinants during Cardiogenesis. Int J Mol Sci. 2022;23(5):2839. https://doi.org/10.3390/ijms23052839.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Yu MS, Spiering S, Colas AR. Generation of first heart field-like cardiac progenitors and ventricular-like Cardiomyocytes from human pluripotent stem cells. J Vis Exp. 2018;136:57688. https://doi.org/10.3791/57688.

    Article  CAS  Google Scholar 

  155. Zhang L, Nomura-Kitabayashi A, Sultana N, Cai W, Cai X, Moon AM, et al. Mesodermal Nkx2.5 is necessary and sufficient for early second heart field development. Dev Biol. 2014;390(1):68–79. https://doi.org/10.1016/j.ydbio.2014.02.023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. George V, Colombo S, Targoff KL. An early requirement for nkx2.5 ensures the first and second heart field ventricular identity and cardiac function into adulthood. Dev Biol. 2015;400(1):10–22. https://doi.org/10.1016/j.ydbio.2014.12.019.

    Article  CAS  PubMed  Google Scholar 

  157. Shouman S, Zaher A, Abdelhameed A, Elshaboury S, Sakr S, Fouda BE, et al. Cardiac Progenitor Cells. Adv Exp Med Biol. 2021;1312:51–73. https://doi.org/10.1007/5584_2020_594.

    Article  CAS  PubMed  Google Scholar 

  158. Arbatlı S, Aslan GS, Kocabaş F. Stem cells in regenerative cardiology. Adv Exp Med Biol. 2018;1079:37–53. https://doi.org/10.1007/5584_2017_113.

    Article  CAS  PubMed  Google Scholar 

  159. Piven OO, Winata CL. The canonical way to make a heart: β-catenin and plakoglobin in heart development and remodeling. Exp Biol Med. 2017;242(18):1735–45. https://doi.org/10.1177/1535370217732737.

    Article  CAS  Google Scholar 

  160. Zhang Y, Wang L, Gao P, Sun Z, Li N, Lu Y, et al. ISL1 promotes cancer progression and inhibits cisplatin sensitivity in triple-negative breast cancer cells. Int J Mol Med. 2018;42(5):2343–52. https://doi.org/10.3892/ijmm.2018.3842.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Alshalalfa M, Abou-Ouf H, Davicioni E, Karnes RJ, Alhajj R, Bismar TA. Expression of ISL1 and its partners in prostate cancer progression and neuroendocrine differentiation. J Cancer Res Clin Oncol. 2021;147(8):2223–31. https://doi.org/10.1007/s00432-021-03634-2.

    Article  CAS  PubMed  Google Scholar 

  162. Li L, Sun F, Chen X, Zhang M. ISL1 is upregulated in breast cancer and promotes cell proliferation, invasion, and angiogenesis. OncoTargets Ther. 2018;11:781–9. https://doi.org/10.2147/OTT.S144241.

    Article  Google Scholar 

  163. Li M, Sun C, Bu X, Que Y, Zhang L, Zhang Y, et al. ISL1 promoted tumorigenesis and EMT via Aurora kinase A-induced activation of PI3K/AKT signaling pathway in neuroblastoma. Cell Death Dis. 2021;12(6):620. https://doi.org/10.1038/s41419-021-03894-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Shi Q, Wang W, Jia Z, Chen P, Ma K, Zhou C. ISL1, a novel regulator of CCNB1, CCNB2 and c-MYC genes, promotes gastric cancer cell proliferation and tumor growth. Oncotarget. 2016;7(24):36489–500. https://doi.org/10.18632/oncotarget.9269.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Alfert A, Moreno N, Kerl K. The BAF complex in development and disease. Epigenetics Chromatin. 2019;12(1):19. https://doi.org/10.1186/s13072-019-0264-y.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Mittal P, Roberts CWM. The SWI/SNF complex in cancer - biology, biomarkers and therapy. Nat Rev Clin Oncol. 2020;17(7):435–48. https://doi.org/10.1038/s41571-020-0357-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Nakashima Y, Yanez DA, Touma M, Nakano H, Jaroszewicz A, Jordan MC, et al. Nkx2-5 suppresses the proliferation of atrial myocytes and conduction system. Circ Res. 2014;114(7):1103–13. https://doi.org/10.1161/CIRCRESAHA.114.303219.

    Article  CAS  PubMed  Google Scholar 

  168. Nagel S, Kaufmann M, Drexler HG, MacLeod RA. The cardiac homeobox gene NKX2-5 is deregulated by juxtaposition with BCL11B in pediatric T-ALL cell lines via a novel t(5;14)(q35.1;q32.2). Cancer Res. 2003;63(17):5329–34.

    CAS  PubMed  Google Scholar 

  169. Penha RCC, Buexm LA, Rodrigues FR, de Castro TP, Santos MCS, Fortunato RS, et al. NKX2.5 is expressed in papillary thyroid carcinomas and regulates differentiation in thyroid cells. BMC Cancer. 2018;18(1):498. https://doi.org/10.1186/s12885-018-4399-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Kojic S, Nestorovic A, Rakicevic L, Protic O, Jasnic-Savovic J, Faulkner G, et al. Cardiac transcription factor Nkx2.5 interacts with p53 and modulates its activity. Arch Biochem Biophys. 2015;569:45–53. https://doi.org/10.1016/j.abb.2015.02.001.

    Article  CAS  PubMed  Google Scholar 

  171. Chung W, Kwabi-Addo B, Ittmann M, Jelinek J, Shen L, Yu Y, et al. Identification of novel tumor markers in prostate, colon and breast cancer by unbiased methylation profiling. PLoS One. 2008;3(4):e2079. https://doi.org/10.1371/journal.pone.0002079.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Moussa H, Sidhom I. NKX2-5, SIL/TAL and TLX3/HOX11L2 expression in Egyptian pediatric T-cell acute lymphoblastic leukemia. Asia Pac J Clin Oncol. 2016;12(1):e1–10. https://doi.org/10.1111/ajco.12119.

    Article  PubMed  Google Scholar 

  173. Przybylski GK, Dik WA, Grabarczyk P, Wanzeck J, Chudobska P, Jankowski K, et al. The effect of a novel recombination between the homeobox gene NKX2-5 and the TRD locus in T-cell acute lymphoblastic leukemia on activation of the NKX2-5 gene. Haematologica. 2006;91(3):317–21.

    CAS  PubMed  Google Scholar 

  174. Iacobucci I, Mullighan CG. Genetic basis of acute lymphoblastic leukemia. J Clin Oncol. 2017;35(9):975–83. https://doi.org/10.1200/JCO.2016.70.7836.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Välimäki MJ, Ruskoaho HJ. Targeting GATA4 for cardiac repair. IUBMB Life. 2020;72(1):68–79. https://doi.org/10.1002/iub.2150.

    Article  CAS  PubMed  Google Scholar 

  176. Katanasaka Y, Suzuki H, Sunagawa Y, Hasegawa K, Morimoto T. Regulation of cardiac transcription factor GATA4 by post-translational modification in Cardiomyocyte hypertrophy and heart failure. Int Heart J. 2016;57(6):672–5. https://doi.org/10.1536/ihj.16-404.

    Article  CAS  PubMed  Google Scholar 

  177. Dai YS, Cserjesi P, Markham BE, Molkentin JD. The transcription factors GATA4 and dHAND physically interact to synergistically activate cardiac gene expression through a p300-dependent mechanism. J Biol Chem. 2002;277(27):24390–8. https://doi.org/10.1074/jbc.M202490200.

    Article  CAS  PubMed  Google Scholar 

  178. Patel RS, Romero R, Watson EV, Liang AC, Burger M, Westcott PMK, et al. A GATA4-regulated secretory program suppresses tumors through recruitment of cytotoxic CD8 T cells. Nat Commun. 2022;13(1):256. https://doi.org/10.1038/s41467-021-27731-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Soini T, Haveri H, Elo JM, Kauppinen M, Kyrönlahti A, Salo MK, et al. Transcription factor GATA-4 is abundantly expressed in childhood but not in adult liver tumors. J Pediatr Gastroenterol Nutr. 2012;54(1):101–8. https://doi.org/10.1097/MPG.0b013e31822d52cf.

    Article  CAS  PubMed  Google Scholar 

  180. Cai KQ, Caslini C, Capo-chichi CD, Slater C, Smith ER, Wu H, et al. Loss of GATA4 and GATA6 expression specifies ovarian cancer histological subtypes and precedes neoplastic transformation of ovarian surface epithelia. PLoS One. 2009;4(7):e6454. https://doi.org/10.1371/journal.pone.0006454.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Han Q, Xu X, Li J, Wang J, Bai L, Wang A, et al. GATA4 is highly expressed in childhood acute lymphoblastic leukemia, promotes cell proliferation and inhibits apoptosis by activating BCL2 and MDM2. Mol Med Rep. 2017;16(5):6290–8. https://doi.org/10.3892/mmr.2017.7369.

    Article  CAS  PubMed  Google Scholar 

  182. Khalid AB, Pence J, Suthon S, Lin J, Miranda-Carboni GA, Krum SA. GATA4 regulates mesenchymal stem cells via direct transcriptional regulation of the WNT signalosome. Bone. 2021;144:115819. https://doi.org/10.1016/j.bone.2020.115819.

    Article  CAS  PubMed  Google Scholar 

  183. Behiry EG, Al-Azzouny MA, Sabry D, Behairy OG, Salem NE. Association of NKX2-5, GATA4, and TBX5 polymorphisms with congenital heart disease in Egyptian children. Mol Genet Genomic Med. 2019;7(5):e612. https://doi.org/10.1002/mgg3.612.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Dong MJ, Zhou Y, Duan M, Gao QM, Zhao JH. Clinical significance and mechanism of TBX5 gene in colorectal cancer. Zhonghua Zhong Liu Za Zhi. 2020;42(5):383–90. https://doi.org/10.3760/cma.j.cn112152-112152-20190829-00560.

    Article  CAS  PubMed  Google Scholar 

  185. Yu J, Ma X, Cheung KF, Li X, Tian L, Wang S, et al. Epigenetic inactivation of T-box transcription factor 5, a novel tumor suppressor gene, is associated with colon cancer. Oncogene. 2010;29(49):6464–74. https://doi.org/10.1038/onc.2010.370.

    Article  CAS  PubMed  Google Scholar 

  186. Cheng L, Han T, Chen B, Nie K, Peng W. TBX5-AS1, an enhancer RNA, is a potential novel prognostic biomarker for lung adenocarcinoma. BMC Cancer. 2021;21(1):794. https://doi.org/10.1186/s12885-021-08517-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Laszlo GS, Alonzo TA, Gudgeon CJ, Harrington KH, Kentsis A, Gerbing RB, et al. High expression of myocyte enhancer factor 2C (MEF2C) is associated with adverse-risk features and poor outcome in pediatric acute myeloid leukemia: a report from the Children's oncology group. J Hematol Oncol. 2015;8:115. https://doi.org/10.1186/s13045-015-0215-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Canté-Barrett K, Pieters R, Meijerink JP. Myocyte enhancer factor 2C in hematopoiesis and leukemia. Oncogene. 2014;33(4):403–10. https://doi.org/10.1038/onc.2013.56.

    Article  CAS  PubMed  Google Scholar 

  189. Vitali C, Bassani C, Chiodoni C, Fellini E, Guarnotta C, Miotti S, et al. SOCS2 controls proliferation and Stemness of hematopoietic cells under stress conditions and its deregulation Marks unfavorable acute Leukemias. Cancer Res. 2015;75(11):2387–99. https://doi.org/10.1158/0008-5472.CAN-14-3625.

    Article  CAS  PubMed  Google Scholar 

  190. Jingjing Z, Lei M, Jie Z, Sha C, Yapeng H, Weimin Z, et al. A novel MEF2C mutation in lymphoid neoplasm diffuse large B-cell lymphoma promotes tumorigenesis by increasing c-JUN expression. Naunyn Schmiedeberg's Arch Pharmacol. 2020;393(8):1549–58. https://doi.org/10.1007/s00210-019-01764-6.

    Article  CAS  Google Scholar 

  191. Krenács D, Borbényi Z, Bedekovics J, Méhes G, Bagdi E, Krenács L. Pattern of MEF2B expression in lymphoid tissues and in malignant lymphomas. Virchows Arch. 2015;467(3):345–55. https://doi.org/10.1007/s00428-015-1796-6.

    Article  CAS  PubMed  Google Scholar 

  192. Pon JR, Wong J, Saberi S, Alder O, Moksa M, Grace Cheng SW, et al. MEF2B mutations in non-Hodgkin lymphoma dysregulate cell migration by decreasing MEF2B target gene activation. Nat Commun. 2015;6:7953. https://doi.org/10.1038/ncomms8953.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Da CM, Cheng ZY, Gong CY, Nan W, Zhou KS, Zhao GH, et al. Role of HAND2-AS1 in human tumors. Clin Chim Acta. 2020;511:189–97. https://doi.org/10.1016/j.cca.2020.10.020.

    Article  CAS  PubMed  Google Scholar 

  194. Jing GY, Zheng XZ, Ji XX. lncRNA HAND2-AS1 overexpression inhibits cancer cell proliferation in hepatocellular carcinoma by downregulating RUNX2 expression. J Clin Lab Anal. 2021;35(4):e23717. https://doi.org/10.1002/jcla.23717.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Yuan Z, Yu X, Chen W, Chen D, Cai J, Jiang Y, et al. Epigenetic silencing and tumor suppressor gene of HAND2 by targeting ERK signaling in colorectal cancer. Cell Commun Signal. 2022;20(1):111. https://doi.org/10.1186/s12964-022-00878-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Wang Y, Cai X. Long noncoding RNA HAND2-AS1 restrains proliferation and metastasis of breast cancer cells through sponging miR-1275 and promoting SOX7. Cancer Biomark. 2020;27(1):85–94. https://doi.org/10.3233/CBM-190530.

    Article  CAS  PubMed  Google Scholar 

  197. Zhang J, Ho JC, Chan YC, Lian Q, Siu CW, Tse HF. Overexpression of myocardin induces partial transdifferentiation of human-induced pluripotent stem cell-derived mesenchymal stem cells into cardiomyocytes. Physiol Rep. 2014;2(2):e00237. https://doi.org/10.1002/phy2.237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Tong X, Wang S, Lei Z, Li C, Zhang C, Su Z, et al. MYOCD and SMAD3/SMAD4 form a positive feedback loop and drive TGF-β-induced epithelial-mesenchymal transition in non-small cell lung cancer. Oncogene. 2020;39(14):2890–904. https://doi.org/10.1038/s41388-020-1189-4.

    Article  CAS  PubMed  Google Scholar 

  199. Shats I, Milyavsky M, Cholostoy A, Brosh R, Rotter V. Myocardin in tumor suppression and myofibroblast differentiation. Cell Cycle. 2007;6(10):1141–6. https://doi.org/10.4161/cc.6.10.4251.

    Article  CAS  PubMed  Google Scholar 

  200. Taurin S, Sandbo N, Yau DM, Sethakorn N, Kach J, Dulin NO. Phosphorylation of myocardin by extracellular signal-regulated kinase. J Biol Chem. 2009;284(49):33789–94. https://doi.org/10.1074/jbc.M109.048983.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Zhou Q, Chen W, Fan Z, Chen Z, Liang J, Zeng G, et al. Targeting hyperactive TGFBR2 for treating MYOCD deficient lung cancer. Theranostics. 2021;11(13):6592–606. https://doi.org/10.7150/thno.59816.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Medjkane S, Perez-Sanchez C, Gaggioli C, Sahai E, Treisman R. Myocardin-related transcription factors and SRF are required for cytoskeletal dynamics and experimental metastasis. Nat Cell Biol. 2009;11(3):257–68. https://doi.org/10.1038/ncb1833.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Martinez-Fernandez A, Nelson TJ, Ikeda Y, Terzic A. C-MYC independent nuclear reprogramming favors cardiogenic potential of induced pluripotent stem cells. J Cardiovasc Transl Res. 2010;3(1):13–23. https://doi.org/10.1007/s12265-009-9150-5.

    Article  PubMed  PubMed Central  Google Scholar 

  204. Wang H, Wang M, Wang Y, Wen Y, Chen X, Wu D, et al. MSX2 suppression through inhibition of TGFβ signaling enhances hematopoietic differentiation of human embryonic stem cells. Stem Cell Res Ther. 2020;11(1):147. https://doi.org/10.1186/s13287-020-01653-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Satoh K, Hamada S, Kimura K, Kanno A, Hirota M, Umino J, et al. Up-regulation of MSX2 enhances the malignant phenotype and is associated with twist 1 expression in human pancreatic cancer cells. Am J Pathol. 2008;172(4):926–39. https://doi.org/10.2353/ajpath.2008.070346.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Satoh K, Hamada S, Shimosegawa T. MSX2 in pancreatic tumor development and its clinical application for the diagnosis of pancreatic ductal adenocarcinoma. Front Physiol. 2012;3:430. https://doi.org/10.3389/fphys.2012.00430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Cheng SL, Behrmann A, Shao JS, Ramachandran B, Krchma K, Bello Arredondo Y, et al. Targeted reduction of vascular Msx1 and Msx2 mitigates arteriosclerotic calcification and aortic stiffness in LDLR-deficient mice fed diabetogenic diets. Diabetes. 2014;63(12):4326–37. https://doi.org/10.2337/db14-0326.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Liu J, An H, Yuan W, Feng Q, Chen L, Ma J. Prognostic relevance and function of MSX2 in colorectal Cancer. J Diabetes Res. 2017;2017:3827037. https://doi.org/10.1155/2017/3827037.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Wu Y, Jin Y, Yamamoto N, Takeuchi A, Miwa S, Tsuchiya H, et al. MSX2 inhibits the growth and migration of osteosarcoma cells by repressing SOX2. Am J Transl Res. 2021;13(6):5851–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Lallemand Y, Bensoussan V, Cloment CS, Robert B. Msx genes are important apoptosis effectors downstream of the Shh/Gli3 pathway in the limb. Dev Biol. 2009;331(2):189–98. https://doi.org/10.1016/j.ydbio.2009.04.038.

    Article  CAS  PubMed  Google Scholar 

  211. Kikuchi M, Katoh H, Waraya M, Tanaka Y, Ishii S, Tanaka T, et al. Epigenetic silencing of HOPX contributes to cancer aggressiveness in breast cancer. Cancer Lett. 2017;384:70–8. https://doi.org/10.1016/j.canlet.2016.10.017.

    Article  CAS  PubMed  Google Scholar 

  212. Yamashita K, Katoh H, Watanabe M. The homeobox only protein homeobox (HOPX) and colorectal cancer. Int J Mol Sci. 2013;14(12):23231–43. https://doi.org/10.3390/ijms141223231.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Yap LF, Lai SL, Patmanathan SN, Gokulan R, Robinson CM, White JB, et al. HOPX functions as a tumour suppressor in head and neck cancer. Sci Rep. 2016;6:38758. https://doi.org/10.1038/srep38758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Pećina-Slaus N. Wnt signal transduction pathway and apoptosis: a review. Cancer Cell Int. 2010;10:22. https://doi.org/10.1186/1475-2867-10-22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Polakis P. Wnt signaling in cancer. Cold Spring Harb Perspect Biol. 2012;4(5):a008052. https://doi.org/10.1101/cshperspect.a008052.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Paul S, Dey A. Wnt signaling and cancer development: therapeutic implication. Neoplasma. 2008;55(3):165–76.

    CAS  PubMed  Google Scholar 

  217. Cohen ED, Tian Y, Morrisey EE. Wnt signaling: an essential regulator of cardiovascular differentiation, morphogenesis and progenitor self-renewal. Development. 2008;135(5):789–98. https://doi.org/10.1242/dev.016865.

    Article  CAS  PubMed  Google Scholar 

  218. Klaus A, Müller M, Schulz H, Saga Y, Martin JF, Birchmeier W. Wnt/β-catenin and bmp signals control distinct sets of transcription factors in cardiac progenitor cells. Proc Natl Acad Sci U S A. 2012;109(27):10921–6. https://doi.org/10.1073/pnas.1121236109.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Cohen ED, Wang Z, Lepore JJ, Lu MM, Taketo MM, Epstein DJ, et al. Wnt/beta-catenin signaling promotes expansion of Isl-1-positive cardiac progenitor cells through regulation of FGF signaling. J Clin Invest. 2007;117(7):1794–804. https://doi.org/10.1172/JCI31731.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Perry JM, He XC, Sugimura R, Grindley JC, Haug JS, Ding S, et al. Cooperation between both Wnt/{beta}-catenin and PTEN/PI3K/Akt signaling promotes primitive hematopoietic stem cell self-renewal and expansion. Genes Dev. 2011;25(18):1928–42. https://doi.org/10.1101/gad.17421911.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Jiang YG, Luo Y, He DL, Li X, Zhang LL, Peng T, et al. Role of Wnt/beta-catenin signaling pathway in epithelial-mesenchymal transition of human prostate cancer induced by hypoxia-inducible factor-1alpha. Int J Urol. 2007;14(11):1034–9. https://doi.org/10.1111/j.1442-2042.2007.01866.x.

    Article  CAS  PubMed  Google Scholar 

  222. Tirosh-Finkel L, Zeisel A, Brodt-Ivenshitz M, Shamai A, Yao Z, Seger R, et al. BMP-mediated inhibition of FGF signaling promotes cardiomyocyte differentiation of anterior heart field progenitors. Development. 2010;137(18):2989–3000. https://doi.org/10.1242/dev.051649.

    Article  CAS  PubMed  Google Scholar 

  223. Wang J, Sontag D, Cattini PA. Heart-specific expression of FGF-16 and a potential role in postnatal cardioprotection. Cytokine Growth Factor Rev. 2015;26(1):59–66. https://doi.org/10.1016/j.cytogfr.2014.07.007.

    Article  CAS  PubMed  Google Scholar 

  224. Hu Y, Li L, Shen L, Gao H, Yu F, Yin W, et al. FGF-16 protects against adverse cardiac remodeling in the infarct diabetic heart. Am J Transl Res. 2017;9(4):1630–40.

    CAS  PubMed  PubMed Central  Google Scholar 

  225. Xu B, Li F, Zhang W, Su Y, Tang L, Li P, et al. Identification of metabolic pathways underlying FGF1 and CHIR99021-mediated cardioprotection. iScience. 2022;25(6):104447. https://doi.org/10.1016/j.isci.2022.104447.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Chorianopoulos E, Heger T, Lutz M, Frank D, Bea F, Katus HA, et al. FGF-inducible 14-kDa protein (Fn14) is regulated via the RhoA/ROCK kinase pathway in cardiomyocytes and mediates nuclear factor-kappaB activation by TWEAK. Basic Res Cardiol. 2010;105(2):301–13. https://doi.org/10.1007/s00395-009-0046-y.

    Article  CAS  PubMed  Google Scholar 

  227. Novoyatleva T, Sajjad A, Pogoryelov D, Patra C, Schermuly RT, Engel FB. FGF1-mediated cardiomyocyte cell cycle reentry depends on the interaction of FGFR-1 and Fn14. FASEB J. 2014;28(6):2492–503. https://doi.org/10.1096/fj.13-243576.

    Article  CAS  PubMed  Google Scholar 

  228. Anderson DJ, Kaplan DI, Bell KM, Koutsis K, Haynes JM, Mills RJ, et al. NKX2-5 regulates human cardiomyogenesis via a HEY2 dependent transcriptional network. Nat Commun. 2018;9(1):1373. https://doi.org/10.1038/s41467-018-03714-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Li J, Lv Y, Wang H, Liu Y, Ren J, Wang H. Cardiomyocyte-like cell differentiation by FGF-2 transfection and induction of rat bone marrow mesenchymal stem cells. Tissue Cell. 2021;73:101665. https://doi.org/10.1016/j.tice.2021.101665.

    Article  CAS  PubMed  Google Scholar 

  230. Ehata S, Miyazono K. Bone morphogenetic protein signaling in Cancer; some topics in the recent 10 years. Front Cell Dev Biol. 2022;10:883523. https://doi.org/10.3389/fcell.2022.883523.

    Article  PubMed  PubMed Central  Google Scholar 

  231. de Pater E, Ciampricotti M, Priller F, Veerkamp J, Strate I, Smith K, et al. Bmp signaling exerts opposite effects on cardiac differentiation. Circ Res. 2012;110(4):578–87. https://doi.org/10.1161/CIRCRESAHA.111.261172.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Bach DH, Park HJ, Lee SK. The dual role of bone morphogenetic proteins in Cancer. Mol Ther Oncolytics. 2017;8:1–13. https://doi.org/10.1016/j.omto.2017.10.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Langenfeld E, Deen M, Zachariah E, Langenfeld J. Small molecule antagonist of the bone morphogenetic protein type I receptors suppresses growth and expression of Id1 and Id3 in lung cancer cells expressing Oct4 or nestin. Mol Cancer. 2013;12(1):129. https://doi.org/10.1186/1476-4598-12-129.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Fazeli Z, Omrani MD, Ghaderian SM. Down-regulation of nestin in mesenchymal stem cells derived from peripheral blood through blocking bone morphogenesis pathway. J Cell Commun Signal. 2016;10(4):273–82. https://doi.org/10.1007/s12079-016-0334-x.

    Article  PubMed  PubMed Central  Google Scholar 

  235. Karwacki-Neisius V, Göke J, Osorno R, Halbritter F, Ng JH, Weiße AY, et al. Reduced Oct4 expression directs a robust pluripotent state with distinct signaling activity and increased enhancer occupancy by Oct4 and Nanog. Cell Stem Cell. 2013;12(5):531–45. https://doi.org/10.1016/j.stem.2013.04.023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Ye L, Bokobza S, Li J, Moazzam M, Chen J, Mansel RE, et al. Bone morphogenetic protein-10 (BMP-10) inhibits aggressiveness of breast cancer cells and correlates with poor prognosis in breast cancer. Cancer Sci. 2010;101(10):2137–44. https://doi.org/10.1111/j.1349-7006.2010.01648.x.

    Article  CAS  PubMed  Google Scholar 

  237. Sharma T, Kapoor A, Mandal CC. Duality of bone morphogenetic proteins in cancer: a comprehensive analysis. J Cell Physiol. 2022;237(8):3127–63. https://doi.org/10.1002/jcp.30785.

    Article  CAS  PubMed  Google Scholar 

  238. Zheng Y, Wang X, Wang H, Yan W, Zhang Q, Chang X. Bone morphogenetic protein 2 inhibits hepatocellular carcinoma growth and migration through downregulation of the PI3K/AKT pathway. Tumour Biol. 2014;35(6):5189–98. https://doi.org/10.1007/s13277-014-1673-y.

    Article  CAS  PubMed  Google Scholar 

  239. Maegdefrau U, Bosserhoff AK. BMP activated Smad signaling strongly promotes migration and invasion of hepatocellular carcinoma cells. Exp Mol Pathol. 2012;92(1):74–81. https://doi.org/10.1016/j.yexmp.2011.10.004.

    Article  CAS  PubMed  Google Scholar 

  240. Maegdefrau U, Amann T, Winklmeier A, Braig S, Schubert T, Weiss TS, et al. Bone morphogenetic protein 4 is induced in hepatocellular carcinoma by hypoxia and promotes tumour progression. J Pathol. 2009;218(4):520–9. https://doi.org/10.1002/path.2563.

    Article  CAS  PubMed  Google Scholar 

  241. Moriyama H, Moriyama M, Ozawa T, Tsuruta D, Iguchi T, Tamada S, et al. Notch signaling enhances Stemness by regulating metabolic pathways through modifying p53, NF-κB, and HIF-1α. Stem Cells Dev. 2018;27(13):935–47. https://doi.org/10.1089/scd.2017.0260.

    Article  CAS  PubMed  Google Scholar 

  242. Nemir M, Pedrazzini T. Functional role of notch signaling in the developing and postnatal heart. J Mol Cell Cardiol. 2008;45(4):495–504. https://doi.org/10.1016/j.yjmcc.2008.02.273.

    Article  CAS  PubMed  Google Scholar 

  243. Wang Z, Da Silva TG, Jin K, Han X, Ranganathan P, Zhu X, et al. Notch signaling drives stemness and tumorigenicity of esophageal adenocarcinoma. Cancer Res. 2014;74(21):6364–74. https://doi.org/10.1158/0008-5472.CAN-14-2051.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Kessler M, Hoffmann K, Brinkmann V, Thieck O, Jackisch S, Toelle B, et al. The notch and Wnt pathways regulate stemness and differentiation in human fallopian tube organoids. Nat Commun. 2015;6:8989. https://doi.org/10.1038/ncomms9989.

    Article  CAS  PubMed  Google Scholar 

  245. Leucker TM, Bienengraeber M, Muravyeva M, Baotic I, Weihrauch D, Brzezinska AK, et al. Endothelial-cardiomyocyte crosstalk enhances pharmacological cardioprotection. J Mol Cell Cardiol. 2011;51(5):803–11. https://doi.org/10.1016/j.yjmcc.2011.06.026.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  246. Witman N, Murtuza B, Davis B, Arner A, Morrison JI. Recapitulation of developmental cardiogenesis governs the morphological and functional regeneration of adult newt hearts following injury. Dev Biol. 2011;354(1):67–76. https://doi.org/10.1016/j.ydbio.2011.03.021.

    Article  CAS  PubMed  Google Scholar 

  247. Bergmann O, Zdunek S, Felker A, Salehpour M, Alkass K, Bernard S, et al. Dynamics of cell generation and turnover in the human heart. Cell. 2015;161(7):1566–75. https://doi.org/10.1016/j.cell.2015.05.026.

    Article  CAS  PubMed  Google Scholar 

  248. Wagner JUG, Dimmeler S. Cellular cross-talks in the diseased and aging heart. J Mol Cell Cardiol. 2020;138:136–46. https://doi.org/10.1016/j.yjmcc.2019.11.152.

    Article  CAS  PubMed  Google Scholar 

  249. Hocker JD, Poirion OB, Zhu F, Buchanan J, Zhang K, Chiou J, et al. Cardiac cell type-specific gene regulatory programs and disease risk association. Sci Adv. 2021;7(20):eabf1444. https://doi.org/10.1126/sciadv.abf1444.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. de Soysa TY, Ranade SS, Okawa S, Ravichandran S, Huang Y, Salunga HT, et al. Single-cell analysis of cardiogenesis reveals basis for organ-level developmental defects. Nature. 2019;572(7767):120–4. https://doi.org/10.1038/s41586-019-1414-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Li Y, Lin B, Yang L. Comparative transcriptomic analysis of multiple cardiovascular fates from embryonic stem cells predicts novel regulators in human cardiogenesis. Sci Rep. 2015;5:9758. https://doi.org/10.1038/srep09758.

    Article  PubMed  PubMed Central  Google Scholar 

  252. Mylonas KS, Ziogas IA, Avgerinos DV. Microenvironment in cardiac tumor development: what lies beyond the event horizon? Adv Exp Med Biol. 2020;1226:51–6. https://doi.org/10.1007/978-3-030-36214-0_4.

    Article  CAS  PubMed  Google Scholar 

  253. Cianflone E, Torella M, Biamonte F, De Angelis A, Urbanek K, Costanzo FS, et al. Targeting cardiac stem cell senescence to treat cardiac aging and disease. Cells. 2020;9(6):1558. https://doi.org/10.3390/cells9061558.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Cianflone E, Torella M, Chimenti C, De Angelis A, Beltrami AP, Urbanek K, et al. Adult cardiac stem cell aging: a reversible stochastic phenomenon? Oxidative Med Cell Longev. 2019;2019:5813147. https://doi.org/10.1155/2019/5813147.

    Article  CAS  Google Scholar 

  255. Ballard VL, Edelberg JM. Stem cells and the regeneration of the aging cardiovascular system. Circ Res. 2007;100(8):1116–27. https://doi.org/10.1161/01.RES.0000261964.19115.e3.

    Article  CAS  PubMed  Google Scholar 

  256. Shafi O. Inverse relationship between Alzheimer's disease and cancer, and other factors contributing to Alzheimer's disease: a systematic review. BMC Neurol. 2016;16(1):236. https://doi.org/10.1186/s12883-016-0765-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Hariharan N, Sussman MA. Cardiac aging - getting to the stem of the problem. J Mol Cell Cardiol. 2015;83:32–6. https://doi.org/10.1016/j.yjmcc.2015.04.008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Aguilar-Sanchez C, Michael M, Pennings S. Cardiac stem cells in the postnatal heart: lessons from development. Stem Cells Int. 2018;2018:1247857. https://doi.org/10.1155/2018/1247857.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Bergmann O, Jovinge S. Cardiac regeneration in vivo: mending the heart from within? Stem Cell Res. 2014;13(3 Pt B):523–31. https://doi.org/10.1016/j.scr.2014.07.002.

    Article  CAS  PubMed  Google Scholar 

  260. Pe'er D, Ogawa S, Elhanani O, Keren L, Oliver TG, Wedge D. Tumor heterogeneity. Cancer Cell. 2021;39(8):1015–7. https://doi.org/10.1016/j.ccell.2021.07.009.

    Article  CAS  PubMed  Google Scholar 

  261. Marusyk A, Polyak K. Tumor heterogeneity: causes and consequences. Biochim Biophys Acta. 2010;1805(1):105–17. https://doi.org/10.1016/j.bbcan.2009.11.002.

    Article  CAS  PubMed  Google Scholar 

  262. Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell plasticity. Nature. 2013;501(7467):328–37. https://doi.org/10.1038/nature12624.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  263. Pietras A. Cancer stem cells in tumor heterogeneity. Adv Cancer Res. 2011;112:255–81. https://doi.org/10.1016/B978-0-12-387688-1.00009-0.

    Article  CAS  PubMed  Google Scholar 

  264. Li X, Martinez-Fernandez A, Hartjes KA, Kocher JP, Olson TM, Terzic A, et al. Transcriptional atlas of cardiogenesis maps congenital heart disease interactome. Physiol Genomics. 2014;46(13):482–95. https://doi.org/10.1152/physiolgenomics.00015.2014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. Ameen M, Sundaram L, Shen M, Banerjee A, Kundu S, Nair S, et al. Integrative single-cell analysis of cardiogenesis identifies developmental trajectories and non-coding mutations in congenital heart disease. Cell. 2022;185(26):4937–4953.e23. https://doi.org/10.1016/j.cell.2022.11.028.

    Article  CAS  PubMed  Google Scholar 

  266. Yang D, Gomez-Garcia J, Funakoshi S, Tran T, Fernandes I, Bader GD, et al. Modeling human multi-lineage heart field development with pluripotent stem cells. Cell Stem Cell. 2022;29(9):1382–1401.e8. https://doi.org/10.1016/j.stem.2022.08.007.

    Article  CAS  PubMed  Google Scholar 

  267. Seeger T, Porteus M, Wu JC. Genome editing in cardiovascular biology. Circ Res. 2017;120(5):778–80. https://doi.org/10.1161/CIRCRESAHA.116.310197.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  268. Vermersch E, Jouve C, Hulot JS. CRISPR/Cas9 gene-editing strategies in cardiovascular cells. Cardiovasc Res. 2020;116(5):894–907. https://doi.org/10.1093/cvr/cvz250.

    Article  CAS  PubMed  Google Scholar 

  269. Jiang L, Liang J, Huang W, Ma J, Park KH, Wu Z, et al. Mol Ther. 2022;30(1):54–74. https://doi.org/10.1016/j.ymthe.2021.10.015.

    Article  CAS  PubMed  Google Scholar 

  270. Liu N, Olson EN. CRISPR modeling and correction of cardiovascular disease. Circ Res. 2022;130(12):1827–50. https://doi.org/10.1161/CIRCRESAHA.122.320496.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Motta BM, Pramstaller PP, Hicks AA, Rossini A. The impact of CRISPR/Cas9 technology on cardiac research: from disease modelling to therapeutic approaches. Stem Cells Int. 2017;2017:8960236. https://doi.org/10.1155/2017/8960236.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. Marotta P, Cianflone E, Aquila I, Vicinanza C, Scalise M, Marino F, et al. Combining cell and gene therapy to advance cardiac regeneration. Expert Opin Biol Ther. 2018;18(4):409–23. https://doi.org/10.1080/14712598.2018.1430762.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Dr. Ovais Shafi (OS) and Dr. Ghazia Siddiqui (GS) are inspired by multiple researchers including Dr. Brian J. Druker - physician-scientist at Oregon Health & Science University, in Portland, Oregon. He is the director of OHSU’s Knight Cancer Institute.

Also inspired by Dr. Betty M. Tyler - Johns Hopkins University School of Medicine, Baltimore, MD. Her work on Brain Tumors is marvelous and amazing.

We are inspired by many marvelous researchers including:

Dr. Jason Sheltzer – Yale Medicine.

Dr. Neil Chi - University of California San Diego.

Dr. Nancy Sweitzer - Washington University in St. Louis.

Dr. June Rhee – City of Hope.

Dr. Stephen Stone - Washington University in St. Louis.

Dr. David Nathanson - University of California, Los Angeles.

Dr. Kristin Koutmou - University of Michigan.

Dr. Joerg Herrmann – Mayo Clinic, Rochester, Minnesota.

Dr. Bruce R. Conklin - University of California, San Francisco.

Dr. Jeanne DeCara – The University of Chicago Medicine.

Dr. Tamar Polonsky – The University of Chicago Medicine.

Dr. Joe Costello - University of California, San Francisco.

Dr. Baoli Hu – University of Pittsburgh.

Dr. Susan Dent – Duke Cancer Institute.

Dr. Lauren A. Baldassarre – Yale Medicine.

Dr. Tomas Neilan – Massachusetts General Hospital.

Dr. Eric Hao Ming Yang - University of California, Los Angeles.

Dr. Arjun Deb - University of California, Los Angeles.

Dr. Atsushi Nakano - University of California, Los Angeles.

Dr. Javid Moslehi - University of California, San Francisco.

Dr. Michael S. Sabbah – Mayo Clinic.

Dr. Richard Lee – Harvard Department of Stem Cell and Regenerative biology.

Dr. Sarah C. Hull – Yale Medicine.

Dr. Anju Nohria – Harvard Medical School.

Dr. Xiaoye Shi - University of California Irvine.

Dr. Robert Allan Weinberg - Massachusetts Institute of Technology (MIT).

We are also hopeful to work with these marvelous researchers and scientists in the future.

I am truly thankful and grateful to my remarkable father ‘Dr. Shafi Uddin’ and my remarkable mother ‘Firdous Shafi’. It is the continuous supporting and understanding nature of my parents that also is always helpful for me.

Funding

I declare that there was not any source of funding for this research work. This research study finds evidence from already published research literature to investigate the benign nature and rare occurrence of cardiac myxoma as a possible consequence of the limited cardiac proliferative/ regenerative potential.

Author information

Authors and Affiliations

Authors

Contributions

Authors: Ovais Shafi (OS)*, Ghazia Siddiqui (GS) and Hassam A Jaffry (HAJ)

1. Ovais Shafi (OS)* is the author of the manuscript and was involved in the idea, concept, design, and methodology of the study, literature search and references. He did the writing, editing, and revision of the manuscript. He was involved in drawing the findings, results, conclusions, implications of the study, interpretation of the data and was involved in all aspects of the study. He prepared and wrote discussion, results, conclusions and all sections of the study. OS extracted and analyzed the data. He was involved in critical evaluation and audit of every aspect of the study including data extraction, limitations of the study, references and all others. He was involved in drawing Fig. 1 and Fig. 2. The author read and approved the manuscript.

He is devoted to the research in cardiogenesis, organoids, atherogenesis, cardiac regenerative/ proliferative potential, primary cardiac tumors, oncology, regenerative medicine, neurodegenerative diseases, neurogenesis, organogenesis, finding gene editing targets, disease development mechanisms and research areas in internal medicine.

OS is a MBBS graduate from Sindh Medical College - Dow University of Health Sciences, Karachi, Pakistan.

Email address*: dr.ovaisshafi@gmail.com

Corresponding author: OS

Correspondence to Ovais Shafi

2. Ghazia Siddiqui (GS) is also the author of the manuscript and was involved in the writing, editing, and revision of the manuscript. GS also contributed to the results of the study, drawing the findings, conclusions, implications of the study, discussion, references and interpretation of the data. Her work also includes adherence of the study to relevant PRISMA guidelines. GS audited data extraction and limitations of the study. She critically audited references and gave important input towards adding additional references. She also contributed to all aspects of the results.

She is devoted to the research in cardiogenesis, organoids, atherogenesis, cardiac regenerative/ proliferative potential, primary cardiac tumors, oncology, regenerative medicine, neurodegenerative diseases, neurogenesis, organogenesis, finding gene editing targets, disease development mechanisms and research areas in internal medicine. GS is a MBBS graduate from Sindh Medical College - Jinnah Sindh Medical University, Karachi, Pakistan.

3. Hassam A Jaffry (HAJ), MD is a MBBS graduate from Sindh Medical College-Dow University of Health Sciences. He is also the author of the manuscript and contributed to the writing, editing, revision, and results section of this manuscript along with working on the findings, interpretation of the data and references. His future areas of interest include gene expressions in cardiac amyloidosis, cardiac organoids and cardiac genetic architecture. He is currently in his third year of Internal Medicine residency. He is an aspiring Cardiologist with a great interest in field of research and clinical cardiology. HAJ is also working on other research areas focused on cardiac amyloidosis and preventive cardiology.

Ovais Shafi (OS)*, Ghazia Siddiqui (GS) and Hassam A Jaffry (HAJ) are the authors of this research study. The work and contributions of all authors have been described in detail. All the authors have read and approved the manuscript.

Authors’ information

Ovais Shafi (OS) is a MBBS graduate from Sindh Medical College - Dow University of Health Sciences, Karachi, Pakistan. He is devoted to the research in cardiogenesis, cardiac organoids, cardiac regenerative/ proliferative potential, primary cardiac tumors, neurogenesis, organogenesis, regeneration, tumorigenesis and internal medicine.

He is truly passionate about research in aging and age-related diseases such as cancers, atherosclerosis, Alzheimer’s disease, and aims to become an eminent ‘Physician Scientist’. OS is also interested in multiple research areas including disease development mechanisms, clinical trials, clinical medicine, therapeutics, regenerative medicine, precision medicine including gene therapies, finding disease specific targets for gene therapy, role of disease genomics and epigenetics in diagnosis, management, and therapeutic development. He is dedicated to the field of research and clinical medicine.

Ovais Shafi (OS) and Ghazia Siddiqui (GS) are interested in finding disease origins and development mechanisms, the cell type-specific aberrations in genetic landscape and role of aging in disease development, to contribute towards the advancement of precision medicine and clinical medicine. They are dedicated to devise and conduct studies to find the targets for gene-editing tools such as CRISPR gene editing or epigenome editing to correct or regulate the genes/signaling pathways which become dysregulated in disease development. Their goal is to devise and conduct studies that may serve as a map for genetic and epigenetic targets for the development of new therapeutic approaches and for the development of organoids.

Ghazia Siddiqui (GS) is a MBBS graduate from Sindh Medical College – Jinnah Sindh Medical University, Karachi, Pakistan. Her goal is to be an eminent ‘Physician Scientist’ and is dedicated to the research areas including clinical medicine, disease development mechanisms, cancer biology, clinical trials, and others. In oncology, she is interested in carcinomas, sarcomas, dysplasia, metaplasia, and neoplasia development mechanisms. She is dedicated to develop expertise in ever evolving landscape of research protocols and clinical medicine. She is devoted to the research in cardiogenesis, cardiac organoids, cardiac regenerative/ proliferative potential, primary cardiac tumors, neurogenesis, organogenesis, regeneration, tumorigenesis and internal medicine.

Another major research goal for Ovais Shafi (OS) and Ghazia Siddiqui (GS) is to transform and bridge the research in disease development mechanisms with clinical medicine. They are passionate about research in organoids including cardiac organoids to develop models addressing the frontiers of research in disease development mechanisms, finding currently unknown disease origins, organ development, homeostasis, cell fate specifications and aging related changes in cell type specific architecture. OS and GS are interested in the following areas: clinical medicine, disease development mechanisms, clinical trials, clinical oncology, nephrology, cardiology, tumor origin, invasion and metastasis, role of single nucleotide polymorphisms (SNPs) in cancer development, Robertsonian translocations in cancer development, impact of trisomies including Down syndrome on oncogenesis, and cytoskeletal alterations in oncogenesis. Interested areas also include role the of genetic variations in development of new therapeutics, effects of pharmacotherapy on genetic architecture including epigenome, the role of polymorphisms in drug target genes, impact of disease-specific alterations in cell biology in development of personalized medicine and others.

Hassam A Jaffry (HAJ), MD is a MBBS graduate from Sindh Medical College-Dow University of Health Sciences. He is also the author of the manuscript and contributed to the writing, editing, revision, and results section of this manuscript along with working on the findings, interpretation of the data and references. His future areas of interest include gene expressions in cardiac amyloidosis, cardiac organoids and cardiac genetic architecture. He is currently in his third year of Internal Medicine residency. He is an aspiring Cardiologist with a great interest in field of research and clinical cardiology. HAJ is also working on other research areas focused on cardiac amyloidosis and preventive cardiology.

Corresponding author

Correspondence to Ovais Shafi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shafi, O., Siddiqui, G. & Jaffry, H.A. The benign nature and rare occurrence of cardiac myxoma as a possible consequence of the limited cardiac proliferative/ regenerative potential: a systematic review. BMC Cancer 23, 1245 (2023). https://doi.org/10.1186/s12885-023-11723-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-023-11723-3

Keywords