Abstract
The adult mammalian heart is incapable of clinically relevant regeneration. The regenerative deficit in adult mammalian heart contrasts with the fetal and neonatal heart, which demonstrate substantial regenerative capacity after injury. This deficiency in adult mammals is attributable to the lack of resident stem cells after birth, combined with an inability of pre-existing cardiomyocytes to complete cytokinesis. Studies of neonatal heart regeneration in mammals suggest that latent regenerative potential can be re-activated. Dissecting the cellular and molecular mechanisms that promote cardiomyocyte proliferation is key to stimulating true regeneration in adult humans. Here, we review recent advances in our understanding of cardiomyocyte proliferation that suggest molecular approaches to heart regeneration.
Keywords: Cardiomyocyte proliferation, heart regeneration, Hippo-YAP, Wnt, growth factors
1. Introduction
Cardiovascular diseases and, in particular, heart failure remain dominant causes of death worldwide [1]. Heart failure can arise from many pathological remodeling processes, but the most common form of systolic heart failure primarily results from a substantial loss of differentiated cardiomyocytes from infarction. With insufficient cardiomyocyte proliferation capacity, the mammalian heart responds to infarction with repair as opposed to regeneration, leading to replacement of the injured muscle tissue with a meshwork of extracellular matrix (ECM) that forms a scar [2, 3]. The loss of muscle tissue eventually compromises contractility of the remaining myocardium, sometimes leading to heart failure and death [4]. Many patients can have their myocardial infarction aborted by catheter and other reperfusion treatments, but after the first day of infarction, this does not preserve myocardium [5]. Thus, there is a worldwide unmet clinical need for therapeutic strategies for heart regeneration in the days to years after an infarction.
Prenatal mammals have a remarkable ability to regenerate their heart following injury; however, this regenerative ability lasts for a brief window of time shortly after birth. Studies have demonstrated a transient cellular and molecular response from pre-existing cardiomyocytes that leads to substantial myocardial regeneration in neonatal mammalian models [6]. Over the past decade, investigators in the field have invested considerable effort and resources for new strategies to achieve effective cardiac regeneration. These therapeutic strategies range from engineered heart patches, exogenous delivery of cardiac progenitor cells, and cell reprogramming strategies to small molecule delivery [7-9]. While many cell-based therapies prompted initial excitement, they have met considerable clinical challenges such as lack of an ideal cell type and no evidence of true cardiomyocyte regeneration in the heart [10, 11].
Amongst the myriad of potential therapies for cardiac regeneration, no clear clinical strategy has emerged thus far, underscoring the need for further insights into the fundamental biology of the cardiomyocyte and its interaction with neighboring cells during the regenerative process. Several signaling pathways- Hippo/YAP [12, 13], Hedgehog (Hh) [14], and most recently thyroid hormone signaling [15], have been investigated as potential therapeutic pathways for adult heart regeneration. In this Review, we will explore recently-defined cellular and molecular signaling pathways that mediate cardiomyocyte cell cycle re-entry as well as emerging concepts in mammalian heart regeneration. We highlight the fundamental biology of cardiomyocyte renewal in designing strategies to potentially achieve adult mammalian heart regeneration.
1.1. Heart homeostasis
Prenatally, the mammalian heart grows through cardiomyocyte cell division (hyperplasia) as opposed to hypertrophic growth seen in adult mammalian heart [16]. Shortly after birth, cardiomyocytes undergo additional DNA synthesis without cytokinesis, resulting in many becoming binucleated and tetraploid [16]. Strong lines of evidence from 14C isotope decay measurements in humans, and complementary stable isotope labeling in combination with fate mapping studies in rodents, demonstrated a modest level of cardiomyocyte renewal in the adult heart. About 1% of cardiomyocytes are replaced per year in adult mammals [17-20], In mammals, the low rate of cardiomyocyte mitosis was traced back to division of pre-existing cardiomyocytes that were also more likely to be mononucleated and diploid [6, 18, 21-23]. Thus, in the absence of injury, adult mammalian heart homeostasis is maintained by the slow turnover of cardiomyocytes at approximately 1% per year, and this rate declines to 0.5% or less with age [17, 18, 20, 24]. The reason cardiomyocyte turnover declines with age in adult mammals is unknown.
1.2. Adult scarring after infarction
The number of cardiomyocytes entering the cell cycle increases after myocardial injury, but cell division (cytokinesis) does not occur sufficiently to allow regeneration in the adult mammalian heart [18]. Instead an injury to the adult heart results in repair by fibrotic scar formation [6]. The repair process leads to deposition of connective tissue (cardiac fibrosis) and only partial restoration of tissue structure [4]. The wound healing process after a myocardial injury relies on the instructive role of an inflammatory microenvironment [25]. The inflammatory reaction and the proliferative response by nondamaged stromal cells that become activated in situ or get recruited from the bloodstream are essential, as dead heart muscle is prone to rupture, and myocardial rupture is frequently instantaneously fatal [25, 26]. Remaining cardiomyocytes develop compensatory hypertrophy, presumably to maintain organ function [16].
1.3. True heart regeneration: Zebrafish, axolotl, newborn mammals.
Lower vertebrates, such as newts and zebrafish, have significantly higher cardiomyocyte renewal rates compared to mammals [23, 27, 28]. They generally maintain > 95 % diploid cardiomyocytes even as adults and exhibit full regeneration of heart muscle by proliferation of pre-existing cardiomyocytes, even after up to 20 % of the heart has been surgically resected [29, 30]. Interestingly, direct experimental evidence for a higher regenerative capacity in neonates of up to ~ 7 days after birth has been shown in mice after injury by ventricular resection [6], infarction [31], cryoinjury [32, 33], or clamping [34]. The apical resection model was reported to increase cardiomyocytes in M-phase and cytokinesis in neonatal mice [6]. Studies examining proliferative cardiomyocytes between species show that the frequency of mononucleated cardiomyocytes of neonatal mice with retained regenerative potential up to ~ 7 days of birth, resemble that of adult lower vertebrates [35] (Figure 1). Furthermore, case reports from corrective human heart surgeries [36] and myocardial infarction in a newborn child [31] show full regeneration of heart muscle. This suggests that there is a window of high regenerative capacity in the neonatal human heart.
Figure 1: Response to cardiac injury in neonatal and adult mammals.
In this figure, we summarize the response to injury in neonatal and adult mammalian hearts. Following injury, neonatal mice have the ability to successfully regenerate their heart by cardiomyocyte proliferation and neovascularization to replace lost tissue and restore cardiac function. On the other hand, adult mammals are not capable of regenerating significant amounts of heart tissue. Instead, injury in the adult mammalian heart leads to scarring and loss of function. Due to the low cell turnover rate, the adult mammalian heart is incapable of replacing the lost cardiomyocytes. Instead, injury results in cardiomyocyte hypertrophy, replacement fibrosis and chronic inflammation, compromising cardiac function.
1.4. Neonatal and adult cardiomyocytes are different
Since the discovery of the transient regenerative capacity of the neonatal mouse heart, many studies have focused on identifying pathways to re-activate the neonatal regenerative program in the adult heart after injury. Neonatal and adult cardiomyocytes are structurally different. For example, adult cardiomyocytes are elongated with highly aligned uniform sarcomeres and dense myofibrillar structures [37, 38]. In mice, the adult cardiomyocytes switch in contractile gene isoforms from the fetal beta myosin heavy chain (β-MHC or MYH7) to the adult alpha (α-MHC or MYH6) isoform [38]. In addition, there are differences in fuel utilization: glycolysis in neonatal cardiomyocytes vs. primarily fatty acid oxidation in adult cardiomyocytes, which allows more efficient production of ATP [39]. Interestingly, a study comparing the transcriptome of neonatal and adult cardiomyocytes revealed that neonatal cardiomyocytes were more transcriptionally related to endothelial cells and adult leukocytes than to the adult cardiomyocytes. The authors argue that neonatal and adult cardiomyocytes could be considered as two distinct cell types [40], suggesting that neonatal cardiomyocytes are primed from the start to maintain an active cell-cycle program that allows them to continue to proliferate after injury. This raises the question of whether the adult heart has retained a subpopulation of cardiomyocytes with a permissive developmental program that allows the heart to maintain the annual 1% cardiomyocyte renewal.
1.5. Barriers to cardiomyocyte proliferation
In the context of cardiac regeneration, the term “terminally differentiated cardiomyocyte” describes a cardiomyocyte that lacks proliferative capacity. This permanent withdrawal from the cell-cycle in majority of postnatal cardiomyocytes correlates with an extensive change in the epigenome and transcriptome, as well as an increase in abundance of multinucleated and polyploid cardiomyocytes [35, 41-43]. Although the degree of cardiomyocyte polyploidization varies across mammalian species, all the mammalian species studied so far have cardiomyocyte cell-cycle exit shortly after birth [24]. As a result, the majority of murine adult ventricular cardiomyocytes (around 80% or more) [44] are binucleated and tetraploid (2Nx2, where N represents the haploid DNA content per nucleus). Recent evidence demonstrates that the percentage of mononuclear diploid cardiomyocytes in the adult heart depends on the genetic background, that varies substantially among different murine strains and is positively correlated with the potential for cardiac regeneration after injury [45]. There is sparse evidence showing that binucleated cardiomyocytes (2nx2) can undergo division [46]. At the time of birth, the neonatal human heart comprises of primarily mononucleated cardiomyocytes and, approximately 30 % binucleated cardiomyocytes. This proportion of mono – and binucleated cardiomyocytes does not change significantly after birth. However, in humans, cardiomyocyte polyploidization increases between birth and adulthood, and the majority of cardiomyocytes become mononucleated tetraploid (ca. 66 %) [17, 24].
Perinatal oxidative stress and consequent DNA damage due to higher oxygen levels may be a major contributor to the established cardiomyocyte cell-cycle withdrawal [47]. The alternative cell cycle routes to cytokinesis may be more advantageous during oxidative stress, since multiple sets of chromosomes may reassure gene expression simultaneous to gene-inactivating mutations [48]. Furthermore, alternative cell cycle routes to cytokinesis are also more favorable from a contractile and energetic standpoint. For example, dividing cardiomyocytes must disassemble their sarcomeres during M-phase to undergo cytokinesis, resulting in a loss of contractile function. Cardiomyocyte endoreduplication increases cardiomyocyte DNA content to enhance expression of contractile proteins and a higher pump strength, at the expense of cell renewal [49, 50]. Interestingly, Huang and colleagues have shown that mononucleated cardiomyocytes isolated from left atrium and pulmonary vein have different resting membrane potential, pacemaker activity and beating rate than binucleated cardiomyocytes [51] and may exert different functions [52]. In summary, it is suggested that loss of regenerative capacity in the hearts of adult mammals may be a trade-off in order for the adult heart to achieve higher contractility and efficient calcium handling to meet increased metabolic and hemodynamic demands, but sacrificing cytokinesis potential [53].
1.6. Manipulating the barriers to adult cardiomyocyte proliferation
The overall arrest in cardiomyocyte division has been attributed to downregulation of positive cell cycle regulators and centrosome disassembly, resulting in a block in the cardiomyocyte cell cycle [16, 35, 54, 55]. Many of the emerging signaling pathways with the potential to reverse cardiomyocyte cell cycle arrest and promote proliferation, were first discovered during heart development when cardiomyocyte proliferation is substantial. The hypothesis is that developmental signaling pathways may also be present in differentiated cardiomyocytes but are kept suppressed until re-activated with the right stimuli.
Cell cycle factors
Molecular interventions such as over-expression of cardiomyocyte cell cycle regulators have been attempted, but overcoming the multiple cell cycle barriers is a challenge, since cell cycle factors do not universally activate cell division [56-58]. For example, over-expression of cyclin B1-CDC2, and concomitant inhibition of p21/p27 induced cardiomyocyte multinucleation instead of proliferation [56-58]. Furthermore, manipulation of the adult cardiomyocyte cell cycle by overexpression of oncogenes has proven less efficient due to ensuing apoptosis [59]. Recently however, Mohamed et al. used a combination of four cell cycle regulators (4F: cyclin-dependent kinase 1 (CDK-1), CDK4, cyclin CCNB, and CCND) and detected a substantial increase in histone H3-phosphorylation in > 10 % of adult and > 20 % of P7 cardiomyocytes [9]. Next, they utilized intramyocardial delivery of adenoviruses encoding the 4F for 2 weeks post-MI and noted a successful induction of cardiomyocyte cell cycle activity and cardiac repair [9]. Similar results, with a rescue of heart function post MI, were also obtained with a simplified cocktail consisting of CDK4, Cyclin D1 and chemical inhibitors of TGF-β and Weel (2F2i) [9] (Figure 2, strategies for adult mammalian heart regeneration). The authors raise the possibility that the high percentage of pHH3 positive cardiomyocytes after 4 F treatment in vitro may be a result of aberrant phosphorylation of pHH3 or a G2/M arrest [9]. The observed repair in vivo warrants further investigation on the beneficial pro-mitotic effects of the 4Fs on cardiomyocyte proliferation.
Figure 2: Strategies for adult mammalian heart regeneration.
In this figure, we highlight therapeutic strategies to augment cardiac regeneration in adult mammalian hearts after injury. Several therapeutic approaches have been described to achieve cardiac regeneration either by stimulation of cardiomyocyte cell cycle re-entry by stimulation of endogenous pathways or by cell-therapy to deliver in-vitro produced induced pluripotent stem cell (iPSC) – derived cardiomyocytes possibly with engineered scaffolds. Stimulation of endogenous pathways of cardiomyocyte proliferation involve administration of mitogens like miR-199a, cyclins, cyclin dependent kinases (CDKs) to induce cardiomyocyte proliferation. Cell therapy comprises of application of 3D engineered scaffolds or extracellular matrix scaffolds to the heart at the site of injury and delivery of iPSC-derived cardiomyocytes at the site of injury.
Developmental factors
Hippo/Yap pathway
Among developmental signaling pathways, the Hippo/Yap pathway has been studied extensively in context of cardiac regeneration [60, 61]. The Hippo pathway comprises of a series of adaptors and kinases that promote phosphorylation of the transcriptional co-activator Yes-associated protein (YAP) [62], and the transcriptional coactivator with PDZ-binding motif (TAZ). YAP/TAZ remain transcriptionally inactive until phosphorylated, when YAP enters the nuclei and activates gene expression by interacting with DNA-binding transcription factors, such as TEAD (TEA domain family members), SMAD1, RUNX4 (RUNT-related transcription factor 4), T-box 5 (TBX5) and p73 [62] in the heart. Genetic deletion of YAP in mice resulted in underdeveloped hearts and embryonic lethality, while embryonic overexpression of YAP induced hyperproliferation of cardiomyocytes and severely disproportional ventricles and death [61, 63, 64]. Furthermore, deletion of a Hippo signaling component (Salv;Sav1) in cardiomyocytes showed evidence of regenerated myocardium after apical resection at P8 [60]. Subsequently, when YAP1 was over-expressed in the adult heart, cardiomyocyte proliferation was detected. In depth experiments revealed that Yap/TEAD complexes induce activation of the insulin-like growth factor pathway and upregulation of nuclear WNT target gene ß-catenin to induce cardiomyocyte cell cycle re-entry and regeneration post injury [64, 65]. Studies in mice have revealed that targeting the Hippo-YAP pathway has potential for increasing cardiomyocyte proliferation; however, it is important to point out that YAP is also a potent oncogene and stimulation of YAP may simultaneously modulate a maladaptive response. Recently, it has been shown that YAP activation induces cardiomyocyte dedifferentiation and heart failure when pressure overload is present [66].
Meis1
Developmental transcription factor Meis1 plays a key role in the cell cycle by inducing expression of p21 and thereby blocking cardiomyocyte cell cycle activity [67]. Conditional deletion of Meis1 in neonatal and adult mouse cardiomyocytes is sufficient to induce cell cycle reentry. However, direct deletion of Meis1 downstream targets, p21 or p27 or both, does not induce cardiomyocyte proliferation [68], but instead increases tumorigenesis. Interestingly, overexpression of the essential transcription factor Tbx20 in adult cardiomyocytes promotes cardiomyogenesis by directly acting on cell-cycle repressors p21, Meis1 and Btg2 [69]. This provides further evidence of the important role of Meis1 in cardiomyocyte cell cycle activity.
E2F
An additional example of developmental transcription factors that act as key regulators of cardiomyocyte proliferation is the E2F transcription factor family. The E2F family controls the synthesis of genes that regulate cell cycle entrance and passage. E2F1, 2 and 3, in particular, are considered “transactivators” that activate transcription of target genes for DNA replication and G1/S transition, thus cell proliferation [70]. E2F activity is regulated by a family of proteins, which includes the retinoblastoma protein (pRB) tumor suppressor, p107 and p130. Overexpression of E2F-1 increases cardiomyocyte apoptosis, while overexpression of the transcription factor E2F4 stimulates cardiomyocyte cycling in vivo [71, 72].
Hedgehog pathway
The role of Hedgehog (Hh) signaling pathway in adult heart homeostasis has been unclear, probably in part due to low expression of this signaling pathway in adult cardiomyocytes. Ischemic heart injury, however, reactivates the Hh pathway [73]. Recently, Singh and colleagues, compared the apical resection heart model in newts and mice and identified a novel HH-Gli1-Mycn gene regulatory network that promotes heart regeneration [14]. Fate-mapping experiments showed that HH-Gli1-Mycn induces cardiomyocyte proliferation of pre-existing cardiomyocytes. Furthermore, chromatin-immunoprecipitation sequencing (ChIP-Seq) analysis revealed that HH-Gli1-Mycn acts regulatory by directly regulating the expression of cyclin-dependent kinases including cyclinD2, cyclinE1 and Cdc7[14].
Neuregulin
The growth factor neuregulin-1 (NRG-1) and its receptors (ERBB2 and ERBB4), are critical for cardiac development and maintenance of adult heart [74]. NRG-1 is an extracellular factor with multiple effects on cell survival, metabolism, angiogenesis, and sarcomere disassembly in addition to cardiomyocyte division [75]. The mode of action of NRG1 seems to be age dependent. During embryogenesis, BMP10 - NRG1 signaling between endocardium and myocardium initiates differentiation and maturation of cardiomyocytes [76]. However, in the adult heart, NRG1 signals via ERBB2 and ERBB4 receptor to stimulate cardiomyocyte proliferation in vivo [77, 78] (Figure 3). EGF/NRG/ERBB signaling in the cardiovascular system is clearly important but has yet to lead to a regenerative or heart failure therapy.
Figure 3: Signaling pathways that modulate cardiomyocyte proliferation.
Here, we outline the major signaling pathways that have been identified to modulate adult cardiomyocyte cell-cycle re-entry. Well-characterized endogenous pathways regulating cardiomyocyte proliferation include Wnt – ß-catenin signaling, neuregulin and Hippo-YAP signaling. Activation of Wnt signaling by the binding of Wnt to its receptor frizzled, results in the translocation of ß-catenin to the nucleus, activating transcriptional activity for cell proliferation. Binding of neuregulin to its receptors ErbB2 and ErbB4 mediate translocation of Yap to the nucleus and activation of transcriptional events that lead to cell proliferation. On the other hand, reactive oxygen species – mediated DNA damage results in cell cycle arrest via Atm phosphorylation and Weel activation.
Hypoxia Inducible Factors and the hypoxic response
Mounting evidence suggests that hypoxic conditions can induce cardiomyocyte proliferation and cardiac regeneration in lower vertebrates and in the adult murine heart after an MI [79-81]. In a study by Nakada et al. [79], adult mice were subjected to chronic severe hypoxemia, with observed reduction in oxidative DNA and altered mitochondrial metabolism. This was sufficient to induce reactivation of cardiomyocyte mitosis and heart repair after induced injury [79, 80]. This reactivation of cell cycle activity was associated with hypoxic activation of hypoxia-inducible factor 1α (HIF1α) [79]. Scavenging of hydrogen peroxide (H2O2) (e.g. catalase overexpression) was suggested to cause a delayed cardiomyocyte cell cycle arrest, whereas hyperoxic or ROS-generating conditions accelerate it [82]. It should be noted, however, that scavenging of H2O2 by transgenic overexpression in zebrafish was shown to impair heart regeneration [83]. Thus, increased ROS levels can lead to activation of the DNA-damage-response pathway, which can induce cell-cycle arrest of postnatal cardiomyocytes, and it will be important to determine how hypoxic signaling may be exploited in adult mammals and humans.
These important discoveries reveal that the cardiomyocyte cell cycle is tightly controlled and highlights that there is a preserved cellular program in place to prevent loss of organ function by uncontrolled proliferation. The functional relevance and the regulatory mechanism that drive post-natal multinucleation and physiological polyploidization in the heart, remain incompletely understood. What is clear is that both in homeostasis and after injury, the majority of adult mammalian cardiomyocytes undergo DNA synthesis but do not complete cytokinesis. A key to enhanced heart regeneration could be to define a subpopulation of cardiomyocytes that might be more amenable and responsive to mitotic stimuli.
2. Secreted factors induce cardiomyocyte proliferation
Although the mass of the heart is primarily cardiomyocytes, cardiomyocytes are outnumbered compared to the total of fibroblast, endothelial, smooth muscle, and inflammatory cells [84]. These cells create a microenvironment that most likely contribute to the regulation of cardiomyocyte proliferation, through paracrine signaling of secreted molecules. A number of secreted factors, cytokines and growth factors appear to play a role in cardiomyocyte cell cycle reentry and cardiac repair [85-87]. The paracrine signaling effects could be important as some cell therapies appear to benefit the heart even in the absence of new cardiomyocyte formation [88-90]. Cardiac regeneration may thus be stimulated by exogenous factors produced by non-cardiomyocytes (Figure 3, signaling pathways that modulate cardiomyocyte proliferation).
2.1. Reactivation of cardiomyocyte cell cycle with secreted factors
Normal thyroid hormone secretion is necessary for optimal cardiac performance [91] and thyroid hormone supplementation has been proposed as treatment for heart failure patients [92]. Recently, Hirose et al. demonstrated that impairing thyroid hormone T3 signaling by either pharmacological inhibition or gene-editing approaches caused an increase in diploid cardiomyocytes by 21 % in mice at postnatal day 14. Moreover, blocking thyroid hormone signaling increased cardiomyocyte proliferative rates, such that the hearts of these mice were 37% larger than controls and contained significantly higher number of cardiomyocytes. This also resulted in a 62 % reduction of scar area and improved cardiac function after myocardial injury [15]. Thyroid (T3) administration in vivo has been previously shown to induce activation of PI3K/Akt/mTOR pathway and stimulate cardiomyocyte protein synthesis [93]. More detailed understanding of how thyroid signaling regulates regenerative capacity is warranted.
Several growth factors have also been described as capable of stimulating cardiomyocyte cell-cycle reentry. Fibroblast growth factor 2 (FGF2) was one of the first growth factors shown to promote neonatal cardiomyocyte DNA synthesis via protein kinase C activation [94]. Phase I trials have shown that intramyocardial injection of fibroblast growth factor 1 (FGF1) during coronary artery bypass graft surgery improves collateral artery growth and capillary proliferation by stimulating anti-apoptotic activity [95]. Treatment with FGF1 and a p38 mitogen-activated protein kinase inhibitor (p38i) in rats after myocardial injury showed promising effects on cardiomyocyte proliferation [96]. Other hormones with similar outcome are Growth hormone-releasing hormone agonist (GHRH) and Hepatocyte growth factor (HGF). Studies in swine models showed that delivery of IGF1/hepatocyte growth factor (HGF) or Growth hormone-releasing hormone agonist (GHRH-A) after ischemic injury significantly reduced infarct size by increasing formation of smaller cardiomyocytes and increased capillary density [97] [98]. At this time, several clinical trials (Phase I-III) using HGF gene therapy for peripheral arterial occlusive disease and coronary heart disease are undergoing [99].
Among the angiogenic secreted factors that have potential to stimulate cardiac regeneration is Follistatin-like 1 (Fstl1) [100, 101]. Fstl1 protects cardiomyocytes from cell death and hypertrophy in vitro, but it also suppresses cardiomyocyte differentiation from stem cells by inhibiting BMPs. In vivo, Fstl1 is normally present in healthy epicardium, but re-distributes to the myocardium upon injury [8]. Surprisingly, transgenic overexpression of Fstl1 in the adult heart does not elicit a regenerative response [8]. Instead, local delivery of Fstl1 through the epicardium, in both mice and swine, induced repair after an MI [8]. The authors conclude that Fstl1 must be synthesized, glycosylated and secreted by epicardium to act like a cardiomyogenic factor and emphasize that the source of the signaling molecule as well as the post translational modifications has a crucial impact.
Acute inflammation occurs immediately after heart injury and stimulates a regenerative response in the neonatal mouse heart [85]. Analyses of the global gene expression changes in the neonatal mouse heart following apical resection identified Interleukin 6 (IL6) [85] and Interleukin 13 (IL13) [102] as regulators of cardiomyocyte proliferation. Downstream regulators, STAT6, STAT3, and periostin were found to be critical mediators of inflammatory signaling in cardiomyocytes. When IL-13 was added to cultured neonatal and adult cardiomyocytes, ERK1/2 and Akt were activated, resulting in increased proliferation [86]. Mice lacking IL13 in the heart showed decreased cardiomyocyte proliferation and impaired heart repair after apical resection. Periostin, however, a secreted extracellular matrix protein, is highly expressed in the developing heart [103] but it is almost undetectable in the adult heart. Remarkably, periostin levels increase following ischemic injury, primarily by fibroblast activation [104]. Extracellular delivery of periostin after an MI stimulated cardiomyocyte mitosis and cardiac regeneration [105]. This finding has been challenged by others, who reported either no effect on cardiomyocyte proliferation or an increased cardiac fibrosis [106]. In summary, manipulation of signaling molecules produced by the non-cardiomyocyte cell populations in the heart might be important for overcoming post-natal cardiomyocyte cell cycle arrest to promote cardiac regeneration.
2.2. miR in cardiac repair
Accumulating evidence has shown that cardiomyocyte proliferation is under control of the miRNA network. MicroRNAs (miRNAs) are small non-coding RNAs (~22 nt in length) that play a role in posttranscriptional regulation of gene expression in the heart (reviewed elsewhere [107, 108]). For example, inhibition of the miR-15 family increased cardiomyocyte proliferation and improved cardiac function after an MI [21]. The miRNA-302-367 cluster has been shown to target the Hippo pathway to promote cardiomyocyte mitosis. Interestingly, this cluster of miRNAs can reactivate cardiomyocyte renewal in a Hippo-independent manner by stimulating homeodomain transcription factor Pitx2 expression. Pitx2 activates gene expression of electron transport chain components and ROS scavengers. Specifically, Nrf2, a regulator of the antioxidant response, is targeted, adding additional evidence of oxidative stress being a barrier to cardiomyocyte mitosis [109].
Other miRNAs, such as the miR-17-92 cluster, miR-214, and the miR-222 cluster have been reported to contribute to cardiac repair in vivo [110-113]. In an unbiased screen of more than 800 human miRNAs, multiple miRNAs were identified and shown to promote neonatal rat cardiomyocyte proliferation [114]. Among these miRNAs, cardiac overexpression of miR-199a or miR-590 induced cardiac repair of infarcted heart while reducing fibrosis. More recently, Borden et al. showed significant cardiac repair after MI, upon miR-294 treatment in mice [115]. Another study by Gabisonia et al. demonstrated that expression of human miR-199a in infarcted pig hearts can stimulate cardiac repair [116]. However, the authors also emphasize that miRNA need to be carefully dosed as repeated delivery and uncontrolled expression of the miR-199a resulted in sudden arrhythmic death of most of the treated pigs. Therefore, the positive outcomes in preclinical studies demonstrate the therapeutic potential of miRNAs, but this therapy needs to be tightly controlled.
3. Conslusion and Future directions (conluding remarks)
In conclusion, promoting de novo cardiomyocyte proliferation and differentiation, while modulating cardiac fibrosis, is a promising approach for restoring cardiac function following ischemic injury. Studies have demonstrated that cardiomyocyte mitosis can be induced with hormones, growth factors, miRNAs and also small molecules. New discoveries and promising preclinical outcomes demonstrating cardiac repair provide optimism regarding novel therapies for human systolic heart failure. However, we must also acknowledge and address the multiple challenges such as increased proliferation of non-myocyte cell types in the heart and the ubiquitous possibility of inducing cardiac arrhythmias. True heart regeneration is a complex and precisely controlled process. Therefore, a deeper understanding of cardiomyocyte biology and the crosstalk between the different cell populations in the heart is required for development of efficient approaches for repairing the mammalian heart.
Acknowledgements
This work was supported by grants from the Leducq Foundation and NIH (AG047131, HL119230, and HL137710 to RTL and F32 HL146000 to NN). Richard Lee is a co-founder of, member of the scientific advisory board for, and holds private equity in Elevian, Inc, a company that aims to develop medicines to restore regenerative capacity. Elevian also provides sponsored research support to the Lee Lab.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Das SR, Delling FN, Djousse L, Elkind MSV, Ferguson JF, Fornage M, Jordan LC, Khan SS, Kissela BM, Knutson KL, Kwan TW, Lackland DT, Lewis TT, Lichtman JH, Longenecker CT, Loop MS, Lutsey PL, Martin SS, Matsushita K, Moran AE, Mussolino ME, O'Flaherty M, Pandey A, Perak AM, Rosamond WD, Roth GA, Sampson UKA, Satou GM, Schroeder EB, Shah SH, Spartano NL, Stokes A, Tirschwell DL, Tsao CW, Turakhia MP, VanWagner LB, Wilkins JT, Wong SS, Virani SS, E. American Heart Association Council on, C. Prevention Statistics, S. Stroke Statistics, Heart Disease and Stroke Statistics-2019 Update: A Report From the American Heart Association, Circulation 139(10) (2019) e56–e528. [DOI] [PubMed] [Google Scholar]
- [2].van den Borne SW, Diez J, Blankesteijn WM, Verjans J, Hofstra L, Narula J, Myocardial remodeling after infarction: the role of myofibroblasts, Nat Rev Cardiol 7(1) (2010) 30–7. [DOI] [PubMed] [Google Scholar]
- [3].Laflamme MA, Murry CE, Heart regeneration, Nature 473(7347) (2011) 326–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Whelan RS, Kaplinskiy V, Kitsis RN, Cell death in the pathogenesis of heart disease: mechanisms and significance, Annu Rev Physiol 72 (2010) 19–44 [DOI] [PubMed] [Google Scholar]
- [5].Verheugt FW, Gersh BJ, Armstrong PW, Aborted myocardial infarction: a new target for reperfusion therapy, Eur Heart J 27(8) (2006) 901–4. [DOI] [PubMed] [Google Scholar]
- [6].Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson, E.N. Olson, H.A. Sadek, Transient regenerative potential of the neonatal mouse heart, Science 331(6020) (2011) 1078–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Liu YW, Chen B, Yang X, Fugate JA, Kalucki FA, Futakuchi-Tsuchida A, Couture L, Vogel KW, Astley CA, Baldessari A, Ogle J, Don CW, Steinberg ZL, Seslar SP, Tuck SA, Tsuchida H, Naumova AV, Dupras SK, Lyu MS, Lee J, Hailey DW, Reinecke H, Pabon L, Fryer BH, MacLellan WR, Thies RS, Murry CE, Human embryonic stem cell-derived cardiomyocytes restore function in infarcted hearts of non-human primates, Nat Biotechnol 36(7) (2018) 597–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Wei K, Serpooshan V, Hurtado C, Diez-Cunado M, Zhao M, Maruyama S, Zhu W, Fajardo G, Noseda M, Nakamura K, Tian X, Liu Q, Wang A, Matsuura Y, Bushway P, Cai W, Savchenko A, Mahmoudi M, Schneider MD, van den Hoff MJ, Butte MJ, Yang PC, Walsh K, Zhou B, Bernstein D, Mercola M, Ruiz-Lozano P, Epicardial FSTL1 reconstitution regenerates the adult mammalian heart, Nature 525(7570) (2015) 479–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Mohamed TMA, Ang YS, Radzinsky E, Zhou P, Huang Y, Elfenbein A, Foley A, Magnitsky S, Srivastava D, Regulation of Cell Cycle to Stimulate Adult Cardiomyocyte Proliferation and Cardiac Regeneration, Cell 173(1) (2018) 104–116 e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Chong X. Yang JJ, Don CW, Minami E, Liu YW, Weyers JJ, Mahoney WM, Van Biber B, Cook SM, Palpant NJ, Gantz JA, Fugate JA, Muskheli V, Gough GM, Vogel KW, Astley CA, Hotchkiss CE, Baldessari A, Pabon L, Reinecke H, Gill EA, Nelson V, Kiem HP, Laflamme MA, Murry CE, Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts, Nature 510(7504) (2014) 273–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Shiba Y, Gomibuchi T, Seto T, Wada Y, Ichimura H, Tanaka Y, Ogasawara T, Okada K, Shiba N, Sakamoto K, Ido D, Shiina T, Ohkura M, Nakai J, Uno N, Kazuki Y, Oshimura M, Minami I, Ikeda U, Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts, Nature 538(7625) (2016) 388–391. [DOI] [PubMed] [Google Scholar]
- [12].Ikeda S, Sadoshima J, Regulation of Myocardial Cell Growth and Death by the Hippo Pathway, Circ J 80(7) (2016) 1511–9. [DOI] [PubMed] [Google Scholar]
- [13].Xiao Y, Leach J, Wang J, Martin JF, Hippo/Yap Signaling in Cardiac Development and Regeneration, Curr Treat Options Cardiovasc Med 18(6) (2016) 38. [DOI] [PubMed] [Google Scholar]
- [14].Singh BN, Koyano-Nakagawa N, Gong W, Moskowitz IP, Weaver CV, Braunlin E, Das S, van Berlo JH, Garry MG, Garry DJ, A conserved HH-Gli1-Mycn network regulates heart regeneration from newt to human, Nat Commun 9(1) (2018) 4237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Hirose K, Payumo AY, Cutie S, Hoang A, Zhang H, Guyot R, Lunn D, Bigley RB, Yu H, Wang J, Smith M, Gillett E, Muroy SE, Schmid T, Wilson E, Field KA, Reeder DM, Maden M, Yartsev MM, Wolfgang MJ, Grutzner F, Scanlan TS, Szweda LI, Buffenstein R, Hu G, Flamant F, Olgin JE, Huang GN, Evidence for hormonal control of heart regenerative capacity during endothermy acquisition, Science 364(6436) (2019) 184–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Soonpaa MH, Kim KK, Pajak L, Franklin M, Field LJ, Cardiomyocyte DNA synthesis and binucleation during murine development, Am J Physiol 271(5 Pt 2) (1996) H2183–9. [DOI] [PubMed] [Google Scholar]
- [17].Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabe-Heider F, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, Jovinge S, Frisen J, Evidence for cardiomyocyte renewal in humans, Science 324(5923) (2009) 98–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Senyo SE, Steinhauser ML, Pizzimenti CL, Yang VK, Cai L, Wang M, Wu TD, Guerquin-Kern JL, Lechene CP, Lee RT, Mammalian heart renewal by pre-existing cardiomyocytes, Nature 493(7432) (2013) 433–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Alkass K, Panula J, Westman M, Wu TD, Guerquin-Kern JL, Bergmann O, No Evidence for Cardiomyocyte Number Expansion in Preadolescent Mice, Cell 163(4) (2015) 1026–36. [DOI] [PubMed] [Google Scholar]
- [20].Eschenhagen T, Bolli R, Braun T, Field LJ, Fleischmann BK, Frisen J, Giacca M, Hare JM, Houser S, Lee RT, Marban E, Martin JF, Molkentin JD, Murry CE, Riley PR, Ruiz-Lozano P, Sadek HA, Sussman MA, Hill JA, Cardiomyocyte Regeneration: A Consensus Statement, Circulation 136(7) (2017) 680–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Porrello ER, Mahmoud AI, Simpson E, Johnson BA, Grinsfelder D, Canseco D, Mammen PP, Rothermel BA, Olson EN, Sadek HA, Regulation of neonatal and adult mammalian heart regeneration by the miR-15 family, Proc Natl Acad Sci U S A 110(1) (2013) 187–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Ali SR, Hippenmeyer S, Saadat LV, Luo L, Weissman IL, Ardehali R, Existing cardiomyocytes generate cardiomyocytes at a low rate after birth in mice, Proc Natl Acad Sci U S A 111(24) (2014) 8850–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Laube F, Heister M, Scholz C, Borchardt T, Braun T, Re-programming of newt cardiomyocytes is induced by tissue regeneration, J Cell Sci 119(Pt 22) (2006) 4719–29. [DOI] [PubMed] [Google Scholar]
- [24].Mollova M, Bersell K, Walsh S, Savla J, Das LT, Park SY, Silberstein LE, Dos Remedios CG, Graham D, Colan S, Kuhn B, Cardiomyocyte proliferation contributes to heart growth in young humans, Proc Natl Acad Sci U S A 110(4) (2013) 1446–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Godwin JW, Pinto AR, Rosenthal NA, Chasing the recipe for a pro-regenerative immune system, Semin Cell Dev Biol 61 (2017) 71–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Sattler S, Fairchild P, Watt FM, Rosenthal N, Harding SE, The adaptive immune response to cardiac injury-the true roadblock to effective regenerative therapies?, NPJ Regen Med 2 (2017) 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Kikuchi K, Holdway JE, Werdich AA, Anderson RM, Fang Y, Egnaczyk GF, Evans T, Macrae CA, Stainier DY, Poss KD, Primary contribution to zebrafish heart regeneration by gata4(+) cardiomyocytes, Nature 464(7288) (2010) 601–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Jopling C, Sleep E, Raya M, Marti M, Raya A, Izpisua Belmonte JC, Zebrafish heart regeneration occurs by cardiomyocyte dedifferentiation and proliferation, Nature 464(7288) (2010) 606–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Gonzalez-Rosa JM, Sharpe M, Field D, Soonpaa MH, Field LJ, Burns CE, Burns CG, Myocardial Polyploidization Creates a Barrier to Heart Regeneration in Zebrafish, Dev Cell 44(4) (2018) 433–446 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Poss KD, Wilson LG, Keating MT, Heart regeneration in zebrafish, Science 298(5601) (2002) 2188–90. [DOI] [PubMed] [Google Scholar]
- [31].Haubner BJ, Adamowicz-Brice M, Khadayate S, Tiefenthaler V, Metzler B, Aitman T, Penninger JM, Complete cardiac regeneration in a mouse model of myocardial infarction, Aging (Albany NY) 4(12) (2012) 966–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Jesty SA, Steffey MA, Lee FK, Breitbach M, Hesse M, Reining S, Lee JC, Doran RM, Nikitin AY, Fleischmann BK, Kotlikoff MI, c-kit+ precursors support postinfarction myogenesis in the neonatal, but not adult, heart, Proc Natl Acad Sci U S A 109(33) (2012) 13380–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Strungs EG, Ongstad EL, O'Quinn MP, Palatinus JA, Jourdan LJ, Gourdie RG, Cryoinjury models of the adult and neonatal mouse heart for studies of scarring and regeneration, Methods Mol Biol 1037 (2013) 343–53. [DOI] [PubMed] [Google Scholar]
- [34].Bryant DM, O'Meara CC, Ho NN, Gannon J, Cai L, Lee RT, A systematic analysis of neonatal mouse heart regeneration after apical resection, J Mol Cell Cardiol 79 (2015) 315–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Walsh S, Ponten A, Fleischmann BK, Jovinge S, Cardiomyocyte cell cycle control and growth estimation in vivo--an analysis based on cardiomyocyte nuclei, Cardiovasc Res 86(3) (2010) 365–73. [DOI] [PubMed] [Google Scholar]
- [36].Fratz S, Hager A, Schreiber C, Schwaiger M, Hess J, Stern HC, Long-term myocardial scarring after operation for anomalous left coronary artery from the pulmonary artery, Ann Thorac Surg 92(5) (2011) 1761–5. [DOI] [PubMed] [Google Scholar]
- [37].Bray MA, Sheehy SP, Parker KK, Sarcomere alignment is regulated by myocyte shape, Cell Motil Cytoskeleton 65(8) (2008) 641–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Weiss A, Leinwand LA, The mammalian myosin heavy chain gene family, Annu Rev Cell Dev Biol 12 (1996) 417–39. [DOI] [PubMed] [Google Scholar]
- [39].Ellen Kreipke R, Wang Y, Miklas JW, Mathieu J, Ruohola-Baker H, Metabolic remodeling in early development and cardiomyocyte maturation, Semin Cell Dev Biol 52 (2016) 84–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Quaife-Ryan GA, Sim CB, Ziemann M, Kaspi A, Rafehi H, Ramialison M, El-Osta A, Hudson JE, Porrello ER, Multicellular Transcriptional Analysis of Mammalian Heart Regeneration, Circulation 136(12) (2017) 1123–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Bruneau BG, Signaling and transcriptional networks in heart development and regeneration, Cold Spring Harb Perspect Biol 5(3) (2013) a008292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Paige SL, Plonowska K, Xu A, Wu SM, Molecular regulation of cardiomyocyte differentiation, Circ Res 116(2) (2015) 341–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Bergmann O, Zdunek S, Felker A, Salehpour M, Alkass K, Bernard S, Sjostrom SL, Szewczykowska M, Jackowska T, Dos Remedios C, Malm T, Andra M, Jashari R, Nyengaard JR, Possnert G, Jovinge S, Druid H, Frisen J, Dynamics of Cell Generation and Turnover in the Human Heart, Cell 161(7) (2015) 1566–75. [DOI] [PubMed] [Google Scholar]
- [44].Naqvi N, Li M, Calvert JW, Tejada T, Lambert JP, Wu J, Kesteven SH, Holman SR, Matsuda T, Lovelock JD, Howard WW, Iismaa SE, Chan AY, Crawford BH, Wagner MB, Martin DI, Lefer DJ, Graham RM, Husain A, A proliferative burst during preadolescence establishes the final cardiomyocyte number, Cell 157(4) (2014) 795–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Patterson M, Barske L, Van Handel B, Rau CD, Gan P, Sharma A, Parikh S, Denholtz M, Huang Y, Yamaguchi Y, Shen H, Allayee H, Crump JG, Force TI, Lien CL, Makita T, Lusis AJ, Kumar SR, Sucov HM, Frequency of mononuclear diploid cardiomyocytes underlies natural variation in heart regeneration, Nat Genet 49(9) (2017) 1346–1353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Matz DG, Oberpriller JO, Oberpriller JC, Comparison of mitosis in binucleated and mononucleated newt cardiac myocytes, Anat Rec 251(2) (1998) 245–55. [DOI] [PubMed] [Google Scholar]
- [47].Puente BN, Kimura W, Muralidhar SA, Moon J, Amatruda JF, Phelps KL, Grinsfelder D, Rothermel BA, Chen R, Garcia JA, Santos CX, Thet S, Mori E, Kinter MT, Rindler PM, Zacchigna S, Mukherjee S, Chen DJ, Mahmoud AI, Giacca M, Rabinovitch PS, Aroumougame A, Shah AM, Szweda LI, Sadek HA, The oxygen-rich postnatal environment induces cardiomyocyte cell-cycle arrest through DNA damage response, Cell 157(3) (2014) 565–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Gu Z, Steinmetz LM, Gu X, Scharfe C, Davis RW, Li WH, Role of duplicate genes in genetic robustness against null mutations, Nature 421(6918) (2003) 63–6. [DOI] [PubMed] [Google Scholar]
- [49].Tiemann K, Weyer D, Djoufack PC, Ghanem A, Lewalter T, Dreiner U, Meyer R, Grohe C, Fink KB, Increasing myocardial contraction art blood pressure in C57BL/6 mice during early postnatal development, Am J Physiol Heart Circ Physiol 284(2) (2003) H464–74. [DOI] [PubMed] [Google Scholar]
- [50].O'Tierney PF, Anderson DF, Faber JJ, Louey S, Thornburg KL, Giraud GD, Reduced systolic pressure load decreases cell-cycle activity in the fetal sheep heart, Am J Physiol Regul Integr Comp Physiol 299(2) (2010) R573–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Huang CF, Chen YC, Yeh HI, Chen SA, Mononucleated and binucleated cardiomyocytes in left atrium and pulmonary vein have different electrical activity and calcium dynamics, Prog Biophys Mol Biol 108(1-2) (2012) 64–73. [DOI] [PubMed] [Google Scholar]
- [52].Stephen MJ, Poindexter BJ, Moolman JA, Sheikh-Hamad D, Bick RJ, Do binucleate cardiomyocytes have a role in myocardial repair? Insights using isolated rodent myocytes and cell culture, Open Cardiovasc Med J 3 (2009) 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Brodsky V, Sarkisov DS, Arefyeva AM, Panova NW, Gvasava IG, Polyploidy in cardiac myocytes of normal and hypertrophic human hearts; range of values, Virchows Arch 424(4) (1994) 429–35. [DOI] [PubMed] [Google Scholar]
- [54].Zebrowski DC, Vergarajauregui S, Wu CC, Piatkowski T, Becker R, Leone M, Hirth S, Ricciardi F, Falk N, Giessl A, Just S, Braun T, Weidinger G, Engel FB, Developmental alterations in centrosome integrity contribute to the post-mitotic state of mammalian cardiomyocytes, Elife 4 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Kang MJ, Kim JS, Chae SW, Koh KN, Koh GY, Cyclins and cyclin dependent kinases during cardiac development, Mol Cells 7(3) (1997) 360–6. [PubMed] [Google Scholar]
- [56].Tane S, Ikenishi A, Okayama H, Iwamoto N, Nakayama KI, Takeuchi T, CDK inhibitors, p21(Cip1) and p27(Kip1), participate in cell cycle exit of mammalian cardiomyocytes, Biochem Biophys Res Commun 443(3) (2014) 1105–9. [DOI] [PubMed] [Google Scholar]
- [57].Poolman RA, Gilchrist R, Brooks G, Cell cycle profiles and expressions of p21CIP1 AND P27KIP1 during myocyte development, Int J Cardiol 67(2) (1998) 133–42. [DOI] [PubMed] [Google Scholar]
- [58].Di Stefano V, Giacca M, Capogrossi MC, Crescenzi M, Martelli F, Knockdown of cyclin-dependent kinase inhibitors induces cardiomyocyte re-entry in the cell cycle, J Biol Chem 286(10) (2011) 8644–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Liu Y, Kitsis RN, Induction of DNA synthesis and apoptosis in cardiac myocytes by E1A oncoprotein, J Cell Biol 133(2) (1996) 325–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Heallen T, Morikawa Y, Leach J, Tao G, Willerson JT, Johnson RL, Martin JF, Hippo signaling impedes adult heart regeneration, Development 140(23) (2013) 4683–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].von Gise A, Lin Z, Schlegelmilch K, Honor LB, Pan GM, Buck JN, Ma Q, Ishiwata T, Zhou B, Camargo FD, Pu WT, YAP1, the nuclear target of Hippo signaling, stimulates heart growth through cardiomyocyte proliferation but not hypertrophy, Proc Natl Acad Sci U S A 109(7) (2012) 2394–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Zhao B, Tumaneng K, Guan KL, The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal, Nat Cell Biol 13(8) (2011) 877–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Xin M, Kim Y, Sutherland LB, Qi X, McAnally J, Schwartz RJ, Richardson JA, Bassel-Duby R, Olson EN, Regulation of insulin-like growth factor signaling by Yap governs cardiomyocyte proliferation and embryonic heart size, Sci Signal 4(196) (2011) ra70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Xin M, Kim Y, Sutherland LB, Murakami M, Qi X, McAnally J, Porrello ER, Mahmoud AI, Tan W, Shelton JM, Richardson JA, Sadek HA, Bassel-Duby R, Olson EN, Hippo pathway effector Yap promotes cardiac regeneration, Pro Natl Acad Sci U S A 110(34) (2013)13839–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Leach JP, Heallen T, Zhang M, Rahmani M, Morikawa Y, Hill MC, Segura A, Willerson JT, Martin JF, Hippo pathway deficiency reverses systolic heart failure after infarction, Nature 550(7675) (2017) 260–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Ikeda S, Mizushima W, Sciarretta S, Abdellatif M, Zhai P, Mukai R, Fefelova N, Oka SI, Nakamura M, Del Re DP, Farrance I, Park JY, Tian B, Xie LH, Kumar M, Hsu CP, Sadayappan S, Shimokawa H, Lim DS, Saoshima J, Hippo Deficiency Leads to Cardiac Dysfunction Accompanied by Cardiomyocyte Dedifferentiation During Pressure Overload, Circ Res 124(2) (2019) 292–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Mahmoud AI, Kocabas F, Muralidhar SA, Kimura W, Koura AS, Thet S, Porrello ER, Sadek HA, Meis1 regulates postnatal cardiomyocyte cell cycle arrest, Nature 497(7448) (2013) 249–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Garcia-Fernandez RA, Garcia-Palencia P, Sanchez MA, Gil-Gomez G, Sanchez B, Rollan E, Martin-Caballero J, Flores JM, Combined loss of p21(waf1/cip1) and p27(kip1) enhances tumorigenesis in mice, Lab Invest 91(11) (2011) 1634–42. [DOI] [PubMed] [Google Scholar]
- [69].Xiang FL, Guo M, Yutzey KE, Overexpression of Tbx20 in Adult Cardiomyocytes Promotes Proliferation and Improves Cardiac Function After Myocardial Infarction, Circulation 133(11) (2016) 1081–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Attwooll C, Lazzerini Denchi E, Helin K, The E2F family: specific functions and overlapping interests, EMBO J 23(24) (2004) 4709–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Ebelt H, Zhang Y, Kampke A, Xu J, Schlitt A, Buerke M, Muller-Werdan U, Werdan K, Braun T, E2F2 expression induces proliferation of terminally differentiated cardiomyocytes in vivo, Cardiovasc Res 80(2) (2008) 219–26. [DOI] [PubMed] [Google Scholar]
- [72].Yurkova N, Shaw J, Blackie K, Weidman D, Jayas R, Flynn B, Kirshenbaum LA, The cell cycle factor E2F-1 activates Bnip3 and the intrinsic death pathway in ventricular myocytes, Circ Res 102(4) (2008) 472–9. [DOI] [PubMed] [Google Scholar]
- [73].Kusano KF, Pola R, Murayama T, Curry C, Kawamoto A, Iwakura A, Shintani S, Ii M, Asai J, Tkebuchava T, Thorne T, Takenaka H, Aikawa R, Goukassian D, von Samson P, Hamada H, Yoon YS, Silver M, Eaton E, Ma H, Heyd L, Kearney M, Munger W, Porter JA, Kishore R, Losordo DW, Sonic hedgehog myocardial gene therapy: tissue repair through transient reconstitution of embryonic signaling, Nat Med 11(11) (2005) 1197–204. [DOI] [PubMed] [Google Scholar]
- [74].Gassmann M, Casagranda F, Orioli D, Simon H, Lai C, Klein R, Lemke G, Aberrant neural and cardiac development in mice lacking the ErbB4 neuregulin receptor, Nature 378(6555) (1995) 390–4. [DOI] [PubMed] [Google Scholar]
- [75].Parodi EM, Kuhn B, Signalling between microvascular endothelium and cardiomyocytes through neuregulin, Cardiovasc Res 102(2) (2014) 194–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Grego-Bessa J, Luna-Zurita L, del Monte G, Bolos V, Melgar P, Arandilla A, Garratt AN, Zang H, Mukouyama YS, Chen H, Shou W, Ballestar E, Esteller M, Rojas A, Perez-Pomares JM, de la Pompa JL, Notch signaling is essential for ventricular chamber development, Dev Cell 12(3) (2007) 415–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Bersell K, Arab S, Haring B, Kuhn B, Neuregulin1/ErbB4 signaling induces cardiomyocyte proliferation and repair of heart injury, Cell 138(2) (2009) 257–70 [DOI] [PubMed] [Google Scholar]
- [78].D'Uva G, Aharonov A, Lauriola M, Kain D, Yahalom-Ronen Y, Carvalho S, Weisinger K, Bassat E, Rajchman D, Yifa O, Lysenko M, Konfino T, Hegesh J, Brenner O, Neeman M, Yarden Y, Leor J, Sarig R, Harvey RP, Tzahor E, ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation, Nat Cell Biol 17(5) (2015) 627–38. [DOI] [PubMed] [Google Scholar]
- [79].Nakada Y, Canseco DC, Thet S, Abdisalaam S, Asaithamby A, Santos CX, Shah AM, Zhang H, Faber JE, Kinter MT, Szweda LI, Xing C, Hu Z, Deberardinis RJ, Schiattarella G, Hill JA, Oz O, Lu Z, Zhang CC, Kimura W Sadek HA, Hypoxia induces heart regeneration in adult mice, Nature 541(7636) (2017) 222–227. [DOI] [PubMed] [Google Scholar]
- [80].Kimura W, Xiao F, Canseco DC, Muralidhar S, Thet S, Zhang HM, Abderrahman Y, Chen R, Garcia JA, Shelton JM, Richardson JA, Ashour AM, Asaithamby A, Liang H, Xing C, Lu Z, Zhang CC, Sadek HA, Hypoxia fate mapping identifies cycling cardiomyocytes in the adult heart, Nature 523(7559) (2015) 226–30. [DOI] [PubMed] [Google Scholar]
- [81].Jopling C, Sune G, Faucherre A, Fabregat C, Izpisua Belmonte JC, Hypoxia induces myocardial regeneration in zebrafish, Circulation 126(25) (2012) 3017–27. [DOI] [PubMed] [Google Scholar]
- [82].Guimaraes-Camboa N, Stowe J, Aneas I, Sakabe N, Cattaneo P, Henderson L, Kilberg MS, Johnson RS, Chen J, McCulloch AD, Nobrega MA, Evans SM, Zambon AC, HIF1alpha Represses Cell Stress Pathways to Allow Proliferation of Hypoxic Fetal Cardiomyocytes, Dev Cell 33(5) (2015) 507–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Han P, Zhou XH, Chang N, Xiao CL, Yan S, Ren H, Yang XZ, Zhang ML, Wu Q, Tang B, Diao JP, Zhu X, Zhang C, Li CY, Cheng H, Xiong JW, Hydrogen peroxide primes heart regeneration with a derepression mechanism, Cell Res 24(9) (2014) 1091–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Skelly DA, Squiers GT, McLellan MA, Bolisetty MT, Robson P, Rosenthal NA, Pinto AR, Single-Cell Transcriptional Profiling Reveals Cellular Diversity and Intercommunication in the Mouse Heart, Cell Rep 22(3) (2018) 600–610. [DOI] [PubMed] [Google Scholar]
- [85].Han C, Nie Y, Lian H, Liu R, He F, Huang H, Hu S, Acute inflammation stimulates a regenerative response in the neonatal mouse heart, Cell Res 25(10) (2015) 1137–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Wodsedalek DJ, Paddock SJ, Wan TC, Auchampach JA, Kenarsary A, Tsaih SW, Flister MJ, O'Meara CC, IL-13 promotes in vivo neonatal cardiomyocyte cell cycle activity and heart regeneration, Am J Physiol Heart Circ Physiol 316(1) (2019) H24–H34. [DOI] [PubMed] [Google Scholar]
- [87].Andreadou I, Cabrera-Fuentes HA, Devaux Y, Frangogiannis NG, Frantz S, Guzik T, Liehn EA, Gomes CPC, Schulz R, Hausenloy DJ, Immune cells as targets for cardioprotection: new players and novel therapeutic opportunities, Cardiovasc Res 115(7) (2019) 1117–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Xiong Q, Hill KL, Li Q, Suntharalingam P, Mansoor A, Wang X, Jameel MN, Zhang P, Swingen C, Kaufman DS, Zhang J, A fibrin patch-based enhanced delivery of human embryonic stem cell-derived vascular cell transplantation in a porcine model of postinfarction left ventricular remodeling, Stem Cells 29(2) (2011) 367–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Riegler J, Tiburcy M, Ebert A, Tzatzalos E, Raaz U, Abilez OJ, Shen Q, Kooreman NG, Neofytou E, Chen VC, Wang M, Meyer T, Tsao PS, Connolly AJ, Couture LA, Gold JD, Zimmermann WH, Wu JC, Human Engineered Heart Muscles Engraft and Survive Long Term in a Rodent Myocardial Infarction Model, Circ Res 117(8) (2015) 720–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Rafatian G, Davis DR, Concise Review: Heart-Derived Cell Therapy 2.0: Paracrine Strategies to Increase Therapeutic Repair of Injured Myocardium, Stem Cells 36(12) (2018) 1794–1803. [DOI] [PubMed] [Google Scholar]
- [91].Biondi B, Palmieri EA, Lombardi G, Fazio S, Effects of subclinical thyroid dysfunction on the heart, Ann Intern Med 137(11) (2002) 904–14. [DOI] [PubMed] [Google Scholar]
- [92].Vale C, Neves JS, von Hafe M, Borges-Canha M, Leite-Moreira A, The Role of Thyroid Hormones in Heart Failure, Cardiovasc Drugs Ther 33(2) (2019) 179–188. [DOI] [PubMed] [Google Scholar]
- [93].Kenessey A, Ojamaa K, Thyroid hormone stimulates protein synthesis in the cardiomyocyte by activating the Akt-mTOR and p70S6K pathways, J Biol Chem 281(30) (2006) 20666–72. [DOI] [PubMed] [Google Scholar]
- [94].Kardami E, Banerji S, Doble BW, Dang X, Fandrich RR, Jin Y, Cattini PA, PKC-dependent phosphorylation may regulate the ability of connexin43 to inhibit DNA synthesis, Cell Commun Adhes 10(4-6) (2003) 293–7. [DOI] [PubMed] [Google Scholar]
- [95].Schumacher B, Pecher P, von Specht BU, Stegmann T, Induction of neoangiogenesis in ischemic myocardium by human growth factors: first clinical results of a new treatment of coronary heart disease, Circulation 97(7) (1998) 645–50. [DOI] [PubMed] [Google Scholar]
- [96].Engel FB, Hsieh PC, Lee RT, Keating MT, FGF1/p38 MAP kinase inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and rescues function after myocardial infarction, Proc Natl Acad Sci U S A 103(42) (2006) 15546–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Bagno LL, Kanashiro-Takeuchi RM, Suncion VY, Golpanian S, Karantalis V, Wolf A, Wang B, Premer C, Balkan W, Rodriguez J, Valdes D, Rosado M, Block NL, Goldstein P, Morales A, Cai RZ, Sha W, Schally AV, Hare JM, Growth hormone-releasing hormone agonists reduce myocardial infarct scar in swine with subacute ischemic cardiomyopathy, J Am Heart Assoc 4(4) (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Koudstaal S, Bastings MM, Feyen DA, Waring CD, van Slochteren FJ, Dankers PY, Torella D, Sluijter JP, Nadal-Ginard B, Doevendans PA, Ellison GM, Chamuleau SA, Sustained delivery of insulin-like growth factor-1/hepatocyte growth factor stimulates endogenous cardiac repair in the chronic infarcted pig heart, J Cardiovasc Transl Res 7(2) (2014) 232–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Wang LS, Wang H, Zhang QL, Yang ZJ, Kong FX, Wu CT, Hepatocyte Growth Factor Gene Therapy for Ischemic Diseases, Hum Gene Ther 29(4) (2018) 413–423. [DOI] [PubMed] [Google Scholar]
- [100].Ouchi N, Oshima Y, Ohashi K, Higuchi A, Ikegami C, Izumiya Y, Walsh K, Follistatin-like 1, a secreted muscle protein, promotes endothelial cell function and revascularization in ischemic tissue through a nitric-oxide synthase-dependent mechanism, J Biol Chem 283(47) (2008) 32802–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Oshima Y, Ouchi N, Sato K, Izumiya Y, Pimentel DR, Walsh K, Follistatin-like 1 is an Akt-regulated cardioprotective factor that is secreted by the heart, Circulation 117(24) (2008) 3099–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].O'Meara CC, Wamstad JA, Gladstone RA, Fomovsky GM, Butty VL, Shrikumar A, Gannon JB, Boyer LA, Lee RT, Transcriptional reversion of cardiac myocyte fate during mammalian cardiac regeneration, Circ Res 116(5) (2015) 804–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Snider P, Hinton RB, Moreno-Rodriguez RA, Wang J, Rogers R, Lindsley A, Li F, Ingram DA, Menick D, Field L, Firulli AB, Molkentin JD, Markwald R, Conway SJ, Periostin is required for maturation and extracellular matrix stabilization of noncardiomyocyte lineages of the heart, Circ Res 102(7) (2008) 752–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Shimazaki M, Nakamura K, Kii I, Kashima T, Amizuka N, Li M, Saito M, Fukuda K, Nishiyama T, Kitajima S, Saga Y, Fukayama M, Sata M, Kudo A, Periostin is essential for cardiac healing after acute myocardial infarction, J Exp Med 205(2) (2008) 295–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Kuhn B, del Monte F, Hajjar RJ, Chang YS, Lebeche D, Arab S, Keating MT, Periostin induces proliferation of differentiated cardiomyocytes and promotes cardiac repair, Nat Med 13(8) (2007) 962–9. [DOI] [PubMed] [Google Scholar]
- [106].Lorts A, Schwanekamp JA, Elrod JW, Sargent MA, Molkentin JD, Genetic manipulation of periostin expression in the heart does not affect myocyte content, cell cycle activity, or cardiac repair, Circ Res 104(1) (2009) e1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Liu N, Olson EN, MicroRNA regulatory networks in cardiovascular development, Dev Cell 18(4) (2010) 510–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].van Rooij E, Olson EN, MicroRNA therapeutics for cardiovascular disease: opportunities and obstacles, Nat Rev Drug Discov 11(11) (2012) 860–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Tao G, Kahr PC, Morikawa Y, Zhang M, Rahmani M, Heallen TR, Li L, Sun Z, Olson EN, Amendt BA, Martin JF, Pitx2 promotes heart repair by activating the antioxidant response after cardiac injury, Nature 534(7605) (2016) 119–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Chen J, Huang ZP, Seok HY, Ding J, Kataoka M, Zhang Z, Hu X, Wang G, Lin Z, Wang S, Pu WT, Liao R, Wang DZ, mir-17-92 cluster is required for and sufficient to induce cardiomyocyte proliferationin postnatal and adult hearts, Circ Res 112(12) (2013) 1557–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Liu X, Xiao J, Zhu H, Wei X, Platt C, Damilano F, Xiao C, Bezzerides V, Bostrom P, Che L, Zhang C, Spiegelman BM, Rosenzweig A, miR-222 is necessary for exercise-induced cardiac growth and protects against pathological cardiac remodeling, Cell Metab 21(4) (2015) 584–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Vujic A, Lerchenmuller C, Wu TD, Guillermier C, Rabolli CP, Gonzalez E, Senyo SE, Liu X, Guerquin-Kern JL, Steinhauser ML, Lee RT, Rosenzweig A, Exercise induces new cardiomyocyte generation in the adult mammalian heart, Nat Commun 9(1) (2018) 1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Aurora AB, Mahmoud AI, Luo X, Johnson BA, van Rooij E, Matsuzaki S, Humphries KM, Hill JA, Bassel-Duby R, Sadek HA, Olson EN, MicroRNA-214 protects the mouse heart from ischemic injury by controlling Ca(2)(+) overload and cell death, J Clin Invest 122(4) (2012) 1222–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Eulalio A, Mano M, Dal Ferro M, Zentilin L, Sinagra G, Zacchigna S, Giacca M, Functional screening identifies miRNAs inducing cardiac regeneration, Nature 492(7429) (2012) 376–81. [DOI] [PubMed] [Google Scholar]
- [115].Borden A, Kurian J, Nickoloff E, Yang Y, Troupes CD, Ibetti J, Lucchese AM, Gao E, Mohsin S, Koch WJ, Houser SR, Kishore R, Khan M, Transient Introduction of miR-294 in the Heart Promotes Cardiomyocyte Cell Cycle Reentry After Injury, Circ Res 125(1) (2019) 14–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Gabisonia K, Prosdocimo G, Aquaro GD, Carlucci L, Zentilin L, Secco I, Ali H, Braga L, Gorgodze N, Bernini F, Burchielli S, Collesi C, Zandona L, Sinagra G, Piacenti M, Zacchigna S, Bussani R, Recchia FA, Giacca M, MicroRNA therapy stimulates uncontrolled cardiac repair after myocardial infarction in pigs, Nature 569(7756) (2019) 418–422. [DOI] [PMC free article] [PubMed] [Google Scholar]



