Growth factors and their roles in cardiac development and regeneration: a narrative review
Review Article

Growth factors and their roles in cardiac development and regeneration: a narrative review

G. Schuyler Brown1, Jihyun Jang1,2, Deqiang Li1,2

1Center for Vascular and Inflammation Diseases, University of Maryland School of Medicine, Baltimore, MD, USA; 2Department of Cardiac Surgery, University of Maryland School of Medicine, Baltimore, MD, USA

Contributions: (I) Conception and design: All authors; (II) Administrative support: None; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: None; (V) Data analysis and interpretation: None; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Deqiang Li, PhD. University of Maryland School of Medicine, 800 West Baltimore St., Room 314, Baltimore, MD 21202, USA. Email: dqli@som.umaryland.edu.

Background and Objective: A network of signaling events is required for the formation of a mature cardiovascular system. During embryogenesis, secretion, or suppression of several growth factors at critical time points and proximities drive this development, including epidermal growth factors (EGFs), fibroblast growth factors (FGFs) and insulin-like growth factors (IGFs) which will be discussed in this review.

Methods: PubMed was searched looking for literatures published between 1990 and March 2022 in English. A review of the literature was completed to find relevant research and terminology around the topics of cardiac development, growth factor signaling and recent advances in cardiovascular regeneration.

Key Content and Findings: Each family of growth factors has been extensively examined by investigators in pre- and postnatal contexts to uncover therapeutic potential against heart disease—a leading cause of death globally. Select ligands with predicted clinical value have been further assessed, such as neuregulin-1 (NRG1)—which binds to the family of EGF receptors (EGFRs) to dimerize and ultimately activate myocyte proliferation through a signal cascade, and IGF-1—which binds to IGF receptors (IGFRs) to regulate metabolism, proliferation, apoptosis, and autophagy.

Conclusions: Collectively, this review supports the need for further investigation on how these critical signaling pathways function during heart development and how the new knowledge might be leveraged to treat cardiovascular disease and promote heart regeneration.

Keywords: Cardiac development; growth factors; regeneration


Received: 18 April 2022; Accepted: 30 September 2022; Published online: 14 October 2022.

doi: 10.21037/pm-22-17


Introduction to cardiac development

During embryogenesis, the heart initially forms a linear heart tube structure, followed by cardiac looping and septation, and eventually grows into a four-chambered heart (1). Meanwhile, multiple components need to form, including the epicardium, myocardium, coronary vascular network, endocardial cushion and ventricular septum and trabeculae (2). This process can be divided into early, mid, and late gestational stages. In the early stages, the linear and looping heart contain two layers of cells. These are the outer layer, comprised of cardiomyocytes (CMs) and an inner layer, comprised of endothelial cells (ECs) (3). Later, the heart will be surrounded by the third layer, known as the epicardium, in the mid stage. The mid stage will also include development of the atrial-ventricular and outflow tract endocardial cushions (1). The process requires constant communications among many cell types such as cardiac progenitor cells, CMs, ECs and fibroblasts. If heart development goes awry, congenital heart disease (CHD) follows. CHD remains to be the number one birth defect in live births worldwide (4).

Importantly, normal cardiac development relies on the proper expression and suppression of growth factors and transcription factors at the right time and place (5). This manuscript will review some of the important signaling events required for proper cardiac development such as epidermal growth factors (EGFs), fibroblast growth factors (FGFs) and insulin-like growth factors (IGFs). We will briefly go over their molecular interactions and outline their noteworthy downstream components. Lastly, we will examine important models which have led to our understanding of the field in recent years and consider how therapeutic heart regeneration can be influenced by the current knowledge of growth factors and their downstream signals. We present this article in accordance with the Narrative Review reporting checklist (available at https://pm.amegroups.com/article/view/10.21037/pm-22-17/rc).


Methods

A review of the literature in PubMed was completed to find relevant research from the mid-1990s to present day—March 2022 (Table 1). The papers were focused more on recent advances and understandings, but we refer to older research to appreciate the progress which has been made over the past 30 years. The primary search terms included Growth Factor Signaling, EGFs, FGFs, IGFs—all as they relate to cardiovascular development. Finally, we compiled recent advances in the field of cardiovascular regeneration and focused on manuscripts relating to the previous growth factors mentioned.

Table 1

The search strategy summary

Items Specification
Date of search March, 2022
Databases and other sources searched PubMed
Search terms used Growth factor signaling, epidermal growth factors, fibroblast growth factors, insulin-like growth factors, cardiovascular development, and cardiovascular regeneration
Timeframe 1990s to March 2022
Inclusion Inclusion: publications in the English language and growth factor actions on either the developmental stage or regeneration potential of the heart
Selection process All authors identified pertinent material for the review

EGF signaling

EGF signaling regulates important processes throughout the body, including the development of a fully functional cardiovascular system. ErbB receptors are well-studied receptor tyrosine kinases (RTKs) which transmit signals from the extracellular environment into the cell via the EGF family of ligands (6). Several ligands have been identified as stimulators of ErbB receptor dimerization, including EGF, neuregulins (NRGs), transforming growth factor-α, amphiregulin, heparin-binding EGF-like growth factor (HB-EGF), betacellulin, epiregulin and epigen (7). These ligands first bind to a monomer to initiate a conformational change in the extracellular domain—for members 1, 3, and 4—which expose a dimerization arm to facilitate pairing (8).

As EGF signaling ligands, NRGs are secreted by endocardial cells and can be tethered to the surface of the cells to assist in the regulation by a transactivating signal discussed later (9). There are 4 NRGs: NRG1–4, with many isoforms for each (10). The NRG-ErbB pathway is mostly involved in ventricular trabeculation and the development of the valves between chambers (11). These steps are valuable in shaping the myocardium and are required for the proper development. Notably, these pathways are involved in differentiation or proliferation, but not simultaneously (12,13).

The first identified EGF receptor (EGFR) is also known as ErbB1 or HER1 (Figure 1) (14). The four members of the receptor family include ErbB1–4, also known as HER1–4 (6). Due to their implications in many diseases—notably with HER2 and breast cancer—they are very well studied and characterized (12). These receptors reside in the plasma membrane in a mostly inactive conformation until activated by their respective ligands. This interaction leads to homo- or heterodimerization with another member of the ErbB family and initiates a downstream signal (15,16). As with other RTKs, the dimerization is followed by transphosphorylation in the cytosolic domains, creating docking sites for proteins to bind and propagate a signal (17). ErbB2 is unique in that it does not have a known ligand. Due to this phenomenon, it typically maintains an active conformation, always revealing a dimerization arm for heterodimerization to take place. It is classified by scientists as a “co-receptor” and tends to be the favorite binding partner (18,19).

Figure 1 The interconnected pathways discussed in the review: EGFs, FGFs, and IGFs and their downstream consequences in the cardiovascular system. β2AR, outlining the current understanding of GPCR transactivation with a typical example. Created with BioRender.com. β2AR, beta-2 adrenergic receptor; MMP, matrix metalloproteinase; ADAM, A disintegrin and metalloproteinase; ErbB, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; FGF, fibroblast growth factor; IGF, insulin-like growth factor; IGFR, insulin-like growth factor receptor; Src, proto-oncogene tyrosine-protein kinase; FAK, focal adhesion kinase; Ras-GTP, rat sarcoma guanosine triphosphate hydrolase oncogene; Raf, serine/threonine-specific protein kinase oncogene; MEK (MAPKK), mitogen-activated protein kinase kinase; ERK, extracellular-signal regulated kinase; PI3K, phosphoinositide 3-kinase; PDK1, phosphoinositide-dependent kinase 1; AKT, protein kinase B; EGF, epidermal growth factor; GPCR, G protein-coupled receptor.

ErbB receptors often begin the mitogen-activated protein kinase (MAPK) pathway, by signaling Ras-GTPase and ultimately leading to extracellular-signal regulated kinase (ERK) translocation into the nucleus (Figure 1); however, more work needs to be done to determine the comprehensive role in cardiac development (20). ErbB signaling is also linked to the Hippo pathway, with the nuclear translocation of Yap/Taz regulating NRG1 production (9).

ErbB activity can also be mediated by transactivation via G protein-coupled receptors (GPCRs). The ErbB system is known to have many of the pro-ligands tethered to the surface of cell membranes (16). After enzymatic cleavage of these ligands, the paracrine signal can be initiated immediately. This transactivation system links GPCRs and ErbBs through various proteinases such as matrix metalloproteinase (MMP) or A disintegrin and metalloproteinase (ADAM) (14). When one of these metalloproteinases is activated, this can lead to an autocrine or paracrine response, which helps to coordinate the signal to nearby cells. This is an important feature of a multicellular system as these GPCR ligands potentially stimulate the receptor of the resident cell followed by activation of a nearby EC, CM, or fibroblast. This creates an opportunity for these GPCRs to directly impact the crosstalk between cell types (21). Within the cardiovascular system, the most common ligand which is shed by metalloproteinase activity is HB-EGF which directly binds to either ErbB1 or ErbB3 (22). A particular example is ADAM17, which is critical for the shedding of NRG1 (23,24). Adding to the intricacy of the system, many relay molecules can also modulate metalloproteinases, including intracellular calcium, protein kinase C, Src kinase and reactive oxygen species (14,25,26).

Early genetic knockout studies have revealed the importance of various ErbB receptors, such as ErbB2, 3, and 4—the most abundant receptors in heart development (19,27-29). Knockout of ErbB2, 3, or 4 led to an embryonic lethal condition between day 10.5 and 13.5 in mice (30,31). This death is attributed to defects in heart morphology, such as hypotrabeculation, thin myocardium and defective cardiac cushion formation (32,33). Knockout of Nrg1 in mice resulted in similar embryonic lethality and cardiac phenotype (34). Taken together, these studies suggest that EGF signaling is critical for early heart morphogenesis, and its continued investigation might be impactful for adult heart regeneration.


FGFs

Another secreted growth factor important in the developing heart is the FGF (Figure 1). This family of proteins has 22 known members divided into 7 subfamilies, which are also needed in the development of many organ systems (35,36). Signaling is typically accomplished through a ligand-receptor interaction, initiated in a paracrine or endocrine manner with a few members performing intracrine functions (37). Paracrine and endocrine ligands are secreted and function along with the cofactor heparan sulfate. This complex will facilitate the dimerization of FGF receptors (FGFRs), leading to the same RTK phenomenon as ErbB signaling (35).

There are 4 receptors in this family, classified as FGFR1–4, with multiple splice variants for each (38). The FGFRs are typically involved in the MAPK and Akt pathways or initiate Jak/STAT signaling and are required for both differentiation and proliferation at different stages and different ligand/receptor interactions (20,39,40). They are also involved in homeostatic events like cell survival, apoptosis or migration (41) as well as angiogenesis—helping to build the coronary network but could also improve the blood flow after an ischemic incident (42). These points highlight the importance of FGF in physiological and pathophysiological contexts.

There are many intricate details involved in the different FGF ligands and receptors, which are still being investigated. In this review, we will discuss only a handful which are involved in heart development. Of the growth factors discussed, this family is the most lacking in available evidence with some controversial discoveries. Multiple FGFs help to regulate CM proliferation via an epicardial- and endocardial-derived signal. These populations of ECs in close contact with the myocytes mediate growth through FGF and other mitogens through paracrine signaling (43).

Several FGFs have been implicated in cardiac development, however there are some disagreements about which ones are involved. There is now a consensus that FGF9, and likely FGF16 and 20, are all essential for myocyte proliferation and could also be necessary for coronary vascular development (1,44). The FGFR1/2 and FGF9-null mice have decreased myocyte proliferation with significant ventricular hypoplasia—an underdeveloped heart (45). Early research suggests that the only relevant FGFs are FGF9, 16 and 20, discounting the importance of others in a developing heart. Later, investigators highlighted the importance of several more ligands, including FGF8, 10, 15, 19 and several others (35). Early work had identified FGF2 as one member which is necessary (46), however this has since been disregarded with later knockout models showing no significant phenotype in mice during embryogenesis.

Elegant studies using embryonic stem cells observed the need for FGFR1 in the differentiation into functional CMs (1,20,47), using both genetic and pharmacological strategies to probe different parts of the signaling pathway. FGFR2 is also an essential component of a developing heart but is lacking in the adult heart (48). Knockout models of FGFRs 1 and 2 in the myocardium led to ventricular wall hypoplasia, giving us a good understanding of how FGF signaling leads to the maturation of the ventricles (33). These findings could help researchers uncover unique strategies in treating cardiovascular disease later in adulthood. There is some conflicting evidence when cells or systems are treated with inhibitors of downstream FGF signaling, such as ERK1/2 or mitogen-activated protein kinase kinase (MEK), with some investigators not able to replicate any findings (49,50). This could be variability in the methods used but could also highlight the importance of time and dose dependence of these signaling events.


IGFs

The last important element we will discuss in the coordinated effort of cardiac development is IGF signaling (Figure 1) (51). IGF-1 and IGF-2 are small growth hormones which perform endocrine, paracrine and autocrine signaling within the system (52,53). A majority of IGF-1 is made and secreted by the liver and expressed postnatally (52), while IGF-2 is predominantly expressed within a developing embryo (54). They are both, however, in many ways critical in embryogenesis and must be well regulated—requiring IGF-binding proteins to stabilize them in their soluble form (55,56). There are two IGF receptors (IGFRs), IGFR-1 and IGFR-2. When a ligand binds, these receptors will autophosphorylate and initiate an appropriate downstream cascade (54).

IGF-1 activates MEF2C, a key transcription factor in cardiac development. This has been confirmed using Luciferase reporter assays showing the promoter enhancement of MEF2C by IGF-1 signaling stimulation (57). IGF-1-induced p38-MAPK activation mediates enhanced phosphorylation of MEF2C. Conversely, inhibitors of phosphoinositide 3-kinase (PI3K) abolish the IGF-1 stimulated MEF2C transcriptional activation (57). IGF-1 also activates other signaling pathways including ERK1/2, PI3K, PKC, PKB, Jak/STAT and PLC (58-60).

IGF-1 and IGF-2 are important for heart development, metabolism, apoptosis, autophagy, and aging (61). An interesting study showed that clinical treatment with IGF-1 could strengthen the heart in late gestation and beyond (62). Downregulation of IGF-1 receptors generate a dilated cardiomyopathy phenotype, while upregulation has led to hypertrophy (51). This phenotype has been observed in a PI3K/Akt dependent manner (63). IGF-1 signaling may not contribute to the maturation of cardiac cells per se, but increases the total number of cells during the developmental phase which may be critical in CVD incidences into adulthood (64). Additionally, reduced amount of IGF-1 in the blood has been associated with a greater risk of acquiring cardiovascular abnormalities (53). IGF-1 could also be anti-fibrotic, an important mechanism which prevents permanent remodeling and scarring—revealing its cardioprotective role (61,65).

Expanding on the key regulatory components of IGF-1, an important consideration for disease risk is now known to be linked to the environment in utero (66). It is evident that issues in gestation are associated with energy balance and regulation throughout life, increasing risk for CVD (67). Many groups have identified a link between fetal growth restriction (FGR) and cardiovascular disease in rats, mice, chickens and sheep (68-70). Long-term consequences of FGR, which were identified over 30 years ago, include decreased cardiac stroke volume, cardiac output, early morbidity and mortality, metabolic impairments leading to obesity, and early CVD-related events as adults (71). IGF-1 was previously speculated as a main driver for this syndrome (72). Authors from the 90s have used sheep as a model of in utero development, particularly using IGF-1 as a treatment for restricted growth. In this sheep model (Figure 2), circulation to the fetus was restricted by partially occluding the umbilical artery and subsequently reintroduce recombinant IGF-1 to evaluate the overall health of the fetus (62). They found that IGF-1 treatment effectively increases the overall heart mass by increasing the proliferation of these ovine cardiac cells (62). These researchers have also taken these experiments a step further and to understand postnatal metabolic control in a myocardial infarction model. These models have shown that administration of various levels of the two IGFs may lead to enhanced healing (54). In a more recent study, scientists treated obese mice with IGF-1 and revealed an improvement in cardiac metabolism, reduction in fibrosis and a decrease in apoptosis; all suggesting a cardioprotective effect of the treatment. These results were accompanied by cardiac progenitor cell survival, proliferation and differentiation which the authors confirm is due to an increase in Akt phosphorylation (73).

Figure 2 FGR ovine model. With the umbilical artery ligation, blood flow is restricted from the embryo leading to many growth abnormalities; however, with the addition of IGF, the embryonic hearts return to a normal phenotype. IGF-1 as a therapeutic option is being explored due to the long-term consequences of the syndrome. Created with BioRender.com. CVD, cardiovascular disease; IGF-1, insulin-like growth factor 1; FGR, fetal growth restriction; IGF, insulin-like growth factor.

Learning from these preclinical studies, an important issue to first understand is the right concentration of IGF to be administered to these FGR patients, as levels over and under the correct value are detrimental to development. This was evident after a series of clinical trials for delivering the right dose of IGF-1 for FGR patients. These clinical trials reveal the complications associated with variable pharmacokinetic properties on fetal and preterm patients, which needed to be treated intravenously (74). In a mouse model of FGR, Alsaied et al. [2017] observe a higher weight gain postnatally compared to control mice, with a higher number of overweight adults (61). This was accompanied by systolic dysfunction at 3 months with no sign of recovery at the end of the experimental timeline (32 weeks).


Cardiac regeneration

Promoting cardiac repair and regeneration has been a huge challenge to the field for several decades. Treatment using stem cells, tissue patches, and trans-differentiation strategies have been tested preclinically and clinically (75). Some have also hypothesized that calling upon endogenous pathways could be the best way to stimulate CM self-renewal (76). Most recently, studies suggest the use of extracellular vesicles could have regenerative potential (77-79). Apparently, a better understanding of these growth signaling pathways such as what are being discussed herein during heart development may give us clearer pictures on when and how we can utilize these pathways to induce cardiac regeneration in adult heart.

Exogenous administration of recombinant NRG-1 into adult mice was shown to stimulate CM proliferation and heart regeneration to various degrees (78,80-82). However, this treatment option may only be effective during earlier periods of life (83). This narrow time window is consistent with an observation of the drastic reduction in the expression of ErbB2 after birth (32). This EGF signaling could be an important element in treating heart failure patients. IGF signaling may have similar effects on heart regeneration (84). Newborn mice can fully regenerate their hearts after injury, although they lose this ability 1 week after birth (56). Interestingly, ablation of IGF-2 in the developing endocardium abolished CM proliferation capacity induced by injury on postnatal day 1 (85). This study illustrated that IGF-2 is an important paracrine factor involved in the early heart regeneration. However, the effects of IGF-1 on adult cardiac repair and regeneration are mixed. For instance, in an MI rat model, administration of IGF-1-adipocyte-derived mesenchymal stem cells in a myocardial infarction rat model showed improved left ventricle ejection fraction (LVEF) but did not decrease infarct scar size when compared to the control group (86). On the contrary, the application of microencapsulated IGF-1 in an ischemia-reperfusion porcine model was reported to improve cardiac function and significantly reduce fibrosis in treated animals compared to controls (79). In a recent study, the overexpression of IGF-1 in cardiac progenitor cells in a pig model of sub-acute myocardial infarction did not achieve significant cardiac functional improvement (87). The overall effects of these exogenous paracrine growth signals on injured hearts might be dependent on the dose, the time and the route of application, as well as the injury models being used. Table 2 outlines some in vivo studies done using these approaches, highlighting which model was used and the therapeutic strategy involved.

Table 2

Studies involving growth factors promoting heart regeneration and repair

Activated growth signaling Organism Intervention/transgenic model References
NRG1 Zebrafish Treatment of AG1478 (a small molecule inhibitor of ErbB receptors) Polizzotti et al., 2015 (83)
NRG1 Zebrafish Homeobox-containing transcription factor prrx1b knockout heart De Bakker et al., 2021 (88)
NRG1/ErbB4 Rat, mouse Injection of recombinant NRG1 Bersell et al., 2009 (80)
ErbB2 Mouse caErbB2 CM D’Uva et al., 2015 (32),
Aharonov et al., 2020 (81)
FGF Zebrafish hsp70:dn-fgfr1; fli1:EGFP transgenic Lepilina et al., 2006 (89)
FGF1 Rat Treatment with FGF1 and p38 inhibitor SB203580 Engel et al., 2006 (90)
FGF2 VEGFA Mouse Reactivation of the fetal epicardial program after MI Zhou et al., 2011 (91)
IGF-2 Zebrafish hsp70:dn-igf1ra-GFP transgenic Huang et al., 2013 (92)
IGF-2 Zebrafish Igf agonist NBI-31772 Choi et al., 2013 (93)
IGFR-1 Mouse Injection of IGF-1 modRNA-injected heart Zangi et al., 2017 (94)
Igf2bp3 Mouse Pre-injection of AAV9-Igfbp3 Wang et al., 2019 (95)

NRG1, neuregulin-1; ErbB, epidermal growth factor receptor; FGF, fibroblast growth factor; VEGFA, vascular endothelial growth factor A; IGF, insulin-like growth factor; IGFR, insulin-like growth factor receptor; Igf2bp, insulin-like growth factor 2 binding protein; PRRX1b, paired-related homeobox 1 transcription factor; hsp70:dn-fgfr1, mouse model dominant negative for the gene fibroblast growth factor receptor 1; caErbB2 CM, cardiomyocyte-specific overexpression of constitutively active ErbB2; fli1:EGFP, mouse model with enhanced green fluorescence protein on friend leukemia integration transcription factor 1 proto-oncogene; MI, myocardial infarction; AAV9, adeno-associated virus serotype 9.


Conclusions and future directions

This review has outlined some of the important growth factor cascades in mediating development of the cardiovascular system. These features are only a fraction of the full interconnected pathway which needs to take place for formation of a fully functioning pump system. EGF, FGF, and IGF families are major contributors to cardiac development and provide an immense number of potential targets for pharmacological or genetic manipulation. However, a more detailed analysis on each of the signaling component will gain a better understanding of these as targets. Growth factor signaling is a promising avenue, and some have even begun clinical treatments with them, such as treating FGR with IGF-1 (74). Similar work is desired to evaluate the therapeutic potential for these growth signals, but these approaches are mostly unresolved in vivo and must be approached with skepticism (82). An important consideration is that these pathways do intertwine at various points. For instance, the MAPK and PI3K pathways are common downstream signaling components for all three growth factor families discussed (Figure 1). Future work is needed to dissect the potential cross talks among these signaling pathways during heart development.

As we have all recognized, developmental events often replay during regeneration, as also illustrated in this review. Hopefully, the knowledge we will gain by studying these signaling events will educate us and then apply them in stimulating heart regeneration.


Acknowledgments

We thank Dr. Li’s lab members for reading and discussing this manuscript.

Funding: This work was supported by the National Heart, Lung, and Blood Institute R01 grant (No. HL153406).


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://pm.amegroups.com/article/view/10.21037/pm-22-17/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://pm.amegroups.com/article/view/10.21037/pm-22-17/coif). All authors report that this work was supported by the National Heart, Lung, and Blood Institute R01 grant (No. HL153406). The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Lavine KJ, Ornitz DM. Fibroblast growth factors and Hedgehogs: at the heart of the epicardial signaling center. Trends Genet 2008;24:33-40. [Crossref] [PubMed]
  2. Gittenberger-de Groot AC, Bartelings MM, Deruiter MC, et al. Basics of cardiac development for the understanding of congenital heart malformations. Pediatr Res 2005;57:169-76. [Crossref] [PubMed]
  3. Dunwoodie SL. Combinatorial signaling in the heart orchestrates cardiac induction, lineage specification and chamber formation. Semin Cell Dev Biol 2007;18:54-66. [Crossref] [PubMed]
  4. Wu W, He J, Shao X. Incidence and mortality trend of congenital heart disease at the global, regional, and national level, 1990-2017. Medicine (Baltimore) 2020;99:e20593. [Crossref] [PubMed]
  5. Rowton M, Guzzetta A, Rydeen AB, et al. Control of cardiomyocyte differentiation timing by intercellular signaling pathways. Semin Cell Dev Biol 2021;118:94-106. [Crossref] [PubMed]
  6. Fuller SJ, Sivarajah K, Sugden PH. ErbB receptors, their ligands, and the consequences of their activation and inhibition in the myocardium. J Mol Cell Cardiol 2008;44:831-54. [Crossref] [PubMed]
  7. Schroeder JA, Jackson LF, Lee DC, et al. Form and function of developing heart valves: coordination by extracellular matrix and growth factor signaling. J Mol Med (Berl) 2003;81:392-403. [Crossref] [PubMed]
  8. Burgess AW, Cho HS, Eigenbrot C, et al. An open-and-shut case? Recent insights into the activation of EGF/ErbB receptors. Mol Cell 2003;12:541-52. [Crossref] [PubMed]
  9. Artap S, Manderfield LJ, Smith CL, et al. Endocardial Hippo signaling regulates myocardial growth and cardiogenesis. Dev Biol 2018;440:22-30. [Crossref] [PubMed]
  10. Falls DL. Neuregulins: functions, forms, and signaling strategies. Exp Cell Res 2003;284:14-30. [Crossref] [PubMed]
  11. Odiete O, Hill MF, Sawyer DB. Neuregulin in cardiovascular development and disease. Circ Res 2012;111:1376-85. [Crossref] [PubMed]
  12. Vagnozzi RJ, Molkentin JD, Houser SR. New Myocyte Formation in the Adult Heart: Endogenous Sources and Therapeutic Implications. Circ Res 2018;123:159-76. [Crossref] [PubMed]
  13. Rentschler S, Zander J, Meyers K, et al. Neuregulin-1 promotes formation of the murine cardiac conduction system. Proc Natl Acad Sci U S A 2002;99:10464-9. [Crossref] [PubMed]
  14. Palanisamy S, Xue C, Ishiyama S, et al. GPCR-ErbB transactivation pathways and clinical implications. Cell Signal 2021;86:110092. [Crossref] [PubMed]
  15. Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 2010;141:1117-34. [Crossref] [PubMed]
  16. Wang Z. Transactivation of Epidermal Growth Factor Receptor by G Protein-Coupled Receptors: Recent Progress, Challenges and Future Research. Int J Mol Sci 2016;17:95. [Crossref] [PubMed]
  17. Jones RB, Gordus A, Krall JA, et al. A quantitative protein interaction network for the ErbB receptors using protein microarrays. Nature 2006;439:168-74. [Crossref] [PubMed]
  18. Citri A, Skaria KB, Yarden Y. The deaf and the dumb: the biology of ErbB-2 and ErbB-3. Exp Cell Res 2003;284:54-65. [Crossref] [PubMed]
  19. Citri A, Yarden Y. EGF-ERBB signalling: towards the systems level. Nat Rev Mol Cell Biol 2006;7:505-16. [Crossref] [PubMed]
  20. Rose BA, Force T, Wang Y. Mitogen-activated protein kinase signaling in the heart: angels versus demons in a heart-breaking tale. Physiol Rev 2010;90:1507-46. [Crossref] [PubMed]
  21. George AJ, Hannan RD, Thomas WG. Unravelling the molecular complexity of GPCR-mediated EGFR transactivation using functional genomics approaches. FEBS J 2013;280:5258-68. [Crossref] [PubMed]
  22. Li Y, Zhang H, Liao W, et al. Transactivated EGFR mediates α1-AR-induced STAT3 activation and cardiac hypertrophy. Am J Physiol Heart Circ Physiol 2011;301:H1941-51. [Crossref] [PubMed]
  23. Shirakabe K, Wakatsuki S, Kurisaki T, et al. Roles of Meltrin beta /ADAM19 in the processing of neuregulin. J Biol Chem 2001;276:9352-8. [Crossref] [PubMed]
  24. Kawai T, Elliott KJ, Scalia R, et al. Contribution of ADAM17 and related ADAMs in cardiovascular diseases. Cell Mol Life Sci 2021;78:4161-87. [Crossref] [PubMed]
  25. Overland AC, Insel PA. Heterotrimeric G proteins directly regulate MMP14/membrane type-1 matrix metalloprotease: a novel mechanism for GPCR-EGFR transactivation. J Biol Chem 2015;290:9941-7. [Crossref] [PubMed]
  26. Sabri A, Short J, Guo J, et al. Protease-activated receptor-1-mediated DNA synthesis in cardiac fibroblast is via epidermal growth factor receptor transactivation: distinct PAR-1 signaling pathways in cardiac fibroblasts and cardiomyocytes. Circ Res 2002;91:532-9. [Crossref] [PubMed]
  27. Zhao YY, Sawyer DR, Baliga RR, et al. Neuregulins promote survival and growth of cardiac myocytes. Persistence of ErbB2 and ErbB4 expression in neonatal and adult ventricular myocytes. J Biol Chem 1998;273:10261-9. [Crossref] [PubMed]
  28. García-Rivello H, Taranda J, Said M, et al. Dilated cardiomyopathy in Erb-b4-deficient ventricular muscle. Am J Physiol Heart Circ Physiol 2005;289:H1153-60. [Crossref] [PubMed]
  29. Tenin G, Clowes C, Wolton K, et al. Erbb2 is required for cardiac atrial electrical activity during development. PLoS One 2014;9:e107041. [Crossref] [PubMed]
  30. Pentassuglia L, Sawyer DB. The role of Neuregulin-1beta/ErbB signaling in the heart. Exp Cell Res 2009;315:627-37. [Crossref] [PubMed]
  31. Kim K, Lee D. ERBB3-dependent AKT and ERK pathways are essential for atrioventricular cushion development in mouse embryos. PLoS One 2021;16:e0259426. [Crossref] [PubMed]
  32. D'Uva G, Aharonov A, Lauriola M, et al. ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation. Nat Cell Biol 2015;17:627-38. [Crossref] [PubMed]
  33. Dong Y, Qian L, Liu J. Molecular and cellular basis of embryonic cardiac chamber maturation. Semin Cell Dev Biol 2021;118:144-9. [Crossref] [PubMed]
  34. Meyer D, Birchmeier C. Multiple essential functions of neuregulin in development. Nature 1995;378:386-90. [Crossref] [PubMed]
  35. Itoh N, Ohta H, Nakayama Y, et al. Roles of FGF Signals in Heart Development, Health, and Disease. Front Cell Dev Biol 2016;4:110. [Crossref] [PubMed]
  36. Farooq M, Khan AW, Kim MS, et al. The Role of Fibroblast Growth Factor (FGF) Signaling in Tissue Repair and Regeneration. Cells 2021;10:3242. [Crossref] [PubMed]
  37. Brewer JR, Mazot P, Soriano P. Genetic insights into the mechanisms of Fgf signaling. Genes Dev 2016;30:751-71. [Crossref] [PubMed]
  38. Dai S, Zhou Z, Chen Z, et al. Fibroblast Growth Factor Receptors (FGFRs): Structures and Small Molecule Inhibitors. Cells 2019;8:614. [Crossref] [PubMed]
  39. Ornitz DM, Itoh N. The Fibroblast Growth Factor signaling pathway. Wiley Interdiscip Rev Dev Biol 2015;4:215-66. [Crossref] [PubMed]
  40. Khosravi F, Ahmadvand N, Bellusci S, et al. The Multifunctional Contribution of FGF Signaling to Cardiac Development, Homeostasis, Disease and Repair. Front Cell Dev Biol 2021;9:672935. [Crossref] [PubMed]
  41. Rochais F, Sturny R, Chao CM, et al. FGF10 promotes regional foetal cardiomyocyte proliferation and adult cardiomyocyte cell-cycle re-entry. Cardiovasc Res 2014;104:432-42. [Crossref] [PubMed]
  42. Schumacher B, Pecher P, von Specht BU, et al. Induction of neoangiogenesis in ischemic myocardium by human growth factors: first clinical results of a new treatment of coronary heart disease. Circulation 1998;97:645-50. [Crossref] [PubMed]
  43. Quijada P, Trembley MA, Small EM. The Role of the Epicardium During Heart Development and Repair. Circ Res 2020;126:377-94. [Crossref] [PubMed]
  44. Colvin JS, Feldman B, Nadeau JH, et al. Genomic organization and embryonic expression of the mouse fibroblast growth factor 9 gene. Dev Dyn 1999;216:72-88. [Crossref] [PubMed]
  45. Lavine KJ, Yu K, White AC, et al. Endocardial and epicardial derived FGF signals regulate myocardial proliferation and differentiation in vivo. Dev Cell 2005;8:85-95. [Crossref] [PubMed]
  46. Pennisi DJ, Ballard VL, Mikawa T. Epicardium is required for the full rate of myocyte proliferation and levels of expression of myocyte mitogenic factors FGF2 and its receptor, FGFR-1, but not for transmural myocardial patterning in the embryonic chick heart. Dev Dyn 2003;228:161-72. [Crossref] [PubMed]
  47. Dell'Era P, Ronca R, Coco L, et al. Fibroblast growth factor receptor-1 is essential for in vitro cardiomyocyte development. Circ Res 2003;93:414-20. [Crossref] [PubMed]
  48. Faul C. Cardiac actions of fibroblast growth factor 23. Bone 2017;100:69-79. [Crossref] [PubMed]
  49. Davidson SM, Morange M. Hsp25 and the p38 MAPK pathway are involved in differentiation of cardiomyocytes. Dev Biol 2000;218:146-60. [Crossref] [PubMed]
  50. Eriksson M, Leppä S. Mitogen-activated protein kinases and activator protein 1 are required for proliferation and cardiomyocyte differentiation of P19 embryonal carcinoma cells. J Biol Chem 2002;277:15992-6001. [Crossref] [PubMed]
  51. Rupert CE, Coulombe KLK. IGF1 and NRG1 Enhance Proliferation, Metabolic Maturity, and the Force-Frequency Response in hESC-Derived Engineered Cardiac Tissues. Stem Cells Int 2017;2017:7648409. [Crossref] [PubMed]
  52. Obradovic M, Zafirovic S, Soskic S, et al. Effects of IGF-1 on the Cardiovascular System. Curr Pharm Des 2019;25:3715-25. [Crossref] [PubMed]
  53. Troncoso R, Ibarra C, Vicencio JM, et al. New insights into IGF-1 signaling in the heart. Trends Endocrinol Metab 2014;25:128-37. [Crossref] [PubMed]
  54. Bergman D, Halje M, Nordin M, et al. Insulin-like growth factor 2 in development and disease: a mini-review. Gerontology 2013;59:240-9. [Crossref] [PubMed]
  55. Puche JE, Castilla-Cortázar I. Human conditions of insulin-like growth factor-I (IGF-I) deficiency. J Transl Med 2012;10:224. [Crossref] [PubMed]
  56. Díaz Del Moral S, Benaouicha M, Muñoz-Chápuli R, et al. The Insulin-like Growth Factor Signalling Pathway in Cardiac Development and Regeneration. Int J Mol Sci 2021;23:234. [Crossref] [PubMed]
  57. Muñoz JP, Collao A, Chiong M, et al. The transcription factor MEF2C mediates cardiomyocyte hypertrophy induced by IGF-1 signaling. Biochem Biophys Res Commun 2009;388:155-60. [Crossref] [PubMed]
  58. Foncea R, Andersson M, Ketterman A, et al. Insulin-like growth factor-I rapidly activates multiple signal transduction pathways in cultured rat cardiac myocytes. J Biol Chem 1997;272:19115-24. [Crossref] [PubMed]
  59. Ibarra C, Estrada M, Carrasco L, et al. Insulin-like growth factor-1 induces an inositol 1,4,5-trisphosphate-dependent increase in nuclear and cytosolic calcium in cultured rat cardiac myocytes. J Biol Chem 2004;279:7554-65. [Crossref] [PubMed]
  60. Nagao H, Cai W, Wewer Albrechtsen NJ, et al. Distinct signaling by insulin and IGF-1 receptors and their extra- and intracellular domains. Proc Natl Acad Sci U S A 2021;118:e2019474118. [Crossref] [PubMed]
  61. Alsaied T, Omar K, James JF, et al. Fetal origins of adult cardiac disease: a novel approach to prevent fetal growth restriction induced cardiac dysfunction using insulin like growth factor. Pediatr Res 2017;81:919-25. [Crossref] [PubMed]
  62. Jonker SS, Giraud GD, Chang EI, et al. Coronary vascular growth matches IGF-1-stimulated cardiac growth in fetal sheep. FASEB J 2020;34:10041-55. [Crossref] [PubMed]
  63. McDevitt TC, Laflamme MA, Murry CE. Proliferation of cardiomyocytes derived from human embryonic stem cells is mediated via the IGF/PI 3-kinase/Akt signaling pathway. J Mol Cell Cardiol 2005;39:865-73. [Crossref] [PubMed]
  64. Sundgren NC, Giraud GD, Schultz JM, et al. Extracellular signal-regulated kinase and phosphoinositol-3 kinase mediate IGF-1 induced proliferation of fetal sheep cardiomyocytes. Am J Physiol Regul Integr Comp Physiol 2003;285:R1481-9. [Crossref] [PubMed]
  65. Perkel D, Naghi J, Agarwal M, et al. The potential effects of IGF-1 and GH on patients with chronic heart failure. J Cardiovasc Pharmacol Ther 2012;17:72-8. [Crossref] [PubMed]
  66. St-Pierre J, Hivert MF, Perron P, et al. IGF2 DNA methylation is a modulator of newborn's fetal growth and development. Epigenetics 2012;7:1125-32. [Crossref] [PubMed]
  67. Symonds ME, Sebert SP, Hyatt MA, et al. Nutritional programming of the metabolic syndrome. Nat Rev Endocrinol 2009;5:604-10. [Crossref] [PubMed]
  68. Menendez-Castro C, Toka O, Fahlbusch F, et al. Impaired myocardial performance in a normotensive rat model of intrauterine growth restriction. Pediatr Res 2014;75:697-706. [Crossref] [PubMed]
  69. Tintu A, Rouwet E, Verlohren S, et al. Hypoxia induces dilated cardiomyopathy in the chick embryo: mechanism, intervention, and long-term consequences. PLoS One 2009;4:e5155. [Crossref] [PubMed]
  70. Habli M, Jones H, Aronow B, et al. Recapitulation of characteristics of human placental vascular insufficiency in a novel mouse model. Placenta 2013;34:1150-8. [Crossref] [PubMed]
  71. Barker DJ, Osmond C, Golding J, et al. Growth in utero, blood pressure in childhood and adult life, and mortality from cardiovascular disease. BMJ 1989;298:564-7. [Crossref] [PubMed]
  72. Katz AB, Keswani SG, Habli M, et al. Placental gene transfer: transgene screening in mice for trophic effects on the placenta. Am J Obstet Gynecol 2009;201:499.e1-8. [Crossref] [PubMed]
  73. Andrade D, Oliveira G, Menezes L, et al. Insulin-like growth factor-1 short-period therapy improves cardiomyopathy stimulating cardiac progenitor cells survival in obese mice. Nutr Metab Cardiovasc Dis 2020;30:151-61. [Crossref] [PubMed]
  74. Hellstrom A, Ley D, Hallberg B, et al. IGF-1 as a Drug for Preterm Infants: A Step-Wise Clinical Development. Curr Pharm Des 2017;23:5964-70. [Crossref] [PubMed]
  75. Zheng L, Du J, Wang Z, et al. Molecular regulation of myocardial proliferation and regeneration. Cell Regen 2021;10:13. [Crossref] [PubMed]
  76. Zuppo DA, Tsang M. Zebrafish heart regeneration: Factors that stimulate cardiomyocyte proliferation. Semin Cell Dev Biol 2020;100:3-10. [Crossref] [PubMed]
  77. Wagner KT, Nash TR, Liu B, et al. Extracellular Vesicles in Cardiac Regeneration: Potential Applications for Tissues-on-a-Chip. Trends Biotechnol 2021;39:755-73. [Crossref] [PubMed]
  78. Heallen TR, Kadow ZA, Kim JH, et al. Stimulating Cardiogenesis as a Treatment for Heart Failure. Circ Res 2019;124:1647-57. [Crossref] [PubMed]
  79. Báez-Díaz C, Blanco-Blázquez V, Sánchez-Margallo FM, et al. Microencapsulated Insulin-Like Growth Factor-1 therapy improves cardiac function and reduces fibrosis in a porcine acute myocardial infarction model. Sci Rep 2020;10:7166. [Crossref] [PubMed]
  80. Bersell K, Arab S, Haring B, et al. Neuregulin1/ErbB4 signaling induces cardiomyocyte proliferation and repair of heart injury. Cell 2009;138:257-70. [Crossref] [PubMed]
  81. Aharonov A, Shakked A, Umansky KB, et al. ERBB2 drives YAP activation and EMT-like processes during cardiac regeneration. Nat Cell Biol 2020;22:1346-56. [Crossref] [PubMed]
  82. Zurek M, Johansson E, Palmer M, et al. Neuregulin-1 Induces Cardiac Hypertrophy and Impairs Cardiac Performance in Post-Myocardial Infarction Rats. Circulation 2020;142:1308-11. [Crossref] [PubMed]
  83. Polizzotti BD, Ganapathy B, Walsh S, et al. Neuregulin stimulation of cardiomyocyte regeneration in mice and human myocardium reveals a therapeutic window. Sci Transl Med 2015;7:281ra45. [Crossref] [PubMed]
  84. Valizadeh A, Asghari S, Mansouri P, et al. The Roles of Signaling Pathways in Cardiac Regeneration. Curr Med Chem 2022;29:2142-66. [Crossref] [PubMed]
  85. Shen H, Gan P, Wang K, et al. Mononuclear diploid cardiomyocytes support neonatal mouse heart regeneration in response to paracrine IGF2 signaling. Elife 2020;9:53071. [Crossref] [PubMed]
  86. Bagno LL, Carvalho D, Mesquita F, et al. Sustained IGF-1 Secretion by Adipose-Derived Stem Cells Improves Infarcted Heart Function. Cell Transplant 2016;25:1609-22. [Crossref] [PubMed]
  87. Prat-Vidal C, Crisóstomo V, Moscoso I, et al. Intracoronary Delivery of Porcine Cardiac Progenitor Cells Overexpressing IGF-1 and HGF in a Pig Model of Sub-Acute Myocardial Infarction. Cells 2021;10:2571. [Crossref] [PubMed]
  88. de Bakker DEM, Bouwman M, Dronkers E, et al. Prrx1b restricts fibrosis and promotes Nrg1-dependent cardiomyocyte proliferation during zebrafish heart regeneration. Development 2021;148:dev198937. [Crossref] [PubMed]
  89. Lepilina A, Coon AN, Kikuchi K, et al. A dynamic epicardial injury response supports progenitor cell activity during zebrafish heart regeneration. Cell 2006;127:607-19. [Crossref] [PubMed]
  90. Engel FB, Hsieh PC, Lee RT, et al. FGF1/p38 MAP kinase inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and rescues function after myocardial infarction. Proc Natl Acad Sci U S A 2006;103:15546-51. [Crossref] [PubMed]
  91. Zhou B, Honor LB, He H, et al. Adult mouse epicardium modulates myocardial injury by secreting paracrine factors. J Clin Invest 2011;121:1894-904. [Crossref] [PubMed]
  92. Huang Y, Harrison MR, Osorio A, et al. Igf Signaling is Required for Cardiomyocyte Proliferation during Zebrafish Heart Development and Regeneration. PLoS One 2013;8:e67266. [Crossref] [PubMed]
  93. Choi WY, Gemberling M, Wang J, et al. In vivo monitoring of cardiomyocyte proliferation to identify chemical modifiers of heart regeneration. Development 2013;140:660-6. [Crossref] [PubMed]
  94. Zangi L, Oliveira MS, Ye LY, et al. Insulin-Like Growth Factor 1 Receptor-Dependent Pathway Drives Epicardial Adipose Tissue Formation After Myocardial Injury. Circulation 2017;135:59-72. [Crossref] [PubMed]
  95. Wang Z, Cui M, Shah AM, et al. Mechanistic basis of neonatal heart regeneration revealed by transcriptome and histone modification profiling. Proc Natl Acad Sci U S A 2019;116:18455-65. [Crossref] [PubMed]
doi: 10.21037/pm-22-17
Cite this article as: Brown GS, Jang J, Li D. Growth factors and their roles in cardiac development and regeneration: a narrative review. Pediatr Med 2023;6:35.

Download Citation