Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 May 24.
Published in final edited form as: Circ Res. 2019 May 24;124(11):1647–1657. doi: 10.1161/CIRCRESAHA.118.313573

Stimulating Cardiogenesis as a Treatment for Heart Failure

Todd R Heallen 1,2, Zachary A Kadow 2,3, Jun Wang 4, Jong W Kim 1, James F Martin 1,2,3,5
PMCID: PMC6534162  NIHMSID: NIHMS1527932  PMID: 31120819

Abstract

Following myocardial injury, cardiomyocyte loss cannot be corrected by using currently available clinical treatments. In recent years, considerable effort has been made to develop cell-based cardiac repair therapies aimed at correcting for this loss. An exciting crop of recent studies reveal that inducing endogenous repair and proliferation of cardiomyocytes may be a viable option for regenerating injured myocardium. Here, we review current heart failure treatments, the state of cardiomyocyte renewal in mammals, and the molecular signals that stimulate cardiomyocyte proliferation. These signals include growth factors, intrinsic signaling pathways, microRNAs, and cell cycle regulators. Animal model cardiac regeneration studies reveal that modulation of exogenous and cell intrinsic signaling pathways can induce re-entry of adult cardiomyocytes into the cell cycle. Using direct myocardial injection, epicardial patch delivery or systemic administration of growth molecules, these studies show that inducing endogenous cardiomyocytes to self-renew is an exciting and promising therapeutic strategy to treat cardiac injury in humans.

Keywords: cardiomyocyte, regeneration, heart failure, signaling pathways, ploidy

Subject Terms: Heart Failure, Treatment

Current heart failure treatments

Heart failure (HF), in which the heart fails to pump sufficient blood to meet the body’s need, is the leading global cause of death1. There are currently estimated 38 million patients suffering from HF worldwide, and this number continues to rise2. HF mortality rates are high, and currently about half of HF patients die within 5 years of diagnosis. Heart failure is a complicated pathological process initiated by heart injury, which arises most commonly from ischemic heart disease associated with coronary vascular disease3, 4. In addition, HF can result from chemotherapy exposure and other forms of cardiomyopathy, such as congenital and viral cardiomyopathies. Due to modern medical advancements, survival rates for pediatric congenital heart disease (CHD) patients have increased in recent years, resulting in more patients surviving into adulthood. Unfortunately however, a significant number of adult CHD patients have shown a dramatic increase in HF risk5, 6. Although current treatment options are useful in the management of HF, there remains an overwhelming need to develop a definitive modality to reverse HF and restore function of damaged tissue.

Pharmacological treatment

Depending on the symptoms that HF patients present, they may require only one or a combination of drugs. Currently available drugs target a symptom of the cardiovascular system, or a contributing factor to HF. Major classes of available drugs include: 1) drugs that help to reduce blood pressure by reducing afterload, such as angiotensin-converting enzyme (ACE) inhibitors and their alternative Angiotensin II receptor blockers, isosorbide dinitrate, beta-blockers, and hydralazine hydrochloride, 2) drugs that reduce blood volume and thus decrease cardiac workload, such as aldosterone antagonists and diuretics, and 3) drugs that increase cardiac contractility, improve cardiac pumping function and slow heart rate, such as digoxin7. Use of these drugs is helpful to prolong the life of HF patients improve cardiac function and reduce symptoms. However, none of these can efficiently reverse heart failure7.

Heart Transplant and Left Ventricular Assist Devices (LVAD)

Patients with severe HF require implantation of a left ventricular assist device (LVAD) or transplantation of an entire donor heart. The only definitive treatment for HF is heart transplantation—an option that is severely limited by the scarcity of donor hearts. From 1987 to 2012, 40,253 people were on a waiting list for a heart transplant, yet only 26,943 received a transplant8. The LVAD, a mechanical pump that is surgically implanted into the left ventricle of the failing heart, has been commonly used as a bridge to heart transplantation. Due to the lack of available donor hearts, the number of LVAD implantations has largely increased, which has improved survival rates of HF patients8. However, LVAD implantation has also resulted in increased costs, with a $234,808 mean cost of LVAD-related hospitalizations in 20118. Although current HF treatments such as drug therapies, LVAD implantation, and heart transplantation have value in improving heart function, hospitalized HF patients still experience very high post-discharge mortality and readmission rates, a number which has not decreased in the last twenty years1. Thus, there is an urgent need to develop new HF therapies.

Cell transplant treatment

In addition to clinical trials, considerable basic and translational research has been conducted with the aim of developing a cell transplantation-based treatment of heart failure. Several sources have been proposed for generating new cardiomyocyte cell sources, including embryonic stem cells (ESC), induced pluripotent stem cells (iPSC), mesenchymal stem cells, cardiac progenitor cells, and transdifferentiation of non-cardiomyocytes7, 912. Considerable effort has been made to overcome the roadblocks of cell transplant treatment. Tissue and genetic engineering techniques are complementary to cell-based therapy and are being developed with the goal of improving survival and integration of transplanted cells into native myocardium. Studies indicate that application of tissue-engineered cell sheet-based patches can increase the survival and engraftment of transplanted cells, help to improve cardiac function and help to prevent cardiac dilation of the infarcted heart13. However, after cell transplantation, engraftment and differentiation of administered cells into the native myocardium is very inefficient13. Recent innovative work indicate that cell-free delivery of extracellular vesicles, or exosomes, via patches to the injured heart improves cardiac recovery14, 15. Thus, patches offer a promising strategy for improving HF treatment. Direct reprogramming of non-cardiomyocytes such as cardiac fibroblasts, is another strategy to generate new cardiomyocytes to restore cardiac function during HF. In vitro and in vivo animal studies indicate that a variety of signaling cocktails can convert cardiac fibroblasts into induced cardiomyocyte-like cells, which helps to reduce the cardiac scarring and improve cardiac function recovery after infarction, yet there are still numerous barriers to overcome before any clinical application should be considered12.

Activation of endogenous cardiac repair

Recent reports have revealed that endogenous adult cardiomyocyte renewal is measurable, yet inefficient and incapable of adequately responding to extensive acute heart damage. Because of these limitations, therapies based on endogenous cardiomyocyte self-renewal are currently unavailable. It was previously thought that cardiomyocyte self-renewal is rare and is unlikely to be a useful method to treat HF. However, recent studies in animal models, such as zebrafish and mouse, have yielded mechanistic insights into the endogenous cardiac regeneration process, including findings concerning cardiomyocyte proliferation, inflammation, fibrosis, neovascularization, and the extracellular matrix16. These findings suggest that it is therapeutically possible to stimulate endogenous heart repair to treat HF following myocardial infarction. Importantly, recent discoveries indicate that inhibiting a critical regulatory genetic pathway called Hippo unleashes a powerful self-reparative capacity in the mammalian heart, and results in reversal of established HF1727. In addition, other signals and pathways such as IGF, Cyclin proteins, and microRNAs, have been reported to improve endogenous cardiac repair by stimulating cardiomyocyte proliferation in rodents following cardiac injury. We will specifically discuss these signals in more depth below. Although we have not reached the point that we can translate these animal model findings into clinical treatments, these exciting advances have uncovered new feasible therapeutic avenues for treating HF by inducing endogenous heart muscle to self-repair. A comparison of potential advantages of creating therapeutics to target endogenous cardiac repair is outlined in Table 1.

Table 1.

Comparison of Inducing Endogenous Repair to Other Potential Therapeutics

Treatment Type Advantages of Endogenous Repair Over Treatment
Current Pharmacological Therapeutics (ACE inhibitors, beta-blockers, diuretics, digoxin, etc.) Endogenous repair can lead to functional improvement and reversal of course of the disease.
Potential for fewer or shorter treatment period, reducing responsibility of patient compliance to medications
Left Ventricular Assist Device (LVAD) Placement LVAD placement requires major surgical intervention, which creates high cost due to hospitalization
LVADs are only rarely curative and often are only act as a bridge to transplant
Heart Transplantation Major shortage of implantable hearts compared to total HF patient population
Patients require immunomodulatory therapies to help slow possibility of organ rejection
Cell Transplantation Therapy Cell transplantation requires incorporation of new cardiomyocytes, both electrically and structurally, to prevent arrhythmia formation

The State of Cardiomyocyte Renewal in Mammals

During neonatal life, the mammalian heart undergoes a period of hyperplastic expansion followed by a stage of hypertrophic growth to accommodate the demand for cardiac output28. In rodents, studies show that neonatal cardiomyocytes undergo a 10% rate of DNA synthesis within the first 4 days of life, but by day 7, this rate quickly reduces down to 1% concomitant with terminal differentiation2933. To investigate the regenerative capacity of neonatal mice, multiple groups have employed various injury models: 1) surgical resection of the ventricular apex, 2) occlusion of the left anterior descending artery (LAD-O), and 3) cryo-infarction. Although informative and useful, these models present limitations. Determining regeneration in the apex resection model is dependent on resection size as neomyogenesis is limited in hearts that receive large resections and have thicker scarring34. Moreover, apex resection does not model physiological injury that the mammalian heart typically experiences, such as an MI. Importantly however, use of the apex resection model led researchers to the novel discovery that cardiac regeneration requires reinnervation of the heart35, 36. Although LAD-O is better suited to model MI, it is more technically demanding than apex resection and requires experience to have consistent infarcts. Cryo-infarction represents an intriguing model as this technique generates an injury and regenerative response similar to LAD-O37, yet can be more easily examined. Importantly however, the LAD-O injury models MI most accurately. Importantly, seminal work in mice revealed that hearts of neonatal mice can regenerate following apex resection or MI injury38, 39. Using these models, groups have reported various degrees of scar formation and regeneration, and the consensus agreement is that the regenerative window closes after day 7, whereby cardiomyocytes terminally differentiate33, 34, 37, 40, 41.

Adult cardiomyocytes are highly specialized, differentiated cells with very limited regenerative capacity, and until recent years, it was believed that cardiomyocytes were not capable of proliferation. However, in an influential study, researchers retroactively analyzed 14C isotope integration into cardiomyocytes of humans who lived during atmospheric nuclear bomb testing conducted from the early 1950s through 196342. Using carbon dating techniques and mathematical modeling, they calculated that human postnatal cardiomyocytes renew at a ~1% rate per year that decreases with age to ~0.3% (Fig. 1)42. Based on these calculations, they found that 40% of cardiomyocytes in the heart have been produced throughout a full life span, whereas the additional 60% are originally formed during prenatal development42, 43. These studies further reveal that hearts of humans 10 years or younger show 14C incorporation rates of ~5–6%, and this rate sharply declines to 1.9% between the ages of ten to twenty (Fig. 1)43.

Figure 1: Renewal rates of human cardiomyocytes.

Figure 1:

A) Timeline and rate calculated retrospectively using mathematical modeling and carbon dating measurements. B) Current theories describing the source of new 14C during aging: Left-labeled non-cardiomyoctyes fuse with cardiomyoctyes, leading to false-positive labeling. Middle-increased ploidy of cardiomyoctyes without cell division, Right-bona fide cardiomyoctye renewal. (Illustration Credit: Ben Smith).

Mounting evidence suggests that postnatally born cardiomyocytes are generated from pre-existing cardiomyocytes4448. Using thymidine-based DNA labeling and mitotic markers in mice, these studies indicate that a small percentage of mammalian adult cardiomyocytes are cycling29, 30, 49, 50. Both of these measuring techniques simply measure cell cycle re-entry, and do not measure completion of cell division. A group in 2014 devised an approach to directly measure cell replication and completion of cytokinesis in mice using a Cre-dependent GFP and RFP labeling system “mosaic analysis with double markers”51. Consistent with other data, they concluded that postnatal cardiomyocytes are born at very low rates, are cardiomyocyte-derived, and do not undergo a second round of division51. Immunofluorescence-based snap-shot profiling of M-phase and cytokinesis in cardiomyocytes from human donor hearts revealed a cardiomyocyte M-phase index of 0.04% from birth to 10 years of age which decreases to 0.009% during the next decade52. However, cardiomyocyte cell division indexes were less than 0.01% and 0.005% at these same respective stages52. It is clear that while adult cardiomyocytes are capable of cell division, these events naturally happen at very low rates that are insufficient to renew myocardium following injury.

Role of Ploidy in Cardiomyocyte Renewal

While the adult mammalian heart has little ability to regenerate following injury, this is not the case of all animal species. It is well established that both zebrafish and newt heart tissue can be repaired following injury through true cardiomyocyte division53, 54. Furthermore, the neonatal mouse heart also has a seven-day regenerative window following birth38. Within this timeframe, cardiomyocytes are able to successfully undergo cellular division and contribute to tissue renewal55. Both of these observations have led to a critical examination of the differences in cardiomyocyte cell biology across species. One interesting difference is the ploidy, or relative DNA content, of the cardiomyocytes. Zebrafish, newt, and neonatal mouse cardiomyocytes are all diploid (2n) cells, containing two sets of chromosomes, while adult mammalian cardiomyocytes exist largely as polyploid cells, containing four or more sets of chromosomes.

During mammalian development, cardiomyocytes are able to freely divide. However, following birth, the renewal capacity of the mammalian cardiomyocytes quickly dissipates56, 57. Instead, the cardiomyocytes exit the cell cycle prematurely. This can either happen following DNA synthesis, a process known as polyploidization, that results in a cardiomyocyte with a single tetraploid (4n) nucleus. The cardiomyocyte can also exit the cell cycle following mitosis but without undergoing cytokinesis, known as binucleation, resulting in a cardiomyocyte with two diploid (2n) nuclei. The relative abundance of polyploidization and binucleation differs between mammalian species, with a majority of adult human cardiomyocytes resulting from polyploidization and a majority of mouse cardiomyocytes resulting from binucleation5658. While the molecular mechanisms explaining these differences in cardiomyocyte cell cycle exit are not well understood, these differences demonstrate that there are potentially different mechanisms regulating cell cycle exit in different mammalian species. There is growing circumstantial evidence of an inverse correlation between cardiomyocyte ploidy and renewal potential. Strikingly, the mouse neonatal regenerative window highly correlates with increased cardiomyocyte ploidy. At birth, all mouse cardiomyocytes are diploid. However, by the end of the first week of life, around half of the cardiomyocytes have undergone binucleation, and cardiomyocyte division following injury also has greatly decreased50. There is also correlative evidence that the pool of diploid cardiomyocytes in the adult mouse are important contributors to the small amount of cardiomyocyte division seen after cardiac injury. For instance, it has been shown that cycling adult cardiomyocytes in the mouse marked by hypoxia fate mapping are more likely to be mononucleated59. Furthermore, mouse strains with greater numbers of diploid cardiomyocytes demonstrate greater functional recovery following myocardial infarction60.

While the evidence that diploid cardiomyocytes have greater regenerative potential in mouse is only correlative, recent work in zebrafish has been able to more directly address this hypothesis. Using a genetic model capable of increasing ploidy in zebrafish cardiomyocytes, it was found that diploid cardiomyocytes were significantly more likely to contribute myocardial renewal following injury61. Interestingly, converting 50% of zebrafish cardiomyocytes to a polyploidy state was sufficient to inhibit cardiac regeneration completely – this same percentage of binucleated cardiomyocytes is found at the end of the regenerative window in neonatal mice. Future work in mammalian systems investigating the role of polyploidy in the mammalian cardiomyocyte will not only improve our understanding of cardiomyocyte renewal but may also lead to new approaches to stimulate cardiogenesis therapeutically as the molecular underpinnings of this phenomenon are revealed.

Measuring True Cardiomyocyte Division

Awareness of polyploidization and binucleation cell cycle variants in cardiomyocytes is also critical when studying cardiac regeneration. While the ultimate goal of regeneration studies is to stimulate cardiomyocyte proliferation to contribute to damaged myocardium, measuring true cardiomyocyte division remains a technical challenge. This is due, in large part, to the fact that cardiomyocyte cell cycle re-entry does not necessarily indicate increased proliferation. In fact, cardiomyocyte ploidy frequently increases in heart failure, suggesting that cell cycle re-entry is an adaptive response to cardiac injury62. Therefore, it is critical that cell cycle assays are properly interpreted when reporting results in order to ensure reproducible conclusions. There are many cell cycle assays commonly used that only indicate cell cycle re-entry, with no indication whether cytokinesis will occur. Nucleotide analogs, such as EdU and BrdU, are incorporated during DNA synthesis and only indicates that the cell has progressed through S phase following analog administration. Histone H3 phosphorylation (pHH3) is a histone modification that occurs in G2 phase and continues to be present throughout mitosis and therefore relays no information regarding cytokinesis63. Aurora B kinase, which is the kinase responsible for Histone H3 phosphorylation, has the added benefit of being expressed throughout cytokinesis. However, Aurora B kinase is expressed in cardiomyocytes both undergoing binucleation and true cellular division, so its expression alone is not sufficient to identify definitive cardiomyocyte division events64.

While each of the previous markers are helpful for building evidence that cardiomyocytes are re-entering the cell cycle, they should be accompanied with more direct proof of cardiomyocyte proliferation. Stereology is a powerful technique for calculating total cardiomyocyte numbers in the heart by measuring cardiomyocyte number and size in successive tissue sections65. While other techniques exist for counting, such as performing counts after isolating cardiomyocytes through tissue dissociation, these often result in inconsistent results due to variation in digestion efficiency. Another useful technique is clonal analysis. There are multiple genetic models that have been developed in mice to perform these experiments, including the Brainbow and Mosaic Analysis with Double Markers (MADM) systems, both of which have been successfully used to demonstrate cardiomyocyte proliferation19, 51, 66, 67. Finally, recent work has indicated that midbody, a critical structural component of the contractile ring, location and distance between daughter nuclei may also be indicative of true cardiomyocyte division68. Time-lapse imaging experiments of acute heart slices demonstrated that the midbody, which contains both Aurora B kinase and Anillin, is expressed symmetrically between daughter nuclei if the cardiomyocyte will undergo cellular division and asymmetrically if binucleation will follow. Furthermore, daughter nuclei of cardiomyocytes that underwent division were much farther apart than daughter nuclei of cardiomyocytes undergoing binucleation. It is possible that adoption of these methods will be helpful in cardiac regeneration studies to more accurately measure cardiomyocyte cell division.

Molecular Signals that Stimulate Cardiomyocyte Proliferation

Given the sharp decline in cell cycle activity and loss of regenerative potential in adult mammalian hearts, considerable effort has been made to understand the cellular mechanism(s) that underlie cardiomyocyte cell division. A number of groups have determined that within the cardiomyocyte, signaling from growth factors, intrinsic pathways, microRNAs and cell cycle regulators stimulate cardiogenesis following injury (Fig. 2).

Figure 2: Molecular signals that stimulate cardiac repair.

Figure 2:

Exogenous signaling by growth factors, such as NRG/ERBB and IGF family members, activates intracellular pathways that promote cardiomyocyte (CM) proliferation and regeneration mechanisms. The Hippo kinase pathway is an intrinsic signaling pathway that triggers inhibitory phosphorylation of the CM proliferation protein Yap. This leads to either Yap sequestration at the plasma membrane with the dystrophin glycoprotein complex (DGC) or in the cytosol with 14–3-3 (dashed arrows). Treatment of CMs with agrin or anti-Hippo (shSalv) molecules promotes localization of dephosphorylated Yap to the nucleus to drive expression of CM renewal genes. MicroRNAs (miRs) can positively or negatively regulate CM proliferation, and development of anti-miR antagomiR molecules have innovated cardiac repair strategies. The CM cell cycle can be reactivated via modulation of regulators such as Cyclin A2 and member of the Cyclin B and Cyclin D families. Delivery of Wee1 and TGFb inihibitor molecules enhances cyclin/CDK-induced cardiac repair. CM proliferation: positive regulators (green), negative regulators (red). . (Illustration Credit: Ben Smith).

Cell Cycle Regulators

As neonatal life ends, cardiac expression of cell cycle regulators such as cyclins and cyclin-dependent kinases is largely silenced32. During the cell cycle, Cyclin A2 positively regulates G1/S and G2/M transitions, whereas Cyclin D2 drives G1/S progression. Overexpression studies in mice suggest that these genes promote proliferation of mature cardiomyocytes in adult hearts. Constitutive cardiac expression of Cyclin A2 in transgenic mice produces cardiomegaly due to the upregulation of cardiomyocyte proliferation69. Following ischemic injury, Cyclin A2 transgenic mice show less ventricular dilation and enhanced cardiac function, concomitant with cell cycle reentry in infarct and border zone myocardium70. Studies in rats and pigs revealed that viral delivery of Cyclin A2 to infarcted hearts was protective to ischemic injury71, 72. Likewise, transgenic overexpression of Cyclin D1 and D2 promotes cardiomyocyte cell cycle activity in infarcted hearts as evidenced by increased multinucleation, DNA synthesis, and proliferation73, 74. Recent investigation of the cardiomyocyte cell cycle reveals that combinatorial overexpression of cell-cycle regulators CDK1/CCNB/CDK4/CCND (referred to as 4F) activates proliferation of post-mitotic cardiomyocytes75. In this study, the authors used rigorous lineage tracing to determine that viral delivery of 4F induced cardiomyocyte proliferation, cell survival and functional recovery to infarcted hearts75. The replacement of CDK1/CCNB with Wee1 and TGFβ inhibitor molecules further enhanced cell-cycle re-entry of cardiomyocytes75, supporting the notion that cell cycle reactivation, if used in a safe manner, may be useful as a therapeutic strategy for cardiac repair.

Growth Factors

In addition to cell-cycle regulators, a growing body of signal transduction studies indicates that several classes of ligand/cell surface receptor complexes can exogenously induce cell cycle re-entry of cardiomyoctes and facilitate regeneration. The Neuregulin1 (NRG1) ligand is an epidermal growth factor that signals through ERBB2–4 tyrosine kinase receptors in the myocardium to regulate cardiac development (reviewed in76). Key regeneration studies reveal that NRG1/ERBB2/ERBB4 signaling promotes cardiomyoctye proliferation in the contexts of cardiac injury and homeostasis77, 78. Exogenous administration of NRG1 into adult mice was shown to stimulate cardiomyocyte proliferation and regeneration77. However, pediatric clinical trials and mice studies reveal that NRG1 delivery stimulates cardiomyocyte proliferation more effectively during a restricted period of early life79. In support of this, cardiac regeneration potential declines significantly after neonatal life due, at least in part, to reduced expression of ERBB280. Deletion of ERBB2 in embryonic or neonatal cardiomyocytes results in myocardial thinning marked by reduced cardiomyocyte numbers, whereas ERBB2 overexpression in neonate and adult mice produces cardiomegaly due to increased cardiomyocyte proliferation, de-differentiation and hypertrophy80. Moreover, transient induction of ERBB2 expression after LAD-O ischemic injury improves functional recovery and regeneration significantly80. These data indicate that increasing NRG1/ERBB activity is a potential avenue for enhancing cardiac regeneration. However, further investigation of NRG1/ERBB2/ERBB4 signaling is warranted, as little is known regarding NRG1 upstream regulators or downstream targets. Importantly however, the ERK, Akt and GSK3β/β-catenin pathways were found to be indispensable downstream mediators of ERBB2-induced cardiomyocyte de-differentiation, proliferation and hypertrophy80. Dual treatment of FGF1 and a p38 MAPK inhibitor (p38i) following myocardial infarction in rats led to increased cardiomyocyte mitoses81. Although cardiomyocyte proliferation is similarly increased, heart function is not rescued by p38i treatment alone81. Consistent with this finding, a 2016 clinical trial revealed that the p38 MAPK inhibitor losmapimod failed to improve the outcome of patients hospitalized after acute myocardial infarction82. Follow-up preclinical studies revealed that catheter delivery of FGF/NRG1-loaded microparticles to ischemia-reperfusion model swine hearts lead to increased myocardial vascularization and remodeling, accompanied by improved cardiac function83.

Epicardial growth factors, including follistatin-lilke 1 (FST1) and insulin growth factor 2 (IGF2) have also been shown to promote cardiomyocyte cell cycle re-entry. Epicardial patch delivery of FST1 to pig and mouse model infarcts promotes proliferation of pre-existing cardiomyocytes, leading to improved cardiac function and survival84. Notably, this effect was only observed when FST1 was epicardially-derived84. This specificity suggests that the origin of a signaling factor influences its capacity to trigger a response in cardiomyocytes. Similarly, IGF2 signaling in the epicardium during mouse development is required for ventricular cardiomyocyte proliferation84. Sustained co-administration of the growth factors insulin growth factor 1 (IGF1) and hepatocyte growth factor (HGF) improved cardiac function and stimulated cardiomyocyte production and capillarization to chronic MI pig hearts85. In addition, injection of growth hormone-releasing hormone (an activator of IGF1 signaling) into pig hearts two weeks after LAD-O injury led to significantly reduced scarring, yet cardiac function was not rescued86. Periostin, an extracellular matrix component, can also promote cardiomyocyte proliferation. Delivery of recombinant periostin via biodegradable Gelfoam extracellular scaffolds to rat hearts after myocardial infarction stimulates cell cycle re-entry of differentiated cardiomyocytes, reduces scaring and fibrosis, and improves cardiac function87. Delivery of recombinant periostin into the pericardial space of post-MI swine stimulated myocardial regeneration, improved cardiac function, and increased capillary density88. However, myocardial fibrosis was significantly increased in these hearts88, indicating that periostin treatment to direct cardiac repair in large mammals likely presents challenges (e.g. defective remodeling and arrhythmias).

Intrinsic Signaling

In recent years, investigators have identified a number of intrinsic cardiomyocyte signaling pathways that regulate cardiac regeneration. Among these pathways are those that execute crucial functions during heart development, cardiomyocyte maturation and postnatal life. Several groups have led the way to determine the transcriptional regulatory network(s) that regulate cardiomyocyte proliferation and regeneration. Recent cardiac regeneration studies identified the transcription factor Meis1 as a crucial regulator of postnatal cardiomyocyte proliferation89. In this work, the authors observed that deletion of Meis1 in cardiomyocytes after the regenerative phase of neonatal life extended the timeframe that cardiomyocytes normally cycle and regenerate89. In adult hearts, Meis1 deletion activated cardiomyocyte mitosis, without compromising cardiomyocyte function, whereas Meis1 overexpression was inhibitory to neonatal cardiac regeneration89. Given that the CDK inhibitors p15, p16, and p21 are Meis1 transcriptional activation targets89, these data collectively indicate that Meis1 is a negative regulator of cardiomyocyte proliferation. Using cryoinjury and apex resection models, investigators discovered that the cardiac transcription factor GATA4 is essential for cardiac regeneration by regulating the paracrine factor FGF1690. Viral overexpression of FGF16 partially rescued the injury-induced defects in GATA4-ablated hearts90, supporting the notion that paracrine factors can be targeted for cardiac repair. Overexpression of the transcription factor Tbx20 activates the YAP, BMP and AKT proliferative pathways, whereas the anti-proliferation genes p21 and Meis1 are repressed by Tbx2091. Importantly, Tbx20 overexpression in adult cardiomyocytes after MI reduced infarct size and markedly improved cardiac function and survival91.

The Hippo signaling pathway is an evolutionarily conserved tissue and organ size regulatory pathway first described in flies92. Core kinase components Mst/Hpo and Lats/Wts relay inhibitory phosphorylation of the downstream effector Yap/Yki, a transcriptional coactivator that promotes expression of survival and pro-growth genes92. In recent years, researchers have identified key Hippo roles in cardiac development, disease, cardiomyocyte homeostasis and regeneration. During development, Hippo signaling restrains cardiomyocyte proliferation to maintain proper heart size, while preserving overall tissue patterning93. Cardiac-specific deletion of the core Hippo component Salvador (Salv) during embryogenesis produced cardiomegaly due to hyper-proliferation of cardiomyocytes, and knockout of Mst and Lats kinases produced similar phenotypes93. Conversely, Yap deletion during cardiogenesis leads to embryonic lethality with hearts exhibiting pronounced myocardial thinning94, 95. More recent studies reveal that Hippo signaling represses a number of cellular mechanisms that are critical for endogenous heart repair. Several reports indicate that Hippo inhibits adult cardiac regeneration1727. In rodents, deletion of Hippo signaling components results in improved cardiac repair and reversal of established HF after myocardial infarction20. In this study, the authors also delivered a small molecule Hippo pathway inhibitor (shSalv) via an adeno-associated virus 9 (AAV9)20, a small virus shown to be safe in humans. AAV9 preferentially infects the heart muscle in mammalian species including mice, pigs, and humans and is a feasible choice for clinical use. Direct myocardial delivery of AAV9-shSalv during ischemic injury, or even weeks after injury, improves heart function20. These data support the possibility of using gene therapy to induce cardiomyocyte regeneration with minimal toxic side effects. Yap overexpression in mouse hearts after MI promoted cardiomyocyte proliferation and reduced infarct size, while preserving heart function19, 96. In addition, recent concurrent reports reveal that the Hippo effector Yap interacts with the dystrophin glycoprotein complex (DGC)21, 26 and the extracellular protein Agrin at the plasma membrane to regulate cardiomyocyte proliferation. One study revealed that Hippo phosphorylation of Yap triggers DGC sequestration of Yap at the plasma membrane in a mechanism to inhibit cardiomyocyte proliferation21. In another study, administration of Agrin to mice after MI promoted cardiac regeneration26. Biochemical assays revealed that Agrin promotes DGC disassembly, disruption of the Yap-DGC interaction and Yap translocation to promote cardiomyocyte proliferation26. Other studies revealed that injury-response genes are activated in regenerating Hippo-deficient hearts20, 23. Following MI, levels of the transcription factor Paired-like homeodomain-2 (Pitx2) are upregulated in Hippo-deficient myocardium23. In this context, Pitx2 and Yap co-regulate expression of antioxidant genes to protect the heart from injury-induced oxidative stress and promote regeneration23. Like Pitx2, the mitochondrial quality control stress response gene Park2 is upregulated in Hippo-deficient hearts and is required for regeneration20. These studies investigating Hippo signaling reveal that besides cardiomyocyte renewal, failing hearts have previously unappreciated cardiac repair genetic programs. Recent profiling of human patient hearts reveal that Hippo signaling is maladaptively upregulated in HF samples20. Altogether, these findings raise the possibility that endogenous cardiomyocytes can be mobilized by inhibiting the Hippo pathway, and that inhibiting Hippo can be used as a strategy to directly treat loss of pumping function in human HF patients.

MicroRNAs

MicroRNAs (miRs) are small non-coding RNAs, which repress gene expression, are required to maintain proper cardiomyocyte homeostasis. Disruption of the miR biogenesis protein Dicer in adult mouse myocardium leads to dilated cardiomyopathy and heart failure97, 98. Moreover, Dicer expression is reduced in end-stage heart failure patients, suggesting that DICER and miRs have important role(s) in cardiomyocyte progression and heart failure97. Indeed, human studies have shown that miR levels are altered in cardiac disease states99101. High-throughput screening of human miRs revealed that miR590 and miR199a promote cardiomyocyte cell cycle re-entry and promote cardiomyocyte proliferation in neonatal and adult mice102. Additional analyses showed that following myocardial infarction, co-delivery of these miRs stimulates cardiac regeneration and rescue of cardiac function102. Individual knockdown of these miRs did not upregulate cardiomyocyte proliferation102, suggesting functional redundancy between miR590 and miR199a targets. Mouse neonatal LAD-O studies revealed that miRs of the miR15 family inhibit cardiomyocyte proliferation39. Furthermore, inhibition of miR15 in adult hearts promotes cardiomyocyte proliferation and improves functional recovery after MI39. Therapeutic targeting of miR15 in mice and pigs using anti-miR oligonucleotides reduced infarct size following ischemia-reperfusion103. miR-17–92, an oncogene that promotes cell proliferation, is required for myocyte proliferation in the heart104. Transgenic miR-17–92 overexpression in mice hearts was protective to ischemic injury104. Cardiac function was improved, scarring was reduced, and border-zone cardiomyocyte proliferation was increased in these hearts104. Expression of miR-31a-5p is upregulated in neonatal rat cardiomyocytes during early regenerative phases105, suggesting a positive regulation of cardiomyocyte proliferation by miR-31a-5p. In support of this notion, cardiomyocyte proliferation marker levels were reduced following injection of neonatal rats with miR-31a-5p antagomiRs105. miR302–367 is essential for cardiomyocyte proliferation during development and overexpression of miR302–367 in the adult heart reduces fibrotic scarring after MI106. However, these hearts exhibited cardiomyocyte de-differentiation, ventricular dilation and reduced cardiac function106. Interestingly, miR302–367 targets the Mst and Lats kinases of the Hippo pathway106, suggesting that miR302–367 regulation of cardiomyocyte renewal and regeneration is mediated, at least in part, by Hippo inhibition. Taken together, these studies reveal that targeting miRs remains a promising therapeutic strategy to induce cardiac reparative processes.

Barriers to current treatments

To varying degrees, pharmacological intervention, LVAD implantation or heart transplantation, and cell transplantation treatments have significant limitations and new approaches are urgently required. Current pharmacological treatments mainly target symptoms or HF contributing factors to improve heart performance but cannot reverse the process of heart failure. Besides the severe scarcity of donor hearts, immune responses and organ rejection are common issues following heart transplantation. Efficacy of LVAD implantation varies in individual patients. Whereas some patients respond favorably to LVAD support and recover cardiac function, other patients do not respond to LVAD support and require heart transplantation. In addition, there are heavy health and financial burdens that come with both LVAD implantation and heart transplantation.

Cell transplantation has been wildly shown to improve cardiac function in cardiac injury animal models, but clinical trials have not revealed consistent benefits107. For example, in the REPAIR-AMI trial, autologous bone marrow-derived mononuclear cell (BMMNC) transplantation led to strong improvement trends of 5-year survival in patients with acute myocardial infarction107. However, there is no evidence of improved left ventricular function in any BMMNC clinical trial10. Observations and outcomes can vary in commonly used lab animal models like rodents, large mammals like swine and dog, and preclinical models like nonhuman primates. Implantation of ESC-derived cardiomyocytes do not cause arrhythmia in rodents but cause ventricular arrhythmia in primates with engrafted cells108. Therefore, variation of interspecies response to cell therapy is a critical issue to keep in mind and varying genetic backgrounds can affect interpreting experimental results. Hence, it is necessary to compare and validate data collected from different species before translating preclinical research to clinical trials. Moreover, these studies rely almost exclusively on ejection fraction as a prognostic measure107. In future studies, an obvious improvement over measuring ejection fraction would be to perform global longitudinal strain to assess newly described forms of HF109. One obvious common biological barrier to cell treatment is that the adult heart is continuously beating. As such, injected cells fail to colonize the diseased heart and are expelled into the circulatory system. In general, there are several challenges for cell transplantation treatments, such as poor engraftment and survival of implanted cells, limited contribution to cardiac contraction activity, immune responses, increased arrhythmogenic risks, risk of teratoma formation, and ethical considerations13.

What needs to be improved

As stated previously, the current therapies of pharmalogical intervention, LVAD placement, and heart transplantation all have drawbacks. Pharmalogical intervention treats HF symptoms and slows disease progression but is not capable of reversing the natural progression of the disease. LVAD implantation prolongs survival, but not all patients respond to treatment, and a majority still eventually require heart transplantation. While heart transplantation is the only curative treatment available, there are not enough donor hearts available for the large number of HF patients and it is a costly procedure. Clearly there is an urgent need for improved HF therapies. In recent years, the heart regeneration field has grown considerably, and has focused almost exclusively on stem cell-based treatments. Nonetheless, researchers have made steady progress towards inducing endogenous cardiomyocyte self-renewal. As a result, numerous animal model studies have shown that inducing endogenous cardiomyocytes to self-renew is a viable and undoubtedly exciting approach to generate new, healthy cardiomyocytes1727. Resident cardiomyocytes can be induced to re-enter cell cycle and proliferate to generate new cardiomyocytes, and a number of cellular signals have been identified as regulatory nodes for this newly unearthed phenomenon1727. Stimulating cardiomyocyte self-renewal through endogenous signaling is an appealing approach, as it would increase the volume of working myocardium after injury and would have the potential to reverse HF progression. Even small increases in heart function may have a large impact for HF patients, potentially prolonging survival and increasing quality of life, meaning combinatorial treatment plans with current standard of care plus novel therapies stimulating cardiomyocyte proliferation have the ability to make a large impact on the growing HF patient population.

Identification of modulators that target cardiomyocyte proliferation signaling pathways is imperative for the development of efficient cardiac repair therapies. Ideally, small molecule targeting of these pathways can be used to promote cardiomyocyte proliferation while minimizing cardiotoxicity. The Hippo-DGC-Agrin studies mentioned above suggest that viral delivery or direct injection of cardiomyocyte-proliferation agonists may be a viable cardiac repair strategy in humans. However, translation of technologies such as heart-specific viral vectors into human therapies requires thoughtful examination especially considering the oncogenic potential of activating growth pathways. In addition to intrinsic activation of cardiomyocyte proliferation pathways, administration of defined extracellular factors via epicardial patch delivery represents a complementary strategy to stimulate endogenous repair. Advantages of this approach include: 1) a cell-free system; reduces the likelihood of recipient cells rejecting delivered factors 2) delivery of peptides would cause transient effects, thereby minimizing the possibility of tumor formation and 3) newly produced cardiomyocytes would more likely incorporate into myocardium and function properly. Importantly however, path delivery can stimulate proliferation of non-cardiomyocytes. Similarly, current anti-MiR cardiac repair strategies lack cardiomyocyte specificity. Hence, innovations to the available patch and miR technologies are needed to overcome these barriers. Moreover, miR studies in non-human primates are warranted to demonstrate whether miR treatment might sufficiently repair cardiac damage in humans.

In addition to these improvements, there remain a number of basic science and clinical challenges to overcome to make stimulating cardiogenesis as a HF treatment strategy a reality (Table 2). Refined and rigorous cardiomyocyte proliferation assays should be developed to ascertain true cardiomyocyte proliferation during regeneration. In addition, the increased use of large animal models in conjunction with careful interpretation of the injury model used will facilitate the translation of HF treatment in human studies. As the clinical application of these findings are being developed, issues such as scalability, tissue-specificity, and integration of newly produced cardiomyocytes into post-injury myocardium need to be thoroughly addressed (Table 2).

Table 2.

Current Challenges for Stimulating Cardiogenesis as a Treatment for Heart Failure

Challenge Potential Solutions
Basic Science
Measuring True Cardiomyocyte Proliferation Practice of rigorous proliferation assays (stereology, lineage tracing, etc.) when measuring and reporting cardiomyocyte proliferation
Variation Among Animal Models Increase use of both large mammal models such as dog and swine at early stages of therapeutic development
Variation Among Cardiac Injury Models Interpretation of results should be specific to the cardiac insult performed and any limitations of injury technique explained
Translational
Scalability of Therapeutic to the Human Heart Increased testing in animal models with similar organ size, such as swine, to determine appropriate delivery techniques
Cell- and Tissue-Type Specificity Further research into epicardial patches, viral delivery modes (AAV), and direct myocardial injection techniques to ensure minimal off-target cellular proliferation / tumor formation
Integration of New Cardiomyocytes into Existing Myocardium (Electrical and Morphological) Extensive characterization of both electrical and structural incorporation of novel cardiomyocytes must be performed

Acknowledgements:

We wish to express our sincere gratitude to the Texas Legislature, both the House and Senate for financial support of THI’s Regenerative Medicine Program, for a philanthropic gift from Mr. Don McGill in support our Regenerative Medicine Program, and to the National Institutes of Health and the American Heart Association for continued research funding support.

Sources of Funding: Supported by the National Institutes of Health (HL 127717, HL 130804, and HL 118761 to J.F.M.; T32HL07676–28 to J.W.K.; R56HL142704, K01 DE026561, and R03DE025873 to J.W.), MacDonald Research Fund Award 16RDM001 (J.F.M.), and the Vivian L. Smith Foundation (J.F.M.). J.F.M. was supported by the LeDucq Foundations Transatlantic Networks of Excellence in Cardiovascular Research (14CVD01: Defining the Genomic Topology of Atrial Fibrillation). T.R.H. was supported by American Heart Association Scientist Development Grant (16SDG26460001). J.W. was supported by American Heart Association Scientist Development Grant 14SDG1984000.

Non-standard Abbreviations and Acronyms:

HF

heart failure

CHD

congenital heart disease

ACE

angiotensin-converting enzyme

LVAD

left ventricular assist device

ESC

embryonic stem cell

iPSC

induced pluripotent stem cell

IGF

insulin growth factor

LAD-O

left anterior descending coronary artery occlusion

MI

myocardial infarction

GFP

green fluorescent protein

RFP

red fluorescent protein

EdU

5-ethynyl-2´-deoxyuridine

BrdU

bromodeoxyuridine

pHH3

phosphohistone H3

MADM

mosaic analysis with double markers

4F

four factors

shSalv

short hairpin Salvador RNA

DGC

dystrophin glycoprotein complex

miR

microRNA

REPAIR-AMI

reinfusion of enriched progenitor cells and infarct remodeling in acute myocardial infarction

BMMNC

bone marrow-derived mononuclear cell

Footnotes

Disclosures: None

References

  • 1.Ambrosy AP, Fonarow GC, Butler J, Chioncel O, Greene SJ, Vaduganathan M, Nodari S, Lam CSP, Sato N, Shah AN and Gheorghiade M. The global health and economic burden of hospitalizations for heart failure: lessons learned from hospitalized heart failure registries. J Am Coll Cardiol. 2014;63:1123–1133. [DOI] [PubMed] [Google Scholar]
  • 2.Braunwald E The war against heart failure: the Lancet lecture. Lancet. 2015;385:812–24. [DOI] [PubMed] [Google Scholar]
  • 3.Yusuf S, Reddy S, Ounpuu S and Anand S. Global burden of cardiovascular diseases: Part II: variations in cardiovascular disease by specific ethnic groups and geographic regions and prevention strategies. Circulation. 2001;104:2855–64. [DOI] [PubMed] [Google Scholar]
  • 4.Lloyd-Jones D, Adams R, Carnethon M, De Simone G, Ferguson TB, Flegal K, Ford E, Furie K, Go A, Greenlund K, Haase N, Hailpern S, Ho M, Howard V, Kissela B, Kittner S, Lackland D, Lisabeth L, Marelli A, McDermott M, Meigs J, Mozaffarian D, Nichol G, O’Donnell C, Roger V, Rosamond W, Sacco R, Sorlie P, Stafford R, Steinberger J, Thom T, Wasserthiel-Smoller S, Wong N, Wylie-Rosett J, Hong Y, American Heart Association Statistics C and Stroke Statistics S. Heart disease and stroke statistics−-2009 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation. 2009;119:480–6. [DOI] [PubMed] [Google Scholar]
  • 5.Ntiloudi D, Giannakoulas G, Parcharidou D, Panagiotidis T, Gatzoulis MA and Karvounis H. Adult congenital heart disease: A paradigm of epidemiological change. International journal of cardiology. 2016;218:269–274. [DOI] [PubMed] [Google Scholar]
  • 6.Gurvitz M, Burns KM, Brindis R, Broberg CS, Daniels CJ, Fuller SM, Honein MA, Khairy P, Kuehl KS, Landzberg MJ, Mahle WT, Mann DL, Marelli A, Newburger JW, Pearson GD, Starling RC, Tringali GR, Valente AM, Wu JC and Califf RM. Emerging Research Directions in Adult Congenital Heart Disease: A Report From an NHLBI/ACHA Working Group. J Am Coll Cardiol. 2016;67:1956–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Leach JP and Martin JF. Cardiomyocyte Proliferation for Therapeutic Regeneration. Curr Cardiol Rep. 2018;20:63. [DOI] [PubMed] [Google Scholar]
  • 8.Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, de Ferranti SD, Floyd J, Fornage M, Gillespie C, Isasi CR, Jimenez MC, Jordan LC, Judd SE, Lackland D, Lichtman JH, Lisabeth L, Liu S, Longenecker CT, Mackey RH, Matsushita K, Mozaffarian D, Mussolino ME, Nasir K, Neumar RW, Palaniappan L, Pandey DK, Thiagarajan RR, Reeves MJ, Ritchey M, Rodriguez CJ, Roth GA, Rosamond WD, Sasson C, Towfighi A, Tsao CW, Turner MB, Virani SS, Voeks JH, Willey JZ, Wilkins JT, Wu JH, Alger HM, Wong SS, Muntner P, American Heart Association Statistics C and Stroke Statistics S. Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association. Circulation. 2017;135:e146–e603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cahill TJ, Choudhury RP and Riley PR. Heart regeneration and repair after myocardial infarction: translational opportunities for novel therapeutics. Nature reviews Drug discovery. 2017;16:699–717. [DOI] [PubMed] [Google Scholar]
  • 10.Telukuntla KS, Suncion VY, Schulman IH and Hare JM. The advancing field of cell-based therapy: insights and lessons from clinical trials. J Am Heart Assoc. 2013;2:e000338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Tang JN, Cores J, Huang K, Cui XL, Luo L, Zhang JY, Li TS, Qian L and Cheng K. Concise Review: Is Cardiac Cell Therapy Dead? Embarrassing Trial Outcomes and New Directions for the Future. Stem Cells Transl Med. 2018;7:354–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Vaseghi H, Liu J and Qian L. Molecular barriers to direct cardiac reprogramming. Protein Cell. 2017;8:724–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Yanamandala M, Zhu W, Garry DJ, Kamp TJ, Hare JM, Jun HW, Yoon YS, Bursac N, Prabhu SD, Dorn GW 2nd, Bolli R, Kitsis RN and Zhang J Overcoming the Roadblocks to Cardiac Cell Therapy Using Tissue Engineering. J Am Coll Cardiol. 2017;70:766–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Heallen TR and Martin JF. Heart repair via cardiomyocyte-secreted vesicles. Nature Biomedical Engineering. 2018;2:271–272. [DOI] [PubMed] [Google Scholar]
  • 15.Liu B Cardiac recovery via extended cell-free delivery of extracellular vesicles secreted by cardiomyocytes derived from induced pluripotent stem cells. Nat Biomed Eng. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cahill TJ, Choudhury RP and Riley PR. Heart regeneration and repair after myocardial infarction: translational opportunities for novel therapeutics. Nat Rev Drug Discov. 2017;16:699–717. [DOI] [PubMed] [Google Scholar]
  • 17.Heallen T, Morikawa Y, Leach J, Tao G, Willerson JT, Johnson RL and Martin JF. Hippo signaling impedes adult heart regeneration. Development. 2013;140:4683–4690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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 and Olson EN. Hippo pathway effector Yap promotes cardiac regeneration. Proc Natl Acad Sci U S A. 2013;110:13839–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lin Z, von Gise A, Zhou P, Gu F, Ma Q, Jiang J, Yau AL, Buck JN, Gouin KA, van Gorp PR, Zhou B, Chen J, Seidman JG, Wang DZ and Pu WT. Cardiac-specific YAP activation improves cardiac function and survival in an experimental murine MI model. Circulation research. 2014;115:354–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Leach JP, Heallen T, Zhang M, Rahmani M, Morikawa Y, Hill MC, Segura A, Willerson JT and Martin JF. Hippo pathway deficiency reverses systolic heart failure after infarction. Nature. 2017;550:260–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Morikawa Y, Heallen T, Leach J, Xiao Y and Martin JF. Dystrophin-glycoprotein complex sequesters Yap to inhibit cardiomyocyte proliferation. Nature. 2017;547:227–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Morikawa Y, Zhang M, Heallen T, Leach J, Tao G, Xiao Y, Bai Y, Li W, Willerson JT and Martin JF. Actin cytoskeletal remodeling with protrusion formation is essential for heart regeneration in Hippo-deficient mice. Science signaling. 2015;8:ra41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tao G, Kahr PC, Morikawa Y, Zhang M, Rahmani M, Heallen TR, Li L, Sun Z, Olson EN, Amendt BA and Martin JF. Pitx2 promotes heart repair by activating the antioxidant response after cardiac injury. Nature. 2016;534:119–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wang J, Liu S, Heallen T and Martin JF. The Hippo pathway in the heart: pivotal roles in development, disease, and regeneration. Nat Rev Cardiol. 2018. [DOI] [PubMed] [Google Scholar]
  • 25.Xiao Y, Leach J, Wang J and Martin JF. Hippo/Yap Signaling in Cardiac Development and Regeneration. Curr Treat Options Cardiovasc Med. 2016;18:38. [DOI] [PubMed] [Google Scholar]
  • 26.Bassat E, Mutlak YE, Genzelinakh A, Shadrin IY, Baruch Umansky K, Yifa O, Kain D, Rajchman D, Leach J, Riabov Bassat D, Udi Y, Sarig R, Sagi I, Martin JF, Bursac N, Cohen S and Tzahor E. The extracellular matrix protein agrin promotes heart regeneration in mice. Nature. 2017;547:179–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wang J and Martin JF. Macro advances in microRNAs and myocardial regeneration. Curr Opin Cardiol. 2014;29:207–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Heineke J and Molkentin JD. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat Rev Mol Cell Biol. 2006;7:589–600. [DOI] [PubMed] [Google Scholar]
  • 29.Soonpaa MH and Field LJ. Survey of studies examining mammalian cardiomyocyte DNA synthesis. Circulation research. 1998;83:15–26. [DOI] [PubMed] [Google Scholar]
  • 30.Walsh S, Ponten A, Fleischmann BK and Jovinge S. Cardiomyocyte cell cycle control and growth estimation in vivo--an analysis based on cardiomyocyte nuclei. Cardiovasc Res. 2010;86:365–73. [DOI] [PubMed] [Google Scholar]
  • 31.Bergmann O, Zdunek S, Alkass K, Druid H, Bernard S and Frisén J. Identification of cardiomyocyte nuclei and assessment of ploidy for the analysis of cell turnover. Experimental Cell Research. 2011;317:188–194. [DOI] [PubMed] [Google Scholar]
  • 32.Ahuja P, Sdek P and MacLellan RW. Cardiac myocyte cell cycle control in development, disease, and regeneration. Physiological reviews. 2007;87:521–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, Olson EN and Sadek HA. Transient Regenerative Potential of the Neonatal Mouse Heart. Science. 2011;331:1078–1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bryant D, O’Meara C, Ho N, Gannon J, Cai L and Lee R. A systematic analysis of neonatal mouse heart regeneration after apical resection. Journal of Molecular and Cellular Cardiology. 2015;79:315–318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.White IA, Gordon J, Balkan W and Hare JM. Sympathetic Reinnervation Is Required for Mammalian Cardiac Regeneration. Circulation research. 2015;117:990–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Mahmoud AI, O’Meara CC, Gemberling M, Zhao L, Bryant DM, Zheng R, Gannon JB, Cai L, Choi WY, Egnaczyk GF, Burns CE, Burns CG, MacRae CA, Poss KD and Lee RT. Nerves Regulate Cardiomyocyte Proliferation and Heart Regeneration. Developmental cell. 2015;34:387–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Strungs EG, Ongstad EL, O’Quinn MP, Palatinus JA, Jourdan LJ and Gourdie RG. Cryoinjury models of the adult and neonatal mouse heart for studies of scarring and regeneration. Methods in molecular biology (Clifton, NJ). 2013;1037:343–53. [DOI] [PubMed] [Google Scholar]
  • 38.Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, Olson EN and Sadek HA. Transient regenerative potential of the neonatal mouse heart. Science. 2011;331:1078–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Porrello ER, Mahmoud AI, Simpson E, Johnson BA, Grinsfelder D, Canseco D, Mammen PP, Rothermel BA, Olson EN and Sadek HA. Regulation of neonatal and adult mammalian heart regeneration by the miR-15 family. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:187–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Jesty SA, Steffey MA, Lee FK, Breitbach M, Hesse M, Reining S, Lee JC, Doran RM, Nikitin A, Fleischmann BK and Kotlikoff MI. c-kit+ precursors support postinfarction myogenesis in the neonatal, but not adult, heart. Proceedings of the National Academy of Sciences. 2012;109:13380–13385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Haubner B, Adamowicz-Brice M, Khadayate S, Tiefenthaler V, Metzler B, Aitman T and Penninger JM. Complete cardiac regeneration in a mouse model of myocardial infarction. Aging. 2012;4:966–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabé-Heider F, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, Jovinge S and Frisén J. Evidence for Cardiomyocyte Renewal in Humans. Science. 2009;324:98–102. [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, Andrä M, Jashari R, Nyengaard JR, Possnert G, Jovinge S, Druid H and Frisén J Dynamics of Cell Generation and Turnover in the Human Heart. Cell. 2015;161:1566–1575. [DOI] [PubMed] [Google Scholar]
  • 44.Cai C-L and Molkentin JD. The Elusive Progenitor Cell in Cardiac Regeneration. Circulation research. 2017;120:400–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Eschenhagen T, Bolli R, Braun T, Field LJ, Fleischmann BK, Frisén J, Giacca M, Hare JM, Houser S, Lee RT, Marbán E, Martin JF, Molkentin JD, Murry CE, Riley PR, Ruiz-Lozano P, Sadek HA, Sussman MA and Hill JA. Cardiomyocyte Regeneration. Circulation. 2017;136:680–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Li Y, He L, Huang X, Bhaloo S, Zhao H, Zhang S, Pu W, Tian X, Li Y, Liu Q, Yu W, Zhang L, Liu X, Liu K, Tang J, Zhang H, Cai D, Ralf AH, Xu Q, Lui KO and Zhou B. Genetic Lineage Tracing of Nonmyocyte Population by Dual Recombinases. Circulation. 2018;138:793–805. [DOI] [PubMed] [Google Scholar]
  • 47.He L, Li Y, Li Y, Pu W, Huang X, Tian X, Wang Y, Zhang H, Liu Q, Zhang L, Zhao H, Tang J, Ji H, Cai D, Han Z, Han Z, Nie Y, Hu S, Wang QD, Sun R, Fei J, Wang F, Chen T, Yan Y, Huang H, Pu WT and Zhou B. Enhancing the precision of genetic lineage tracing using dual recombinases. Nat Med. 2017;23:1488–1498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ali SR, Hippenmeyer S, Saadat LV, Luo L, Weissman IL and Ardehali R. Existing cardiomyocytes generate cardiomyocytes at a low rate after birth in mice. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:8850–8855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Senyo SE, Steinhauser ML, Pizzimenti CL, Yang VK, Cai L, Wang M, Wu T-D, Guerquin-Kern J-L, Lechene CP and Lee RT. Mammalian heart renewal by pre-existing cardiomyocytes. Nature. 2012;493:433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Soonpaa MH, Kim KK, Pajak L, Franklin M and Field LJ. Cardiomyocyte DNA synthesis and binucleation during murine development. Am J Physiol. 1996;271:H2183–9. [DOI] [PubMed] [Google Scholar]
  • 51.Ali SR, Hippenmeyer S, Saadat LV, Luo L, Weissman IL and Ardehali R. Existing cardiomyocytes generate cardiomyocytes at a low rate after birth in mice. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:8850–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mollova M, Bersell K, Walsh S, Savla J, Das L, Park S-Y, Silberstein LE, dos Remedios CG, Graham D, Colan S and Kühn B. Cardiomyocyte proliferation contributes to heart growth in young humans. Proceedings of the National Academy of Sciences. 2013;110:1446–1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Poss KD, Wilson LG and Keating MT. Heart regeneration in zebrafish. Science. 2002;298:2188–90. [DOI] [PubMed] [Google Scholar]
  • 54.Oberpriller JO and Oberpriller JC. Response of the adult newt ventricle to injury. J Exp Zool. 1974;187:249–53. [DOI] [PubMed] [Google Scholar]
  • 55.Senyo SE, Steinhauser ML, Pizzimenti CL, Yang VK, Cai L, Wang M, Wu TD, Guerquin-Kern JL, Lechene CP and Lee RT. Mammalian heart renewal by pre-existing cardiomyocytes. Nature. 2013;493:433–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Alkass K, Panula J, Westman M, Wu TD, Guerquin-Kern JL and Bergmann O. No Evidence for Cardiomyocyte Number Expansion in Preadolescent Mice. Cell. 2015;163:1026–36. [DOI] [PubMed] [Google Scholar]
  • 57.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 and Frisen J. Dynamics of Cell Generation and Turnover in the Human Heart. Cell. 2015;161:1566–75. [DOI] [PubMed] [Google Scholar]
  • 58.Mollova M, Bersell K, Walsh S, Savla J, Das LT, Park SY, Silberstein LE, Dos Remedios CG, Graham D, Colan S and Kuhn B. Cardiomyocyte proliferation contributes to heart growth in young humans. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:1446–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.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 and Sadek HA. Hypoxia fate mapping identifies cycling cardiomyocytes in the adult heart. Nature. 2015;523:226–30. [DOI] [PubMed] [Google Scholar]
  • 60.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 and Sucov HM. Frequency of mononuclear diploid cardiomyocytes underlies natural variation in heart regeneration. Nat Genet. 2017;49:1346–1353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Gonzalez-Rosa JM, Sharpe M, Field D, Soonpaa MH, Field LJ, Burns CE and Burns CG. Myocardial Polyploidization Creates a Barrier to Heart Regeneration in Zebrafish. Developmental cell. 2018;44:433–446 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Herget GW, Neuburger M, Plagwitz R and Adler CP. DNA content, ploidy level and number of nuclei in the human heart after myocardial infarction. Cardiovasc Res. 1997;36:45–51. [DOI] [PubMed] [Google Scholar]
  • 63.Crosio C, Fimia GM, Loury R, Kimura M, Okano Y, Zhou H, Sen S, Allis CD and Sassone-Corsi P. Mitotic phosphorylation of histone H3: spatio-temporal regulation by mammalian Aurora kinases. Mol Cell Biol. 2002;22:874–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Engel FB, Schebesta M and Keating MT. Anillin localization defect in cardiomyocyte binucleation. J Mol Cell Cardiol. 2006;41:601–12. [DOI] [PubMed] [Google Scholar]
  • 65.Bruel A and Nyengaard JR. Design-based stereological estimation of the total number of cardiac myocytes in histological sections. Basic Res Cardiol. 2005;100:311–9. [DOI] [PubMed] [Google Scholar]
  • 66.Livet J, Weissman TA, Kang H, Draft RW, Lu J, Bennis RA, Sanes JR and Lichtman JW. Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system. Nature. 2007;450:56–62. [DOI] [PubMed] [Google Scholar]
  • 67.Zong H, Espinosa JS, Su HH, Muzumdar MD and Luo L. Mosaic analysis with double markers in mice. Cell. 2005;121:479–92. [DOI] [PubMed] [Google Scholar]
  • 68.Hesse M, Doengi M, Becker A, Kimura K, Voeltz N, Stein V and Fleischmann BK. Midbody Positioning and Distance Between Daughter Nuclei Enable Unequivocal Identification of Cardiomyocyte Cell Division in Mice. Circulation research. 2018;123:1039–1052. [DOI] [PubMed] [Google Scholar]
  • 69.Chaudhry HW, Dashoush NH, Tang H, Zhang L, Wang X, Wu EX and Wolgemuth DJ. Cyclin A2 mediates cardiomyocyte mitosis in the postmitotic myocardium. The Journal of biological chemistry. 2004;279:35858–66. [DOI] [PubMed] [Google Scholar]
  • 70.Cheng RK, Asai T, Tang H, Dashoush NH, Kara RJ, Costa KD, Naka Y, Wu EX, Wolgemuth DJ and Chaudhry HW. Cyclin A2 induces cardiac regeneration after myocardial infarction and prevents heart failure. Circulation research. 2007;100:1741–8. [DOI] [PubMed] [Google Scholar]
  • 71.Shapiro SD, Ranjan AK, Kawase Y, Cheng RK, Kara RJ, Bhattacharya R, Guzman-Martinez G, Sanz J, Garcia MJ and Chaudhry HW. Cyclin A2 induces cardiac regeneration after myocardial infarction through cytokinesis of adult cardiomyocytes. Sci Transl Med. 2014;6:224ra27. [DOI] [PubMed] [Google Scholar]
  • 72.Woo YJ, Panlilio CM, Cheng RK, Liao GP, Atluri P, Hsu VM, Cohen JE and Chaudhry HW. Therapeutic delivery of cyclin A2 induces myocardial regeneration and enhances cardiac function in ischemic heart failure. Circulation. 2006;114:I206–13. [DOI] [PubMed] [Google Scholar]
  • 73.Pasumarthi KB, Nakajima H, Nakajima HO, Soonpaa MH and Field LJ. Targeted expression of cyclin D2 results in cardiomyocyte DNA synthesis and infarct regression in transgenic mice. Circulation research. 2005;96:110–8. [DOI] [PubMed] [Google Scholar]
  • 74.Soonpaa MH, Koh GY, Pajak L, Jing S, Wang H, Franklin MT, Kim KK and Field LJ. Cyclin D1 overexpression promotes cardiomyocyte DNA synthesis and multinucleation in transgenic mice. J Clin Invest. 1997;99:2644–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mohamed TMA, Ang YS, Radzinsky E, Zhou P, Huang Y, Elfenbein A, Foley A, Magnitsky S and Srivastava D. Regulation of Cell Cycle to Stimulate Adult Cardiomyocyte Proliferation and Cardiac Regeneration. Cell. 2018;173:104–116 e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Citri A and Yarden Y. EGF-ERBB signalling: towards the systems level. Nat Rev Mol Cell Biol. 2006;7:505–16. [DOI] [PubMed] [Google Scholar]
  • 77.Bersell K, Arab S, Haring B and Kuhn B. Neuregulin1/ErbB4 signaling induces cardiomyocyte proliferation and repair of heart injury. Cell. 2009;138:257–70. [DOI] [PubMed] [Google Scholar]
  • 78.Wadugu B and Kuhn B. The role of neuregulin/ErbB2/ErbB4 signaling in the heart with special focus on effects on cardiomyocyte proliferation. Am J Physiol Heart Circ Physiol. 2012;302:H2139–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Polizzotti BD, Ganapathy B, Walsh S, Choudhury S, Ammanamanchi N, Bennett DG, dos Remedios CG, Haubner BJ, Penninger JM and Kuhn B. Neuregulin stimulation of cardiomyocyte regeneration in mice and human myocardium reveals a therapeutic window. Sci Transl Med. 2015;7:281ra45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.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 and Tzahor E. ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation. Nature cell biology. 2015;17:627–38. [DOI] [PubMed] [Google Scholar]
  • 81.Engel FB, Hsieh PC, Lee RT and Keating MT. FGF1/p38 MAP kinase inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and rescues function after myocardial infarction. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:15546–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.O’Donoghue ML, Glaser R, Cavender MA, Aylward PE, Bonaca MP, Budaj A, Davies RY, Dellborg M, Fox KA, Gutierrez JA, Hamm C, Kiss RG, Kovar F, Kuder JF, Im KA, Lepore JJ, Lopez-Sendon JL, Ophuis TO, Parkhomenko A, Shannon JB, Spinar J, Tanguay JF, Ruda M, Steg PG, Theroux P, Wiviott SD, Laws I, Sabatine MS, Morrow DA and Investigators L-T. Effect of Losmapimod on Cardiovascular Outcomes in Patients Hospitalized With Acute Myocardial Infarction: A Randomized Clinical Trial. Jama. 2016;315:1591–9. [DOI] [PubMed] [Google Scholar]
  • 83.Garbayo E, Gavira JJ, de Yebenes MG, Pelacho B, Abizanda G, Lana H, Blanco-Prieto MJ and Prosper F. Catheter-based Intramyocardial Injection of FGF1 or NRG1-loaded MPs Improves Cardiac Function in a Preclinical Model of Ischemia-Reperfusion. Sci Rep. 2016;6:25932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.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 and Ruiz-Lozano P. Epicardial FSTL1 reconstitution regenerates the adult mammalian heart. Nature. 2015;525:479–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Koudstaal S, Bastings MM, Feyen DA, Waring CD, van Slochteren FJ, Dankers PY, Torella D, Sluijter JP, Nadal-Ginard B, Doevendans PA, Ellison GM and 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. 2014;7:232–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.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 and Hare JM. Growth hormone-releasing hormone agonists reduce myocardial infarct scar in swine with subacute ischemic cardiomyopathy. J Am Heart Assoc. 2015;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kuhn B, del Monte F, Hajjar RJ, Chang YS, Lebeche D, Arab S and Keating MT. Periostin induces proliferation of differentiated cardiomyocytes and promotes cardiac repair. Nat Med. 2007;13:962–9. [DOI] [PubMed] [Google Scholar]
  • 88.Ladage D, Yaniz-Galende E, Rapti K, Ishikawa K, Tilemann L, Shapiro S, Takewa Y, Muller-Ehmsen J, Schwarz M, Garcia MJ, Sanz J, Hajjar RJ and Kawase Y. Stimulating myocardial regeneration with periostin Peptide in large mammals improves function post-myocardial infarction but increases myocardial fibrosis. PloS one. 2013;8:e59656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Mahmoud AI, Kocabas F, Muralidhar SA, Kimura W, Koura AS, Thet S, Porrello ER and Sadek HA. Meis1 regulates postnatal cardiomyocyte cell cycle arrest. Nature. 2013;497:249–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Yu W, Huang X, Tian X, Zhang H, He L, Wang Y, Nie Y, Hu S, Lin Z, Zhou B, Pu W, Lui KO and Zhou B. GATA4 regulates Fgf16 to promote heart repair after injury. Development. 2016;143:936–49. [DOI] [PubMed] [Google Scholar]
  • 91.Xiang FL, Guo M and Yutzey KE. Overexpression of Tbx20 in Adult Cardiomyocytes Promotes Proliferation and Improves Cardiac Function After Myocardial Infarction. Circulation. 2016;133:1081–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Halder G and Johnson RL. Hippo signaling: growth control and beyond. Development. 2011;138:9–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Heallen T, Zhang M, Wang J, Bonilla-Claudio M, Klysik E, Johnson RL and Martin JF. Hippo pathway inhibits Wnt signaling to restrain cardiomyocyte proliferation and heart size. Science. 2011;332:458–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.von Gise A, Lin Z, Schlegelmilch K, Honor LB, Pan GM, Buck JN, Ma Q, Ishiwata T, Zhou B, Camargo FD and Pu WT. YAP1, the nuclear target of Hippo signaling, stimulates heart growth through cardiomyocyte proliferation but not hypertrophy. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:2394–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Xin M, Kim Y, Sutherland LB, Qi X, McAnally J, Schwartz RJ, Richardson JA, Bassel-Duby R and Olson EN. Regulation of insulin-like growth factor signaling by Yap governs cardiomyocyte proliferation and embryonic heart size. Science signaling. 2011;4:ra70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.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 and Olson EN. Hippo pathway effector Yap promotes cardiac regeneration. Proceedings of the National Academy of Sciences of the United States of America. 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Chen JF, Murchison EP, Tang R, Callis TE, Tatsuguchi M, Deng Z, Rojas M, Hammond SM, Schneider MD, Selzman CH, Meissner G, Patterson C, Hannon GJ and Wang DZ. Targeted deletion of Dicer in the heart leads to dilated cardiomyopathy and heart failure. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:2111–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Rao PK, Toyama Y, Chiang HR, Gupta S, Bauer M, Medvid R, Reinhardt F, Liao R, Krieger M, Jaenisch R, Lodish HF and Blelloch R. Loss of cardiac microRNA-mediated regulation leads to dilated cardiomyopathy and heart failure. Circulation research. 2009;105:585–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Thum T, Gross C, Fiedler J, Fischer T, Kissler S, Bussen M, Galuppo P, Just S, Rottbauer W, Frantz S, Castoldi M, Soutschek J, Koteliansky V, Rosenwald A, Basson MA, Licht JD, Pena JT, Rouhanifard SH, Muckenthaler MU, Tuschl T, Martin GR, Bauersachs J and Engelhardt S. MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature. 2008;456:980–4. [DOI] [PubMed] [Google Scholar]
  • 100.Ikeda S, Kong SW, Lu J, Bisping E, Zhang H, Allen PD, Golub TR, Pieske B and Pu WT. Altered microRNA expression in human heart disease. Physiol Genomics. 2007;31:367–73. [DOI] [PubMed] [Google Scholar]
  • 101.Matkovich SJ, Van Booven DJ, Youker KA, Torre-Amione G, Diwan A, Eschenbacher WH, Dorn LE, Watson MA, Margulies KB and Dorn GW 2nd. Reciprocal regulation of myocardial microRNAs and messenger RNA in human cardiomyopathy and reversal of the microRNA signature by biomechanical support. Circulation. 2009;119:1263–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Eulalio A, Mano M, Dal Ferro M, Zentilin L, Sinagra G, Zacchigna S and Giacca M. Functional screening identifies miRNAs inducing cardiac regeneration. Nature. 2012;492:376–81. [DOI] [PubMed] [Google Scholar]
  • 103.Hullinger TG, Montgomery RL, Seto AG, Dickinson BA, Semus HM, Lynch JM, Dalby CM, Robinson K, Stack C, Latimer PA, Hare JM, Olson EN and van Rooij E. Inhibition of miR-15 protects against cardiac ischemic injury. Circulation research. 2012;110:71–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Chen J, Huang ZP, Seok HY, Ding J, Kataoka M, Zhang Z, Hu X, Wang G, Lin Z, Wang S, Pu WT, Liao R and Wang DZ. mir-17–92 cluster is required for and sufficient to induce cardiomyocyte proliferation in postnatal and adult hearts. Circulation research. 2013;112:1557–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Xiao J, Liu H, Cretoiu D, Toader DO, Suciu N, Shi J, Shen S, Bei Y, Sluijter JP, Das S, Kong X and Li X. miR-31a-5p promotes postnatal cardiomyocyte proliferation by targeting RhoBTB1. Exp Mol Med. 2017;49:e386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Tian Y, Liu Y, Wang T, Zhou N, Kong J, Chen L, Snitow M, Morley M, Li D, Petrenko N, Zhou S, Lu M, Gao E, Koch WJ, Stewart KM and Morrisey EE. A microRNA-Hippo pathway that promotes cardiomyocyte proliferation and cardiac regeneration in mice. Sci Transl Med. 2015;7:279ra38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Assmus B, Leistner DM, Schachinger V, Erbs S, Elsasser A, Haberbosch W, Hambrecht R, Sedding D, Yu J, Corti R, Mathey DG, Barth C, Mayer-Wehrstein C, Burck I, Sueselbeck T, Dill T, Hamm CW, Tonn T, Dimmeler S, Zeiher AM and Group R-AS. Long-term clinical outcome after intracoronary application of bone marrow-derived mononuclear cells for acute myocardial infarction: migratory capacity of administered cells determines event-free survival. Eur Heart J. 2014;35:1275–83. [DOI] [PubMed] [Google Scholar]
  • 108.Chong JJ, Yang X, 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 and Murry CE. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature. 2014;510:273–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Marwick TH. Ejection Fraction Pros and Cons: JACC State-of-the-Art Review. J Am Coll Cardiol. 2018;72:2360–2379. [DOI] [PubMed] [Google Scholar]

RESOURCES