Abstract
Ischemic cardiac injury causes irreversible muscle loss and scarring, but recent years have seen dramatic advances in cardiac reprogramming, the field focused on regenerating cardiac muscle. With SARS-CoV2 increasing the age-adjusted cardiovascular disease mortality rate, it is worth evaluating the state of this field. Here, we summarize novel innovations in reprogramming strategies, insights into their mechanisms, and technologies for factor delivery. We also propose a broad model of reprogramming to suggest directions for future research. Poet Emily Dickinson wrote, “If I can stop one heart from breaking, I shall not live in vain.” Today, researchers studying cardiac reprogramming view this line as a call to action to translate this revolutionary approach into life-saving treatments for patients with cardiovascular diseases.
Keywords: Cardiac reprogramming, direct reprogramming, ischemic heart disease, regenerative medicine, cardiomyocytes, transdifferentiation, cell fate, cardiovascular biology, chromatin remodeling, cell signaling, metabolic reprogramming
Introduction
A 2023 update to the American Heart Association’s statistics on cardiovascular disease revealed that 2020, the first year of the SARS-CoV2 pandemic in the United States, also saw a record high in CVD-related deaths, topping the previous high in 2003 by nearly 19,000 deaths. What’s more, this increase has hit Asian, Black, and Hispanic communities disproportionately. (1) Ischemic injury, such as a myocardial infarction, accounts for a large chunk of this mortality. (1,2) In mammalian hearts, non-muscle cells stop contributing to the pool of heart muscle cells, cardiomyocytes (CMs), early in fetal development, and CMs lose nearly all proliferative capacity shortly after birth. Beating CMs lost during a heart attack are therefore largely replaced by less functional scar tissue, and this loss of function often leads to chronic heart failure. Whole organ transplant is the gold standard of treatment because there do not currently exist therapies to reduce scar size and improve cardiac function after ischemic injury. (3)
If medication or gene therapy could cause a patient's own CMs to reenter the cell cycle or cause the cardiac fibroblasts (CFs) already populating the scar area to change into beating CMs, then we could improve cardiac function after ischemic injury reducing loss of life and the need for invasive therapies like organ transplant. (3) The first hints that such an intervention might be possible came in 1987 when researchers converted fibroblasts to myoblasts by expressing the cardiac lineage regulator MYOCD. (2) However, it is only in the decade immediately preceding the pandemic that research into direct cardiac reprogramming—reprogramming of non-myocytes, typically fibroblasts, into induced cardiomyocytes (iCMs) without passing through a pluripotent intermediate—as a potential therapeutic for ischemic injury graduated from a curiosity to an inevitability. (3) However, low reprogramming efficiency and incomplete maturation of iCMs remain significant barriers to clinical application.
Transcription factors, micro-RNAs, and small molecules are the three broad categories of factors in reprogramming cocktails. Table 1 presents a survey of cocktails used in literature published since 2019. Whichever factors are included, these cocktails are designed to repress starting cell identity and promote CM identity. The most common TF reprogramming strategy for CMs uses three transcription factors: GATA4 (G), MEF2C (M) and TBX5 (T). Often included with GMT is HAND2, (3) which may lead to more mature iCMs. (4) The most common microRNA strategy is “miRCombo”, miR-1, miR-133, miR-208 and miR-499. (5) Strategies using small molecules typically include inhibitors of fibrosis, inflammation and DNA and histone methylation and promoters of chromatin opening and cAMP production. (2)
Table 1:
Survey of recently published literature on cardiac reprogramming and maturation strategies. The factors listed form the optimal cocktail for each study except where otherwise noted. The efficiency of each strategy given represents the most specific assessment assay reported (e.g., for flow cytometry experiments with multiple CM markers, only the double-positive percentage is listed). Where possible, the assay is listed first followed by the effieciency, and when studies use multiple cocktails or models they are given in parentheses. Abbreviations: CM-cardiomyocyte; pm-pacemaker; CPC-cardiac progenitor cell; EC-endothelial cell; Fb-fibroblast; CF-cardiac fibroblast; MEF-mouse embryonic fibroblast; DF-dental fibroblast; H9-human embryonic stem cell line; ADMSC-adipose-derived mesenchymal stromal/stem cell; UC-urine cell; TTF-tail tip fibroblast; VM-ventricular myocytes; AM-atrial myocytes; EHM-engineered human myocardium; HFF-human foreskin fibroblast; m-mouse; r-rat; h-human; a-adult; n-neonatal; i-induced; d-dividing; miR-microRNA; TF-transcription factor; SM-small molecule; DN-dominant negative; Np-nanoparticle; AA-ascorbic acid; ElecStim-electrical stimulation; RA-retanoic acid; Mat-Matrigel; RCCS-rotary cell culture system; FC-flow cytometry; IF-immunofluorescence.
| Target | Starting | Factors | Type | Delivery | Maturation | Max Efficiency |
Ref |
|---|---|---|---|---|---|---|---|
| iCM | mnCF; in vivo | miRCombo (miR-1, miR-133, miR-208, miR-499) | miR | Carbon dots | AA + Se | 100x cells/field vs cnt (cell morphology); 1/3 Fibrosis area post-MI | (36) |
| mnCF | miRCombo | miR | H pep-mod neutrophil-mimicking membrane-mesoporous Si Np | FC cTnT+ 15.2% | (72) | ||
| haCF | miRCombo | miR | lipoplexes | FC cTnT+ 15% | (73) | ||
| haCF | miRCombo | miR | DharmaFect 1 | FC cTnT+ 11% | (5) | ||
| MEF, maCF | PTC-209 (BMIi) -> CHIR990 21 + RepSox (TGFβR1i) + Forskolin + VPA (HDACi) + Parnate (LSD1i) + TTNPB (RA analogue) | SM | SM | FC aMHC+ 41% (MEF); 27.2% (maCF) | (39) | ||
| MEF | 5-azac + A83-01 | SM | SM | FC cTnT+ 48.7%; FC ActA+ 57.3% FC aMHC-GFP+ 22.2% (MEF); IF aMHC+ 40% (HDFs); 1/8 scar area post-MI FC cTnT+ 17% (rCF); 7.5% (hCF) FC cTnT+ 13% (EC); 10.3% (CF) IF ActA+ 10.5% (MEF AM); 9.5% (MEF AMGT); FC cTnT+ActA+ 9.5% (mnCF AM); 14% (mnCF AMGT); 6.5% (hESC-Fb AM); 11.9% (hESC-Fb AMGT) | (42) | ||
| MEF; hnDF; in vivo | GMT | TF | Au Np | (74) | |||
| MEF; rnCF; hnCF | GM + Tead1 | TF | Lenti | (46) | |||
| rECs; rnCF | EC: GMT CF: GMT + ETV2 | TF | Lenti | (28) | |||
| MEF; mnCF; H9-Fb | Ascl1 + M(133); AMGT(13 3) | TF | Lenti + Retro-Polycistronic | (24) | |||
| H9c2 cardiomyoblast | MH | TF | Lenti tet-on Mef2c + recombinant Hand2 | FC aMHC-GFP+ 30% | (76) | ||
| hADMSC | GMT or MESP1 | TF | mRNA | Cardiac DIff Media | 10-100x expression of actn2, tbx5, gja1, tnni3 | (75) | |
| hnDF | GMTH + MYOCD | TF | Neon Transfection | Coculture w/hECs in layers + ElecStim | 3x cTnT expression vs monoculture | (4) | |
| MEF; in vivo | MGTH | TF | Retro | IF TnI+ 39%, IF Lin+ 2% iVM, 12% iAMs (MEF); 100 iCM/section, 100% iVM (in vivo) | (21) | ||
| hUC | MT + Mesp1 + MyoCD PHF7 + GMTH + Akt1 | TF | Retro | FC cTnT+ 22.4% | (19) | ||
| TTF | TF | Retro | FC cTnT+aMHC-GFP+ 18.8% | (32) | |||
| Fb | MGT | TF | Retro-polycistronic | NA | (43) | ||
| MEF; TTF; mnCF | Mef2c isoform 2 + GT | TF | Retro-polycistronic | FC aMHC-GFP+ 5.5% (MEF); 11.5% (mnCF); 12.8% (TTF) IF Titin-eGFP+ActA+ 25% | (30) | ||
| Fb | MGTH | TF | Retro-polycistronic | (78) | |||
| MEF | GMTH | TF | Sendai | 8kPa Mat | IF ActA+ 20% | (84) | |
| in vivo | GMT | TF | Sendai | IF cTnT+ 2.5%; 1/3 fibrosis area post-MI | (70) | ||
| Mouse non-CM; in vivo | GMTH + DN-TGFb + DN-Wnt8a + acid ceramidase | TF + DN + SM | modRNA | IF aMHC-mCherry+ ActA+ 42% (nonCM); 1/2 scar area post-MI | (68) | ||
| H9-Fb; hCF | MGT133 + TBX20 | TF + miR | Lenti-polycistronic | IF cTnI+ 40% (H9-Fb); 70% (HCF) | (27) | ||
| hCF | GMT or HMyocd + shp63 | TF + shRNA | Lenti | FC cTnT+ 4.3% (GMT shP); 14.9% (HMyshP) | (69) | ||
| H9-Fb | MGT133 + shEZH2 | TF + shRNA | Retro-polycistronic | FC cTnT+ 32.7% | (38) | ||
| mnCF; maCF; MEF: H9-Fb | MGT + Atg5 or shBeclin1 | TF + shRNA | Retro/Lenti-polycistronic | FC cTnT+ MGTA 9.4% (mnCF); MGTshB 21% (mnCF); 5% (maCF); 6% (MEF); 20% (H9-Fb) | (51) | ||
| MEF; raCF; haCF | rat: GMT; human: GMTH + MYOCD + miR-590 GHMT + DMSO GMT + CREB/CB Pi MGT + IGF1 + Mll1i (MM589) + A83–1 + PTC-209 shox2 + hcn2 + tbx5 or tbx18 | TF | Lenti | HDACi (NaBut) + WNTi (ICG-001) + RA | FC cTnT+ 23.2% (raCF); 25% (haCF) | (44) | |
| MEF | TF + SM | Retro | IF cTnT+ 13% | (41) | |||
| MEF | TF + SM | Retro | FC aMHC-GFP+ 20% | (40) | |||
| MEF; mnCF | TF + SM | Retro-polycistronic | FC aMHC-GFP+ 25% (MEF); 12.9% (mnCF) | (49) | |||
| hADMSC-iCPC | TF | Lenti | CX30.2-mCherry+ 5.9% | (17) | |||
| ipmCM | MEF | GHMT + Isl1; GMT + d1-32HAND2 | TF | Retro | IF Lin+ GHMTI 0.6% iPM, 0.5% iAM, 0.4% iVM; GMTdH 1% iPM, 0.1% iAM, 0.1% iVM | (22) | |
| Purkinje Cells | hCM; hiPSC-CM | Rolipram + Forskolin + CHIR990 21 + SB43154 2 + VPA + RG108 + Parnate + Resveratrol | SM | SM | RA + NRG1 + Epi | Contactin2-mCherry+ 15.5% | (18) |
| dCM | EHM; in vivo | mouse epicardial ECVs | ECVs | Media/Injection | Removal of factors | FUCCI SG2M CM 4%; 15% D7 after P1MI; 4% D7 after P7MI; IF PH3+ 0.5% (EHM) | (10) |
| in vivo rat | SubQ W8B2+ hCSC | ECVs | TheraCyte device | 2/3 scar area post-MI | (11) | ||
| In vivo | WNTi (CDMG1/2) | SM | SM | Removal of factors | IF PH3+ 0.14%; 2/3 scar area post-MI | (9) | |
| H9-CM | miR-10b | miR | Lipo-fectamine RNAiMAX MYH6-Cre-caErbB2 aMHC-Cre-YAP5SA | IF Edu+ 5.5% | (6) | ||
| maCM | caErbB2 | TF | IF AurKB 2% | (58) | |||
| in vivo | caYap | TF | IF PH3+ 0.95% IF PH3+ 0.7% (ex vivo maCM); 2/3 scar area post-MI, IF EdU+ 5-11% at injury border | (7) | |||
| maCM; in vivo | transient OSKM | TF | i4F-Cre-heart | Removal of factors | (8) | ||
| CPC | MEF; HFF | CHIR990 21 + A83-01 + GSK126 + Forskolin + CTPB + AM580 GT + Nkx2.5 + CHIR990 21 | SM | SM | FC Flk1+PdgfRα + 75.2% | (15) | |
| Fb | TF + SM | Lenti CRISPRa | Nkx2.5-eGFP+ 85.8% | (12) | |||
| HFF | GMTH + MEIS1 | TF | Lenti CRISPRa | ActA + BMP4 -> Epi -> RCCS | FC cTnT+ 8.7%; TPM11+ 62.95% | (13) | |
| hADMSC | MESP1 + ETS2 | TF | Recombinant Protein | 3D Spheres | FC KDR+PDGFR a+ 71.4% | (14) |
Novel reprogramming strategies aim to improve the reprogramming efficiency of these base strategies and/or the maturity of the resulting CMs. The gold standard for assessing novel reprogramming strategies in vitro is to evaluate iCM morphology and expression of CM markers (typically, sarcomeric proteins such as cardiac troponin T and α-actinin evaluated with rt-qPCR, flow cytometry, and immunocytochemistry). In vivo assessments include lineage tracing and evaluating cardiac function post-MI. Assays for assessing maturity of iCMs include evaluating for expression of chamber-specific markers, regularity of sarcomere organization, proportion of cells exhibiting calcium cycling, and frequency of spontaneously beating loci. (2,3)
The purpose of the present literature review is to summarize the current state of the field, with particular focus on the advances occurring since the pandemic brought new levels of urgency to the issue. We will lay out the key factors that have been used for reprogramming in the heart with an eye toward common mechanisms. We will also look at modulation of signaling pathways to improve reprogramming cocktails. Finally, we will explore recent research into safe and efficacious delivery of factors and remaining barriers to clinical translation. Our hope is that this review will synthesize work from a broad range of researchers in various fields to help direct future research to realize that goal.
1. Advances in Cardiac Reprogramming Factors
1.1. Cardiac Reprogramming Strategies to Specific Target Cell Types
One attractive target for cardiac regenerative medicine is causing mature CMs to reenter the cell cycle like developing, proliferative CMs. Strategies to induce proliferation in mature CMs are varied. A fairly straightforward strategy involves exogenous expression of cell cycle regulators or treatment with their agonists could induce proliferation in post-mitotic rat and mouse CMs. (6) Some investigators have found that promotion of metabolic reprogramming to a more glycolytic state (7) is sufficient to promote cell cycle entry for mature CMs. Others have accomplished this by transiently promoting expression of some stem cell regulators (8) or inhibition of others. (9) Still others have shown that ECVs from cultured epicardial cells (10) or signaling from subcutaneously implanted cardiac stem cells (11) can also induce mature CM proliferation among other cardioprotective effects.
Going back even further in the development timeline, many recent papers have explored strategies to reprogram various fibroblasts into multipotent cardiac progenitor cells. These cells, when transplanted, could differentiate into CMs, smooth muscle cells, and endothelial cells at the site of an injury. The strategies include variations of GMT: GT with NKX2.5 and inhibition of GSK3 (12) and GMTH with vascular patterning and circadian regulator MEIS1. (13) One group formed multipotent CPCs using MESP1 and ETS2, a transcription factor involved in a broad range of cellular processes. They then activated β-adrenergic signaling and cultured cells in cardiac spheroids with electrical stimulation to derive highly conductive ventricular CM-like cells. (14) Lastly, one group designed a small molecule cocktail that reprogrammed fibroblasts into CPCs that could be cultured long term and differentiated both in vitro and in vivo. The cocktail included inhibitors for GSK3, TGFβ and EZH2 methyltransferase, with activators of adenylyl cyclase and histone acetyltransferase and a retinoic acid derivative. (15)
Healthy hearts contain not only contractile CMs, but also conductive pacemaker and Purkinje cells that transmit electrical impulses to regulate beating. Recent work has shown it is possible to generate these conductive cells from their contractile cousins. One group used only TBX18, the transcription factor regulating formation of the sinoatrial node, to induce pacemaker cells from neonatal rat ventricular CMs. This was improved by coculture with CFs which helped drive aerobic glycolysis and regular rhythm in the nascent pacemakers. (16) Another showed that expression of three factors, pattern formation regulator SHOX2, the pacemaker channel HCN2, and TBX5, in cardiac progenitor cells induced pacemaker formation. (17) Lastly, Prodan, et al., recently developed a strategy using a small molecule cocktail to convert human CMs into Purkinje cells, which allow rapid proliferation of action potentials from the sinoatrial node to the rest of the heart. (18)
The bulk of this review, however, concerns the direct reprogramming of non-myocytes into mature iCMs. Recent years have seen significant advances in both the efficiency of direct cardiac reprogramming strategies and the maturity of the resulting iCMs. Starting cells for iCM reprogramming have included multiple fibroblast types and non-myocyte cardiac cells from mice, rats and humans as well as human urine cells (19) and adipose-derived mesenchymal stem cells. (14) Different starting cells require variations on reprogramming factors, but broadly speaking reprogramming is currently most efficient in CFs and endothelial cells isolated from neonatal (specifically, P0-P3) mice and rats. (2) Not all the mechanisms of this variation are known, but it is likely that expression of cardiac lineage factors has a priming effect for CFs compared with, for example, MEFs or tail-tip fibroblasts. Additionally, reprogramming efficiency declines rapidly with organismal age. It was recently shown that knockdown of the TF EPAS1 could improve reprogramming specifically for adult CFs, suggesting a greater role for hypoxia and metabolic reprogramming in promoting plasticity of the adult cells. (20)
The most common cardiac reprogramming strategies largely induce non-chamber-specific CMs or ventricular CM-like cells, particularly the latter in in vivo reprogramming, with pacemaker-like cells coming in a distant third. (21) One group showed that inclusion of HAND2 in GMTH led to an increase in the proportion of pacemaker-like cells induced from CFs compared to GMT alone. (22) Much less work has targeted generation of significant proportions of atrial CM-like cells. Recently, Zhao, et al., developed Biowire II, a platform to push cells toward either ventricular or atrial fates through directed differentiation of pluripotent cells and electrical conditioning, (23) but this strategy has yet to be attempted for direct reprogramming.
1.2. Genetic regulators as key cardiac reprogramming factors
Historically, strategies for reprogramming have relied on an understanding of the factors that bring about the target cell type during organismal development. Indeed, most published strategies for direct cardiac reprogramming have relied on an understanding of CM differentiation when selecting factors to screen for transdifferentiation potential. (24) The development rationale broadly underpins all TF-based reprogramming. Introduction of just a few TFs can lead to systemic changes in gene expression, which means that introduction of TFs crucial for the normal development of CMs can drive the suppression of starter cell identity and induction of a CM gene program. The factors making up the major TF-based cardiac reprogramming strategy GMT(H) are all involved in normal CM differentiation at various developmental phases. GATA4 and MEF2C are involved in directing cardiac development from nearly the earliest differentiation of the 3 germ layers. TBX5 regulates chamber formation and atrioventricular septation, (25) and HAND2 is important in ventricular development. (26)
Many recent attempts to modify this core strategy to make cardiac reprogramming more efficient have explored a variety of TFs important to cardiac development. These include adding MYOCD (4) to GMTH, the master-regulator of CM development and the cardiac conductive system TBX20 to MGT133 (27) and the endothelial lineage regulator ETV2 to GMT. (28) The latter pushed CFs through a transient endothelial cell-like state, which is in keeping with the recent observations that CMs can derive from progenitors expressing ETV2 at low levels. (29) Additionally, it has been recently shown that specific isoforms of reprogramming factors, in this case isoform 2 of MEF2C, can lead to higher reprogramming efficiencies. (30)
At the same time, it is becoming more appreciated that reprogramming strategies need not hew so closely to development as to exclude all factors not associated with the target lineage. Some reprogramming mechanisms are common to multiple lineages. For instance, reprogramming strategies typically include a pioneer factor, which locally opens repressive chromatin to make cells more amenable to changes in gene expression. (31) GATA4 fills this role in MGT(+) reprogramming strategies, (24) but recent work has shown that ASCL1, a neuronal reprogramming factor, could replace GATA4 and TBX5, (24) and PHF7, a factor associated with spermatogenesis, replaced GATA4. (32) These factors do not even have to occur naturally. Directed evolution through site saturation mutagenesis and chimeragenesis of Oct4 and other POU family proteins led to the development of an enhanced POU protein which could more effectively reprogram cells. (33) This strategy could be applied to pioneer factors of many lineages, but Oct4 and other pluripotency reprogramming factors have also been used to create more permissive genomes for cardiac reprogramming. (8)
1.3. Epigenetic and Post-Transcriptional Regulation as a Crucial Reprogramming Mechanisms
Lineage specific chromatin patterns represent a major barrier to reprogramming. Missinato, et al., identified four TFs (ATF7IP, JUNB, SP7, and ZNF207) that maintain closed chromatin around TFs important for reprogramming toward multiple lineages. (34) Pioneer factors begin the work of undoing this repression by promoting chromatin opening at their various recognition sites. Additionally, our lab recently showed through integrated scATAC-seq and scRNA-seq that MGT operates at cis-regulatory elements to cooperatively directly open chromatin in regions related to the cardiovascular lineage and indirectly facilitate closing chromatin regions related to fibroblast lineage. (35) Ascorbic acid has also been used alongside miR-Combo to promote chromatin opening in its capacity as a cofactor for TET and H3K36 demethylase. (36,37)
A related mechanism is inhibiting factors that promote repressive chromatin or chromatin modifications leading to differentiation. One potent strategy in this vein is inhibition of polycomb repressive conplex subunits like EZH2 and BMI1 either with shRNAs (38) or small molecules. (39) Inhibition of CREB/CBP chromatin binding and/or remodeling using specific inhibitors or culture in 1% DMSO has also been shown to improve cardiac reprogramming efficiency. (40,41) Finally, KO/shRNA or small molecule inhibitors of histone (e.g., MM589, Forskolin) (15,42) or DNA methyltransferases (e.g., DMAP1-KO, 5-azacytidine, Parnate) have also been shown in recent years to promote cardiac reprogramming. (39)
MicroRNAs represent another avenue for altering gene expression by directing cofactors to specific genes or mRNAs, modulating transcription for the former and translation or degradation dynamics for the latter. The miRs that make up the miRcombo reprogramming cocktail each regulate targets related to silencing fibroblast identity and promoding CM identity. Specifically, miR-1 promotes a litany of cardioprotective phenotypes, miR-133 represses TGFβ, which is important for fibroblast activation and function, miR-208 regulates expression of the sarcomeric myosin heavy chain, and miR-499 regulates calcium signaling. (5)
2. Pathways to Modulate Cardiac Reprogramming
2.1. Classic Signaling Pathways in Cardiac Reprogramming
Epigenetic remodeling is also one of many consequences of altering cell signaling, others including metabolic transition, modulation of transcription landscape, and cell cycle regulation. Various reprogramming strategies take advantage of small molecule inhibitors of signaling pathways such as TGFβ, (15,42,43) WNT, (44) GSK3, (43) BMI1, (39) etc. This section will summarize the recent advances in the understanding of the cell signaling events involved in promoting or inhibiting cardiac reprogramming.
For cardiac reprogramming, suppression of Hippo signaling has long been known to be advantageous by interfering with YAP/TAZ interactions with both the activating SWI/SNF and repressive NuRD complexes. (45) Additionally, inhibition of Hippo signaling frees TEAD1, itself a cardiac reprogramming factor, for binding with MEF2C to promote activation of cardiac lineage transcription patterns. (19,46) Interestingly, expression of a constitutively active YAP in mature CMs promoted CM proliferation in vivo, suggestive of a less differentiated state. (7) This is in keeping with YAP/TAZ as a barrier to induction of mature CMs but may suggest a role for Hippo signaling in cardiac regenerative medicine.
Inhibition of downstream cAMP or p38/MAPK signaling (40) were shown to improve cardiac reprogramming efficiency at least in part by inhibiting CREB-CBP chromatin remodeling. Similarly, inhibition of pro-inflammatory and pro-fibrotic signaling, which prevents activation of fibroblasts to myofibroblasts, improved reprogramming efficiency. (47) Myofibroblast-specific actin isoforms have been shown to be a potent barrier to reprogramming. (48) C-C chemokine signaling was shown to inhibit cardiac reprogramming while inhibitors of CCR1, CCR4, and CCR5 improved reprogramming efficiency. (49) In contrast, agonists of the xeno-RNA sensor Rig1 improved miRCombo reprogramming efficiency. (50) Inhibition of WNT signaling has also been shown to enhance cardiac reprogramming efficiency (44) as well as to induce proliferation in postmitotic CMs. (9) Complicating this, we have recently shown that early activation of WNT signaling by knockdown of Beclin1 before day 4 of MGT reprogramming increases reprogramming efficiency. (51)
This effect was autophagy-independent, related to Beclin1’s role in endosome maturation. (52) Beclin1 has also been shown to inhibit STAT3/JAK2 interactions. (53) Additionally, Beclin1 knockdown has been shown to increase signaling of AKT1, which is used in some cardiac reprogramming cocktails. (32) AKT is a broad-specificity kinase and operates through various mechanisms in reprogramming. One intriguing possibility is its capacity to phosphorylate and inactivate translational repressor YBX1, thereby promoting translation of its target mRNAs. (54) Indeed, we have seen that YBX1 knockdown at early time points enhances cardiac reprogramming efficiency (manuscript in revision). This may represent a generalizable post-transcriptional regulatory pathway for reprogramming.
2.2. Metabolic and Prosurvival Pathways in Cardiac Reprogramming
CMs show high metabolic dependence on oxidative phosphorylation. Consequently, cardiac reprogramming involves a metabolic shift away from glycolysis and towards oxidative phosphorylation. (55) Strategies to alter metabolism and metabolic signaling remain an untapped potential resource in cardiac reprogramming (55) This section will cover new insights into metabolic interventions which should be investigated for their capacity to facilitate cardiac reprogramming.
Terminal differentiation and cell cycle exit of CMs was found to be associated with loss of PKM2 and a shift towards mitochondrial oxidative phosphorylation. (56) Indeed, several groups have recently reported that shifting metabolism towards glycolysis after cardiac injury, through malonate inhibition of SDH, (57) Neuregulin 1 promotion of ErbB2 signaling (58) or metformin treatment at higher doses (59) is sufficient to promote cell cycle entry and proliferation of terminally differentiated CMs. Among the factors that have been shown to promote cardiac reprogramming are agents known to either inhibit pro-glycolytic pathways (e.g. IGF2, (49) GSK3 and YAP/TAZ inhibitors) or promote mitochondrial oxidative phosphorylation (e.g. AKT1 (32,60) retinoic acid (61)). Whether these activities are causative during cardiac reprogramming remains to be explored.
CMs are typically characterized by higher mitochondrial fusion activity, leading to more energetic efficiency. (62) MiR-499, one fourth of miRCombo, has been shown to promote mitochondrial fusion by binding and silencing the mRNA of the master fission regulator DRP1. (63) Likewise, GSK3 inhibition, widely used in cardiac reprogramming, (15,43) has been shown to promote fusion. (60) Indeed, AKT1 is an upstream inhibitor of GSK3 and a potent promoter of fusion and mitochondrial biogenesis. (32) In our own unpublished work, we have seen that inhibition of mitochondrial fission and promotion of mitochondrial biogenesis were sufficient to improve reprogramming efficiency.
Mitochondrial clearance through mitophagy is crucial for normal mitochondrial organization during both differentiation of CPCs (64) and perinatal CM maturation. (65) The same holds for other prosurvival pathways. Cardiac stem cell differentiation is characterized in part by increases in autophagy, mediated in part by an intracellular pool of matrix metalloprotease 9. (66) We showed that knocking down the autophagy regulator ATG5 dramatically reduced reprogramming efficiency in an autophagy-dependent manner. (51) Additionally, a major function of AKT1 is mTORC1 activation, which promotes autophagy, cell growth and metabolic reprogramming. (67) On the other hand, inhibiting proapoptotic pathways by eliminating stress-induced ceramides with acid ceramidase (68) or knocking down p63 (69) has been shown to improve cardiac reprogramming.
3. Advances in Factor Delivery and In Vivo Cardiac Reprogramming
Much work has recently been done exploring alternative delivery systems, both viral and nonviral, that will be both more efficient and safer for clinical translation than lenti- and retroviruses. In the former category are Sendai, adeno and adeno-associated viruses, which do not integrate into the genome. Sendai viruses are attractive for clinical applications because of their ability to replicate in the cytoplasm, leading to higher transgene expression with a lower viral load. (25) In cardiac reprogramming, Sendai virus vectors have improved efficiency and effectively reduced infarct size and cardiac dysfunction in MI models. (70) Adenoviral vectors are comparatively resistant to mutagenesis and allow for tissue specificity. (71) They have also been used clinically as vectors for a subset of SARS-CoV2 vaccines with minimal safety concerns. Finally, adeno-associated viruses have much the same advantages of adenoviruses but much lower immunogenicity. However, they have about half the packaging capacity of Sendai and adenoviruses (~4kbp vs ~8kbp). (71)
Nonviral delivery mechanisms include techniques to deliver plasmid, mRNA, protein or RNPs to the starting cells. Groups have recently used carbon dots, (36) H peptide-modified neutrophil-mimicking membranes on mesoporous silicon nanoparticles (72) and lipoplexes (73) to improve delivery of miRCombo to the starting cells both in vitro and in vivo. Additionally, GMT has been effectively delivered nonvirally using gold nanoparticles. (74) An mRNA delivery system, which allows for more precise control of the timing of factor expression, was used to efficiently transdifferentiate human adipose-derived MSCs to iCMs using GMT or MESP1. (75) Further, using modified mRNA, such as that used in the Pfizer/BioNTech SARS-CoV2 vaccine, can further fine-tune expression level and timing. (68) Similarly, directly delivering protein to starting cells has been used to efficiently differentiate human cardiomyoblasts to beating CMs and to induce hCPCs from hADMSCs. (76)(14) Finally, advances have been made in the use of CRISPR activation (CRISPRa) to increase the expression of endogenous genes to facilitate reprogramming. The current CRISPRa cardiac reprogramming literature uses lentiviral dCas9 and guide RNAs, (12,13) but the possibility remains of using assembled RNPs allowing for nonviral delivery. (77) This strategy is particularly attractive because the actual reprogramming is performed by endogenous gene expression, but endogenously generating the necessary expression levels of reprogramming factors remains a significant barrier.
Controlling the stoichiometry of reprogramming factors can also have a profound effect on efficiency. One group recently showed that a ratio of 1/8:1:1:1:1 of viruses for GMT + MESP1 + MYOCD was optimal for reprogramming. Another way researchers have accomplished this is through development of polycistronic vectors wherein the order of genes after the promoter affects their relative expression. For GMT and GMTH reprogramming polycistronic vectors have been optimized with the order MGT and MGTH. (51,78) In keeping with this, one group recently showed that increasing MEF2C expression 30-fold by fusion with a powerful transactivating domain dramatically improved MGT reprogramming efficiency. (79) Stoichiometry is also important with nonviral delivery. The group that used modRNA for GMTH along with DN-TGFβ, DN-WNT8a, and acid ceramidase tried several stoichiometric ratios with equal or higher proportion of MEF2c or HAND2 required for optimal reprogramming. (68)
4. Remaining Barriers, Future Directions and Concluding Remarks
Despite the rapid progress the field of cardiac reprogramming has seen over the past few years, significant barriers remain for clinical translation. The efficiency of converting starting cells to mature iCMs remains low. Encouragingly, strides have already been made to improve both reprogramming efficiency and maturation. Interestingly, recent work demonstrates that soft, three-dimensional matrices, promotion of mitochondrial energetics, coculture with endothelial cells, and electrical and mechanical stimulation are effective combinatory strategies to produce CMs with mature phenotypes. (4,14,27) The field has benefited in this respect from work on maturation of PSC-derived CMs where recent advances include overexpression of SIRT3 (80) and TRPA1 (81) promoting mitochondrial oxidative phosphorylation, regular cycling of mechanical stimulation (82) and modulation of WNT, MAPK, and AKT signaling. (83) Even accounting for the reduced efficiency of reprogramming in vivo, these techniques have already shown tremendous benefits to cardiac function in animal models of myocardial infarction. (70,84) Along with recent advances in factor delivery, this suggests transition to the clinic is more a matter of time than of needing paradigm-shifting breakthroughs.
Nonetheless, we have barely scratched the surface of what is possible in regenerative medicine. One group recently identified 290 transcription factors which alone are sufficient to change cell fate. (85) Most of them had not been previously reported as reprogramming factors for any lineage, and several were likely able to induce lineages that do not match those in which the factors play a role in normal development. In cardiac reprogramming specifically, there are still promising avenues that remain un- or under-explored. Designing strategies to induce metabolic reprogramming toward oxidative phosphorylation and promote mitochondrial fusion and motility is likely to benefit reprogramming efficiency. Likewise, pro-survival and cell cycle interventions may further improve reprogramming strategies.
In conclusion, while tremendous progress has been made in the study of direct cardiac reprogramming, much work remains before it can be applied to putting a dent in the pandemic-associated rise in CVD-related mortality. Exploring both novel and well-characterized mechanisms of reprogramming and cardiac development will make our strategies smarter and more efficient, and advances in bioengineering will help us deploy these techniques more safely and on a wider scale. When it comes to the future of cardiac reprogramming, the world is our bi(cuspid)valve.
Highlights.
Innovations to cardiac reprogramming improve efficiency and increase iCM maturation
Strategies have been designed to induce proliferative CMs and cardiac progenitor cells
Epigenetics, cell signaling, and metabolism are crucial reprogramming mechanisms
Advances in factor delivery promising for cardiac reprogramming move to clinic
Acknowledgements
B. Spurlock and L. Qian are supported by the National Institutes of Health [] and the American Heart Association []. J. Liu is supported by the National Institutes of Health [].
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.
Declaration of Competing Interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
- 1.Tsao CW, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, et al. Heart Disease and Stroke Statistics-2023 Update: A Report From the American Heart Association. Circulation. 2023. Feb 21;147(8):e93–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Xie Y, Liu J, Qian L. Direct cardiac reprogramming comes of age: Recent advance and remaining challenges. Semin Cell Dev Biol. 2022. Feb;122:37–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sadahiro T, Ieda M. In vivo reprogramming as a new approach to cardiac regenerative therapy. Semin Cell Dev Biol. 2022. Feb;122:21–7. [DOI] [PubMed] [Google Scholar]
- 4.Kwon SP, Song SY, Yoo J, Kim HY, Lee J-R, Kang M, et al. Multilayered cell sheets of cardiac reprogrammed cells for the evaluation of drug cytotoxicity. Tissue Eng Regen Med. 2021. Oct;18(5):807–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Paoletti C, Divieto C, Chiono V. Direct Reprogramming of Adult Human Cardiac Fibroblasts into Induced Cardiomyocytes Using miRcombo. Methods Mol Biol. 2022;2573:31–40. [DOI] [PubMed] [Google Scholar]
- 6.Xie Y, Wang Q, Gao N, Wu F, Lan F, Zhang F, et al. MircroRNA-10b Promotes Human Embryonic Stem Cell-Derived Cardiomyocyte Proliferation via Novel Target Gene LATS1. Mol Ther Nucleic Acids. 2020. Mar 6;19:437–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Monroe TO, Hill MC, Morikawa Y, Leach JP, Heallen T, Cao S, et al. YAP partially reprograms chromatin accessibility to directly induce adult cardiogenesis in vivo. Dev Cell. 2019. Mar 25;48(6):765–779.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Chen Y, Lüttmann FF, Schoger E, Schöler HR, Zelarayán LC, Kim K-P, et al. Reversible reprogramming of cardiomyocytes to a fetal state drives heart regeneration in mice. Science. 2021. Sep 24;373(6562):1537–40. [DOI] [PubMed] [Google Scholar]
- 9.Xie S, Fu W, Yu G, Hu X, Lai KS, Peng X, et al. Discovering small molecules as Wnt inhibitors that promote heart regeneration and injury repair. J Mol Cell Biol. 2020. Jan 22;12(1):42–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Del Campo CV, Liaw NY, Gunadasa-Rohling M, Matthaei M, Braga L, Kennedy T, et al. Regenerative potential of epicardium-derived extracellular vesicles mediated by conserved miRNA transfer. Cardiovasc Res. 2022. Jan 29;118(2):597–611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kompa AR, Greening DW, Kong AM, McMillan PJ, Fang H, Saxena R, et al. Sustained subcutaneous delivery of secretome of human cardiac stem cells promotes cardiac repair following myocardial infarction. Cardiovasc Res. 2021. Feb 22;117(3):918–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Jiang L, Liang J, Huang W, Ma J, Park KH, Wu Z, et al. CRISPR activation of endogenous genes reprograms fibroblasts into cardiovascular progenitor cells for myocardial infarction therapy. Mol Ther. 2022. Jan 5;30(1):54–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wang J, Jiang X, Zhao L, Zuo S, Chen X, Zhang L, et al. Lineage reprogramming of fibroblasts into induced cardiac progenitor cells by CRISPR/Cas9-based transcriptional activators. Acta Pharm Sin B. 2020. Feb;10(2):313–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Islas JF, Abbasgholizadeh R, Dacso C, Potaman VN, Navran S, Bond RA, et al. β-Adrenergic stimuli and rotating suspension culture enhance conversion of human adipogenic mesenchymal stem cells into highly conductive cardiac progenitors. J Tissue Eng Regen Med. 2020. Feb;14(2):306–18. [DOI] [PubMed] [Google Scholar]
- 15. Wang J, Gu S, Liu F, Chen Z, Xu H, Liu Z, et al. Reprogramming of fibroblasts into expandable cardiovascular progenitor cells via small molecules in xeno-free conditions. Nat Biomed Eng. 2022. Apr;6(4):403–20. • Use of small molecule inhibitors of GSK3, TGFβ and histone methyltransferase EZH2 and agonists of adenylyl cyclase, histone acetyltransferase, and retanoic acid receptor to reprogram mouse and human fibroblasts into cardiovascular progenitor cells that could be passaged long term. The CPCs were effectively differentiated into CMs, ECs, and SMCs.
- 16.Chou P-C, Liu C-M, Weng C-H, Yang K-C, Cheng M-L, Lin Y-C, et al. Fibroblasts drive metabolic reprogramming in pacemaker cardiomyocytes. Circ Res. 2022. Jun 24;131(1):6–20. [DOI] [PubMed] [Google Scholar]
- 17.Raghunathan S, Islas JF, Mistretta B, Iyer D, Shi L, Gunaratne PH, et al. Conversion of human cardiac progenitor cells into cardiac pacemaker-like cells. J Mol Cell Cardiol. 2020. Jan;138:12–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Prodan N, Ershad F, Reyes-Alcaraz A, Li L, Mistretta B, Gonzalez L, et al. Direct reprogramming of cardiomyocytes into cardiac Purkinje-like cells. iScience. 2022. Nov 18;25(11):105402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhang X, Chen L, Huang X, Chen H, Cai B, Qin Y, et al. MYOCD is Required for Cardiomyocyte-like Cells Induction from Human Urine Cells and Fibroblasts Through Remodeling Chromatin. Stem Cell Rev and Rep. 2022. Oct;18(7):2414–30. [DOI] [PubMed] [Google Scholar]
- 20.Sun H, Pratt RE, Dzau VJ, Hodgkinson CP. Neonatal and adult cardiac fibroblasts exhibit inherent differences in cardiac regenerative capacity. J Biol Chem. 2023. May;299(5):104694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Zhang Z, Villalpando J, Zhang W, Nam Y-J. Chamber-Specific Protein Expression during Direct Cardiac Reprogramming. Cells. 2021. Jun 16;10(6). •• Evaluation of chamber-specific identity of iCMs reprogrammed with MGTH. In vitro reprogramming generated mostly non-chamber-specific cells followed by VM-like then AM-like cells while in vivo reprogramming generated almost exclusively VM-like cells.
- 22.Fernandez-Perez A, Sathe AA, Bhakta M, Leggett K, Xing C, Munshi NV. Hand2 Selectively Reorganizes Chromatin Accessibility to Induce Pacemaker-like Transcriptional Reprogramming. Cell Rep. 2019. May 21;27(8):2354–2369.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zhao Y, Rafatian N, Feric NT, Cox BJ, Aschar-Sobbi R, Wang EY, et al. A Platform for Generation of Chamber-Specific Cardiac Tissues and Disease Modeling. Cell. 2019. Feb 7;176(4):913–927.e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Wang H, Keepers B, Qian Y, Xie Y, Colon M, Liu J, et al. Cross-lineage potential of Ascl1 uncovered by comparing diverse reprogramming regulatomes. Cell Stem Cell. 2022. Oct 6;29(10):1491–1504.e9. •• The neuronal reprogramming factor ASCL1 can operate as a pioneer factor for various reprogramming strategies, including cardiac and hepatic. ASCL1 can replace GATA4 and TBX5 in MGT reprogramming with the same or higher efficiency.
- 25.López-Muneta L, Miranda-Arrubla J, Carvajal-Vergara X. The future of direct cardiac reprogramming: any GMT cocktail variety? Int J Mol Sci. 2020. Oct 26;21(21). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Dal-Pra S, Hodgkinson CP, Dzau VJ. Induced cardiomyocyte maturation: Cardiac transcription factors are necessary but not sufficient. PLoS ONE. 2019. Oct 17;14(10):e0223842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Tang Y, Aryal S, Geng X, Zhou X, Fast VG, Zhang J, et al. TBX20 improves contractility and mitochondrial function during direct human cardiac reprogramming. Circulation. 2022. Nov 15;146(20):1518–36. • Addition of TBX20 to the MGT133 cocktail significantly improved reprogramming efficiency in H9-derived fibroblasts and hCFs. Additionally, iCMs generated with this cocktail showed increased sarcomere formation, contractility, and mitochondrial oxidative phosphorylation compared to MGT133 alone.
- 28. Mathison M, Sanagasetti D, Singh VP, Pugazenthi A, Pinnamaneni JP, Ryan CT, et al. Fibroblast transition to an endothelial “trans” state improves cell reprogramming efficiency. Sci Rep. 2021. Nov 19;11(1):22605. • GMT reprogramming to iCMs was more efficient using ECs as starting cells rather than CFs. Pushing fibroblasts to a transient endothelial-like state by overexpressing ETV2 enhanced GMT reprogramming to iCMs.
- 29.Cao X, Mircea M, Yakala GK, van den Hil FE, Brescia M, Mei H, et al. ETV2 Upregulation Marks the Specification of Early Cardiomyocytes and Endothelial Cells During Co-differentiation. Stem Cells. 2023. Mar 2;41(2):140–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wang L, Huang P, Near D, Ravi K, Xu Y, Liu J, et al. Isoform Specific Effects of Mef2C during Direct Cardiac Reprogramming. Cells. 2020. Jan 22;9(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kim K-P, Choi J, Yoon J, Bruder JM, Shin B, Kim J, et al. Permissive epigenomes endow reprogramming competence to transcriptional regulators. Nat Chem Biol. 2021. Jan;17(1):47–56. [DOI] [PubMed] [Google Scholar]
- 32. Garry GA, Bezprozvannaya S, Chen K, Zhou H, Hashimoto H, Morales MG, et al. The histone reader PHF7 cooperates with the SWI/SNF complex at cardiac super enhancers to promote direct reprogramming. Nat Cell Biol. 2021. May 3;23(5):467–75. •• PHF7, a histone reader required for normal spermatogenesis, enhanced GMTH reprogramming efficiency by working with SWI/SNF to open chromatin at cardiac super enhancers. PHF7 was also able to replace GATA4 in a cardiac reprogramming cocktail, but with lower efficiency.
- 33. Tan DS, Chen Y, Gao Y, Bednarz A, Wei Y, Malik V, et al. Directed evolution of an enhanced POU reprogramming factor for cell fate engineering. Mol Biol Evol. 2021. Jun 25;38(7):2854–68. •• Directed evolution using iterative site saturation mutagenesis of Oct4 and chimeragenesis using domains from other POU-family genes produced an enhanced POU which could replace Oct4 in OSKM reprogramming to hiPSCs with higher reprogramming efficiency.
- 34.Missinato MA, Murphy S, Lynott M, Yu MS, Kervadec A, Chang Y-L, et al. Conserved transcription factors promote cell fate stability and restrict reprogramming potential in differentiated cells. Nat Commun. 2023. Mar 27;14(1):1709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wang H, Yang Y, Qian Y, Liu J, Qian L. Delineating chromatin accessibility re-patterning at single cell level during early stage of direct cardiac reprogramming. J Mol Cell Cardiol. 2022. Jan;162:62–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Yang L, Xue S, Du M, Lian F. Highly efficient microrna delivery using functionalized carbon dots for enhanced conversion of fibroblasts to cardiomyocytes. Int J Nanomedicine. 2021. Jun 1;16:3741–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wang X, Hodgkinson CP, Dzau VJ. Production of Cardiomyocytes by microRNA-Mediated Reprogramming in Optimized Reprogramming Media. Methods Mol Biol. 2021;2239:47–59. [DOI] [PubMed] [Google Scholar]
- 38.Tang Y, Zhao L, Yu X, Zhang J, Qian L, Jin J, et al. Inhibition of EZH2 primes the cardiac gene activation via removal of epigenetic repression during human direct cardiac reprogramming. Stem Cell Res. 2021. May;53:102365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Testa G, Russo M, Di Benedetto G, Barbato M, Parisi S, Pirozzi F, et al. Bmi1 inhibitor PTC-209 promotes Chemically-induced Direct Cardiac Reprogramming of cardiac fibroblasts into cardiomyocytes. Sci Rep. 2020. Apr 28;10(1):7129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Bektik E, Sun Y, Dennis AT, Sakon P, Yang D, Deschênes I, et al. Inhibition of CREB-CBP Signaling Improves Fibroblast Plasticity for Direct Cardiac Reprogramming. Cells. 2021. Jun 22;10(7). •• Small molecule inhibitors of CREB/CBP and other downstream cAMP signaling effectors improved cardiac reprogramming efficiency. Preconditioning of fibroblasts with CREB/CBP inhibitors led to 2-fold increase in iCM generation.
- 41.Lim CK, Efthymios M, Tan W, Autio MI, Tiang Z, Li PY, et al. Dimethyl sulfoxide (DMSO) enhances direct cardiac reprogramming by inhibiting the bromodomain of coactivators CBP/p300. J Mol Cell Cardiol. 2021. Nov;160:15–26. [DOI] [PubMed] [Google Scholar]
- 42.Jin Y, Kim H, Min S, Choi YS, Seo SJ, Jeong E, et al. Three-dimensional heart extracellular matrix enhances chemically induced direct cardiac reprogramming. Sci Adv. 2022. Dec 14;8(50):eabn5768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bektik E, Fu J-D. Production of Cardiomyocyte-Like Cells by Fibroblast Reprogramming with Defined Factors. Methods Mol Biol. 2021;2239:33–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Singh VP, Pinnamaneni JP, Pugazenthi A, Sanagasetti D, Mathison M, Wang K, et al. Enhanced Generation of Induced Cardiomyocytes Using a Small-Molecule Cocktail to Overcome Barriers to Cardiac Cellular Reprogramming. J Am Heart Assoc. 2020. Jun 16;9(12):e015686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hillmer RE, Link BA. The roles of hippo signaling transducers yap and taz in chromatin remodeling. Cells. 2019. May 24;8(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Singh VP, Pinnamaneni JP, Pugazenthi A, Sanagasetti D, Mathison M, Martin JF, et al. Hippo pathway effector tead1 induces cardiac fibroblast to cardiomyocyte reprogramming. J Am Heart Assoc. 2021. Dec 10;e022659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Van Handel B, Wang L, Ardehali R. Environmental factors influence somatic cell reprogramming to cardiomyocyte-like cells. Semin Cell Dev Biol. 2022. Feb;122:44–9. [DOI] [PubMed] [Google Scholar]
- 48.Zhang Z, Zhang W, Blakes R, Sundby LJ, Shi Z, Rockey DC, et al. Fibroblast fate determination during cardiac reprogramming by remodeling of actin filaments. Stem Cell Reports. 2022. Jul 12;17(7):1604–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Guo Y, Lei I, Tian S, Gao W, Hacer K, Li Y, et al. Chemical suppression of specific C-C chemokine signaling pathways enhances cardiac reprogramming. J Biol Chem. 2019. Jun 7;294(23):9134–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Baksh SS, Hu J, Pratt RE, Dzau VJ, Hodgkinson CP. Rig1 receptor plays a critical role in cardiac reprogramming via YY1 signaling. Am J Physiol, Cell Physiol. 2023. Apr 1;324(4):C843–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Wang L, Ma H, Huang P, Xie Y, Near D, Wang H, et al. Down-regulation of Beclin1 promotes direct cardiac reprogramming. Sci Transl Med. 2020. Oct 21;12(566). •• Researchers screened genes related to autophagy as potential reprogramming factors and found that loss of Atg5 inhibited MGT reprogramming and its overexpression enhanced reprogramming efficiency. Surprisingly, the screen identified another autophagy regulator Beclin1 as a potent barrier to reprogramming. Knockdown of Beclin1 enhanced reprogramming efficiency by regulating WNT signaling through an autophagy-independent mechanism.
- 52.Galluzzi L, Green DR. Autophagy-Independent Functions of the Autophagy Machinery. Cell. 2019. Jun 13;177(7):1682–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hu F, Li G, Huang C, Hou Z, Yang X, Luo X, et al. The autophagy-independent role of BECN1 in colorectal cancer metastasis through regulating STAT3 signaling pathway activation. Cell Death Dis. 2020. May 1;11(5):304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Evdokimova V, Ruzanov P, Anglesio MS, Sorokin AV, Ovchinnikov LP, Buckley J, et al. Akt-mediated YB-1 phosphorylation activates translation of silent mRNA species. Mol Cell Biol. 2006. Jan;26(1):277–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jahng JWS, Zhang M, Wu JC. The role of metabolism in directed differentiation versus trans-differentiation of cardiomyocytes. Semin Cell Dev Biol. 2022. Feb;122:56–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Magadum A, Singh N, Kurian AA, Munir I, Mehmood T, Brown K, et al. Pkm2 regulates cardiomyocyte cell cycle and promotes cardiac regeneration. Circulation. 2020. Apr 14;141(15):1249–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bae J, Salamon RJ, Brandt EB, Paltzer WG, Zhang Z, Britt EC, et al. Malonate promotes adult cardiomyocyte proliferation and heart regeneration. Circulation. 2021. May 18;143(20):1973–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Honkoop H, de Bakker DE, Aharonov A, Kruse F, Shakked A, Nguyen PD, et al. Single-cell analysis uncovers that metabolic reprogramming by ErbB2 signaling is essential for cardiomyocyte proliferation in the regenerating heart. eLife. 2019. Dec 23;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Emelyanova L, Bai X, Yan Y, Bosnjak ZJ, Kress D, Warner C, et al. Biphasic effect of metformin on human cardiac energetics. Transl Res. 2021. Mar;229:5–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Papadopoli D, Pollak M, Topisirovic I. The role of GSK3 in metabolic pathway perturbations in cancer. Biochim Biophys Acta Mol Cell Res. 2021. Jul;1868(8):119059. [DOI] [PubMed] [Google Scholar]
- 61.Miao S, Zhao D, Wang X, Ni X, Fang X, Yu M, et al. Retinoic acid promotes metabolic maturation of human Embryonic Stem Cell-derived Cardiomyocytes. Theranostics. 2020. Aug 1;10(21):9686–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Spurlock B, Tullet J, Hartman JL, Mitra K. Interplay of mitochondrial fission-fusion with cell cycle regulation: Possible impacts on stem cell and organismal aging. Exp Gerontol. 2020. Jul 1;135:110919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wang J-X, Jiao J-Q, Li Q, Long B, Wang K, Liu J-P, et al. miR-499 regulates mitochondrial dynamics by targeting calcineurin and dynamin-related protein-1. Nat Med. 2011. Jan;17(1):71–8. [DOI] [PubMed] [Google Scholar]
- 64.Lampert MA, Orogo AM, Najor RH, Hammerling BC, Leon LJ, Wang BJ, et al. BNIP3L/NIX and FUNDC1-mediated mitophagy is required for mitochondrial network remodeling during cardiac progenitor cell differentiation. Autophagy. 2019. Jul;15(7):1182–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Gong G, Song M, Csordas G, Kelly DP, Matkovich SJ, Dorn GW. Parkin-mediated mitophagy directs perinatal cardiac metabolic maturation in mice. Science. 2015. Dec 4;350(6265):aad2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Yadav SK, Mishra PK. Intracellular matrix metalloproteinase-9 mediates epigenetic modifications and autophagy to regulate differentiation in human cardiac stem cells. Stem Cells. 2021. Apr;39(4):497–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Zhang W, Feng G, Wang L, Teng F, Wang L, Li W, et al. MeCP2 deficiency promotes cell reprogramming by stimulating IGF1/AKT/mTOR signaling and activating ribosomal protein-mediated cell cycle gene translation. J Mol Cell Biol. 2018. Dec 1;10(6):515–26. [DOI] [PubMed] [Google Scholar]
- 68. Kaur K, Hadas Y, Kurian AA, Żak MM, Yoo J, Mahmood A, et al. Direct reprogramming induces vascular regeneration post muscle ischemic injury. Mol Ther. 2021. Oct 6;29(10):3042–58. •• Reprogramming was performed with modified mRNA for MGTH, DN-TGFβ, DN-WNT8a, and acid ceramidase. The cocktail effectively reprogrammed mouse cardiac non-CMs with 42% of cells double-positive for αMHC and α-actinin. Interestingly, in vivo the cocktail promoted neovascularization to the scar area and also to skeletal muscle after hind limb injury.
- 69. Pinnamaneni JP, Singh VP, Kim MB, Ryan CT, Pugazenthi A, Sanagasetti D, et al. p63 silencing induces epigenetic modulation to enhance human cardiac fibroblast to cardiomyocyte-like differentiation. Sci Rep. 2022. Jul 6;12(1):11416. • Reprogramming of human CFs to iCMs using either MGT or HAND2 and MYOCD was enhanced using an shRNA for the apoptosis regulator p63. Interfering with p63’s interaction with HDAC1 by overexpression of p63-transactivatiion inhibitor domain led to a similar enhancement, suggesting this as a plausible mechanism.
- 70.Isomi M, Sadahiro T, Fujita R, Abe Y, Yamada Y, Akiyama T, et al. Direct reprogramming with Sendai virus vectors repaired infarct hearts at the chronic stage. Biochem Biophys Res Commun. 2021. Jun 30;560:87–92. [DOI] [PubMed] [Google Scholar]
- 71.Kang MH, Hu J, Pratt RE, Hodgkinson CP, Asokan A, Dzau VJ. Optimizing delivery for efficient cardiac reprogramming. Biochem Biophys Res Commun. 2020. Nov 26;533(1):9–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wang Q, Song Y, Chen J, Li Q, Gao J, Tan H, et al. Direct in vivo reprogramming with non-viral sequential targeting nanoparticles promotes cardiac regeneration. Biomaterials. 2021. Sep;276:121028. [DOI] [PubMed] [Google Scholar]
- 73.Nicoletti L, Paoletti C, Tarricone G, Andreana I, Stella B, Arpicco S, et al. Lipoplexes for effective in vitro delivery of microRNAs to adult human cardiac fibroblasts for perspective direct cardiac cell reprogramming. Nanomedicine. 2022. Sep;45:102589. [DOI] [PubMed] [Google Scholar]
- 74.Chang Y, Lee E, Kim J, Kwon Y-W, Kwon Y, Kim J. Efficient in vivo direct conversion of fibroblasts into cardiomyocytes using a nanoparticle-based gene carrier. Biomaterials. 2019. Feb;192:500–9. [DOI] [PubMed] [Google Scholar]
- 75.Mueller P, Wolfien M, Ekat K, Lang CI, Koczan D, Wolkenhauer O, et al. RNA-Based Strategies for Cardiac Reprogramming of Human Mesenchymal Stromal Cells. Cells. 2020. Feb 22;9(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Haridhasapavalan KK, Sundaravadivelu PK, Joshi N, Das NJ, Mohapatra A, Voorkara U, et al. Generation of a recombinant version of a biologically active cell-permeant human HAND2 transcription factor from E. coli. Sci Rep. 2022. Sep 27;12(1):16129. • Purified recombinant HAND2, grown in E. coli, enhanced differentiation of cardiomyoblasts to CMs when paired with a tet-on MEF2C.
- 77.Jensen TI, Mikkelsen NS, Gao Z, Foßelteder J, Pabst G, Axelgaard E, et al. Targeted regulation of transcription in primary cells using CRISPRa and CRISPRi. Genome Res. 2021. Nov;31(11):2120–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zhang Z, Zhang W, Nam Y-J. Stoichiometric optimization of Gata4, Hand2, Mef2c, and Tbx5 expression for contractile cardiomyocyte reprogramming. Sci Rep. 2019. Oct 18;9(1):14970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kojima H, Sadahiro T, Muraoka N, Yamakawa H, Hashimoto H, Ishii R, et al. MEF2C/p300-mediated epigenetic remodeling promotes the maturation of induced cardiomyocytes. Stem Cell Reports. 2023. Jun 13;18(6):1274–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wang R, Xu H, Tan B, Yi Q, Sun Y, Xiang H, et al. SIRT3 promotes metabolic maturation of human iPSC-derived cardiomyocytes via OPA1-controlled mitochondrial dynamics. Free Radic Biol Med. 2023. Feb 1;195:270–82. [DOI] [PubMed] [Google Scholar]
- 81.Ding Q, Liu X, Qi Y, Yao X, Tsang SY. TRPA1 promotes the maturation of embryonic stem cell-derived cardiomyocytes by regulating mitochondrial biogenesis and dynamics. Stem Cell Res Ther. 2023. Jun 7;14(1):158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Song M, Jang Y, Kim S-J, Park Y. Cyclic Stretching Induces Maturation of Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes through Nuclear-Mechanotransduction. Tissue Eng Regen Med. 2022. Aug;19(4):781–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Garay BI, Givens S, Abreu P, Liu M, Yücel D, Baik J, et al. Dual inhibition of MAPK and PI3K/AKT pathways enhances maturation of human iPSC-derived cardiomyocytes. Stem Cell Reports. 2022. Sep 13;17(9):2005–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kurotsu S, Sadahiro T, Fujita R, Tani H, Yamakawa H, Tamura F, et al. Soft matrix promotes cardiac reprogramming via inhibition of YAP/TAZ and suppression of fibroblast signatures. Stem Cell Reports. 2020. Sep 8;15(3):612–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ng AHM, Khoshakhlagh P, Rojo Arias JE, Pasquini G, Wang K, Swiersy A, et al. A comprehensive library of human transcription factors for cell fate engineering. Nat Biotechnol. 2021. Apr;39(4):510–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
