MRNA-Enhanced Cell Therapy: Comparison
Please note this is a comparison between Version 2 by Vivi Li and Version 1 by Palas Chanda.

mRNA has emerged as an important biomolecule in the global call for the development of therapies during the COVID-19 pandemic. Synthetic in vitro-transcribed (IVT) mRNA can be engineered to mimic naturally occurring mRNA and can be used as a tool to target “undruggable” diseases. Recent advancement in the field of RNA therapeutics have addressed the challenges inherent to this drug molecule and this approach is now being applied to several therapeutic modalities, from cancer immunotherapy to vaccine development.

  • RNA therapeutics
  • cell therapy
  • cardiovascular regeneration
  • inflammatory signaling
  • nuclear reprogramming
  • iPSCs
  • transdifferentiation
  • cardiovascular ageing

1. Introduction

Advances in the biopharmaceutical industry were accelerated in the global race toward therapies for the COVID-19 pandemic [1]. Most notably, messenger ribonucleic acid (mRNA) vaccines galvanized the field, with lightspeed generation of new therapeutic molecules. For example, within 42 days of the publication of the SARS-CoV-2 sequence by Chinese scientists in January 2020 [2], Moderna sent its RNA vaccine candidate to the National Institute of Allergy and Infectious Disease for preclinical testing. By April 2020, Moderna launched its first clinical trial. Less than 8 months later, Moderna will be seeking Emergency Use Authorization for its vaccine, after phase III trials revealed 95% efficacy and excellent safety. Out of the 236 COVID-19 vaccines being developed, 29 of them are mRNA-based [1] and the first two (BNT162 from Pfizer and MRNA1273 from Moderna) of all the vaccines to complete the phase III clinical trial belong to this category [3,4]. These vaccines will be the first mRNA therapeutics to reach the market. The speed by which mRNA vaccines were developed, and their high degree of efficacy and safety, has brought attention to the great promise of mRNA therapeutics.

Advances in the biopharmaceutical industry were accelerated in the global race toward therapies for the COVID-19 pandemic [1]. Most notably, messenger ribonucleic acid (mRNA) vaccines galvanized the field, with lightspeed generation of new therapeutic molecules. For example, within 42 days of the publication of the SARS-CoV-2 sequence by Chinese scientists in January 2020 [2], Moderna sent its RNA vaccine candidate to the National Institute of Allergy and Infectious Disease for preclinical testing. By April 2020, Moderna launched its first clinical trial. Less than 8 months later, Moderna will be seeking Emergency Use Authorization for its vaccine, after phase III trials revealed 95% efficacy and excellent safety. Out of the 236 COVID-19 vaccines being developed, 29 of them are mRNA-based [1] and the first two (BNT162 from Pfizer and MRNA1273 from Moderna) of all the vaccines to complete the phase III clinical trial belong to this category [3][4]. These vaccines will be the first mRNA therapeutics to reach the market. The speed by which mRNA vaccines were developed, and their high degree of efficacy and safety, has brought attention to the great promise of mRNA therapeutics.

Whereas the majority of drugs approved by U.S. Food and Drug Administration (FDA) are small molecules, such drugs have limitations in the range of diseases that are “druggable” [5,6]. In contrast, mRNA has nearly limitless range, as this biological software can be rapidly modified to encode any therapeutic protein or antigen of interest. Furthermore, with advances in delivery methods, pharmacokinetic and pharmacodynamic properties, enhanced efficacy and stability and reduced immunogenicity and production costs [7,8], mRNA therapeutics have an almost limitless potential.

Whereas the majority of drugs approved by U.S. Food and Drug Administration (FDA) are small molecules, such drugs have limitations in the range of diseases that are “druggable” [5][6]. In contrast, mRNA has nearly limitless range, as this biological software can be rapidly modified to encode any therapeutic protein or antigen of interest. Furthermore, with advances in delivery methods, pharmacokinetic and pharmacodynamic properties, enhanced efficacy and stability and reduced immunogenicity and production costs [7][8], mRNA therapeutics have an almost limitless potential.

mRNA therapeutics offers several advantages over the contemporary small molecule, protein or DNA-based therapies. For example, it is difficult to generate small molecules that will allosterically enhance the activity of a deficient enzyme. It may also be difficult to generate a properly folded and post-translationally modified recombinant protein for the same deficit. By contrast, mRNA encoding the wild-type enzyme is easily generated, and when delivered to the appropriate cell type, can replace the deficient enzyme. Compared to DNA delivered gene vectors, RNA is biologically active in both dividing and non-dividing cells [9] and does not need to enter the cell nucleus to generate its therapeutic effect. Furthermore, with standard mRNA, there is no risk of altering the host genome. Synthetic in vitro-transcribed (IVT) mRNA is designed to mimic naturally occurring mRNA [10], i.e., a single-stranded open reading frame flanked by untranslated regions, a 5′ cap for translation, and a 3′ poly(A) tail for stability [11,12]. Modified nucleosides (e.g., 5-methylcytosine and pseudouridine) are included to reduce the cellular toxicity associated with immunogenicity of exogenous mRNA [13]. Furthermore, the mRNA sequence can be modified to include synonymous codons that are optimal for a specific cell type (reflecting tRNA abundance), or a “translational ramp” (specific initial amino acid sequence that enhances translation) [14,15].

mRNA therapeutics offers several advantages over the contemporary small molecule, protein or DNA-based therapies. For example, it is difficult to generate small molecules that will allosterically enhance the activity of a deficient enzyme. It may also be difficult to generate a properly folded and post-translationally modified recombinant protein for the same deficit. By contrast, mRNA encoding the wild-type enzyme is easily generated, and when delivered to the appropriate cell type, can replace the deficient enzyme. Compared to DNA delivered gene vectors, RNA is biologically active in both dividing and non-dividing cells [9] and does not need to enter the cell nucleus to generate its therapeutic effect. Furthermore, with standard mRNA, there is no risk of altering the host genome. Synthetic in vitro-transcribed (IVT) mRNA is designed to mimic naturally occurring mRNA [10], i.e., a single-stranded open reading frame flanked by untranslated regions, a 5′ cap for translation, and a 3′ poly(A) tail for stability [11][12]. Modified nucleosides (e.g., 5-methylcytosine and pseudouridine) are included to reduce the cellular toxicity associated with immunogenicity of exogenous mRNA [13]. Furthermore, the mRNA sequence can be modified to include synonymous codons that are optimal for a specific cell type (reflecting tRNA abundance), or a “translational ramp” (specific initial amino acid sequence that enhances translation) [14][15].

An mRNA-based approach lends itself to a number of therapeutic modalities, including (a) replacement therapy to compensate for a defective gene/protein, or to supply therapeutic proteins [16]; (b) vaccination, where mRNA encoding specific antigen(s) is administered to trigger protective immunity [16]; (c) cell therapy, which involves transfection of mRNA into the cells ex vivo to therapeutically enhance cell survival, proliferation and/or function [17]; (d) generation of new monoclonal antibodies using mRNA [18]; (e) gene editing, where mRNA is used to express an enzyme that edits and corrects the disease-causing defective gene [19].

Advances in mRNA design, production and delivery has sparked the exploration of mRNA therapy in different fields, such as immunotherapy against cancer and infectious diseases [20], production of growth factors, generation of engineered mesenchymal stem cells (MSCs) and regenerative medicine. 

2. Employing mRNA to Generate iPSCs for Stem Cell Therapy

In 2006, Yamanaka and colleagues [21] demonstrated that terminally differentiated adult somatic cells can be reprogrammed to generate induced pluripotent stem cells (iPSCs) by ectopic expression of a specific set of transcription factors, 

In 2006, Yamanaka and colleagues [21] demonstrated that terminally differentiated adult somatic cells can be reprogrammed to generate induced pluripotent stem cells (iPSCs) by ectopic expression of a specific set of transcription factors, 

Pou5f1

Sox2

Klf4

 and c-

Myc (OSKM) using retroviral vectors. These iPSCs are highly similar to embryonic stem cells (ESC) in terms of self-renewal and the ability to be differentiated to all three germ layers. Thus, human iPSCs can be used as an alternative for human ESCs, thereby avoiding potential ethical issues. This seminal discovery was transformative for the field of regenerative medicine. Patient-specific iPSCs generated from somatic cells can be differentiated to understand the pathobiology of genetic diseases. Furthermore, differentiated derivatives of iPSCs could serve as therapeutic cells. For example, iPSCs generated from a patient with a genetic disease (such as cardiomyopathy due to muscular dystrophy) could undergo ex vivo genome editing [22]. These iPSCs could be differentiated into therapeutic cells which could be transplanted into the patient with minimum risk of genetic incompatibility or immune rejection.

(OSKM) using retroviral vectors. These iPSCs are highly similar to embryonic stem cells (ESC) in terms of self-renewal and the ability to be differentiated to all three germ layers. Thus, human iPSCs can be used as an alternative for human ESCs, thereby avoiding potential ethical issues. This seminal discovery was transformative for the field of regenerative medicine. Patient-specific iPSCs generated from somatic cells can be differentiated to understand the pathobiology of genetic diseases. Furthermore, differentiated derivatives of iPSCs could serve as therapeutic cells. For example, iPSCs generated from a patient with a genetic disease (such as cardiomyopathy due to muscular dystrophy) could undergo ex vivo genome editing [22]. These iPSCs could be differentiated into therapeutic cells which could be transplanted into the patient with minimum risk of genetic incompatibility or immune rejection.

However, viral vectors (retroviral, lentiviral or adenoviral vectors) used to generate iPSCs confer risk of genomic integration and limit the clinical application of such iPSCs [23]. Consequently, several integration-free approaches have been developed, including the Sendai virus [24], cell permeant recombinant proteins [25], non-integrating plasmids or episomal DNA [26,27]. Although these approaches presented minimal risk of genome insertion, the efficiency of iPSC generation is very low. Subsequently, iPSCs were generated using mRNAs encoding 

However, viral vectors (retroviral, lentiviral or adenoviral vectors) used to generate iPSCs confer risk of genomic integration and limit the clinical application of such iPSCs [23]. Consequently, several integration-free approaches have been developed, including the Sendai virus [24], cell permeant recombinant proteins [25], non-integrating plasmids or episomal DNA [26][27]. Although these approaches presented minimal risk of genome insertion, the efficiency of iPSC generation is very low. Subsequently, iPSCs were generated using mRNAs encoding 

POU5F1

SOX2

LIN28A

 and 

NANOG [28], or mRNA encoding the Yamanaka factors [29]. Characterization of the iPSCs [29,30] generated by both mRNA-based methods showed higher identity of the global transcriptional signature with human ESCs by comparison to retroviral-derived iPSCs. Single nucleotide polymorphism analysis revealed that, in comparison to mRNA-derived iPSCs, those derived using the retroviral vectors had more mutations.

[28], or mRNA encoding the Yamanaka factors [29]. Characterization of the iPSCs [29][30] generated by both mRNA-based methods showed higher identity of the global transcriptional signature with human ESCs by comparison to retroviral-derived iPSCs. Single nucleotide polymorphism analysis revealed that, in comparison to mRNA-derived iPSCs, those derived using the retroviral vectors had more mutations.

These observations suggested that an mRNA-based method is a safer choice for clinical applications than retrovirus-derived iPSCs. Although mRNA-based method generated transgene-free iPSCs with reasonable reprogramming efficiency (4%) [31], this protocol required daily transfection for 2 weeks. Recent efforts have simplified and optimized the mRNA-based reprogramming protocols [32,33]. However, once generated using mRNA technology, such iPSCs can be differentiated into clinical grade cardiomyocytes [34] using standard differentiation protocols. Furthermore, mRNA can be used to accelerate differentiation of iPSCs to the desired derivative. For example, mRNA encoding 

These observations suggested that an mRNA-based method is a safer choice for clinical applications than retrovirus-derived iPSCs. Although mRNA-based method generated transgene-free iPSCs with reasonable reprogramming efficiency (4%) [31], this protocol required daily transfection for 2 weeks. Recent efforts have simplified and optimized the mRNA-based reprogramming protocols [32][33]. However, once generated using mRNA technology, such iPSCs can be differentiated into clinical grade cardiomyocytes [34] using standard differentiation protocols. Furthermore, mRNA can be used to accelerate differentiation of iPSCs to the desired derivative. For example, mRNA encoding 

ETV2 has been used to generate iPSC-derived ECs with high (90%) efficiency [35].

has been used to generate iPSC-derived ECs with high (90%) efficiency [35].

3. Employing mRNA to Directly Generate or Enhance Therapeutic Cells

It is also possible to use mRNA to directly generate therapeutic cells and/or enhance their proliferation, survival or functions. For example, mRNA encoding reprogramming or differentiation factors can be transfected into somatic cells that are derived from easily accessible somatic cells (e.g., skin fibroblasts) to directly generate cardiovascular cells ex vivo. Theoretically, these cells could be administered back to the patient by direct injection, or incorporated into biocompatible scaffolds. For example, fibroblasts have been transdifferentiated directly into cardiomyocytes in vitro and in vivo by overexpressing master regulators of cardiomyocyte lineage (i.e., 

Gata4

Mef2c

 and 

Tbx5

) using a retroviral approach [36]. Similarly, we and others have used viral vectors overexpressing master regulators of endothelial lineage (e.g., 

ETV2

FLI1

GATA2

 and 

KLF4

) to transdifferentiate fibroblasts into induced endothelial cells [37]. Rather than viral vectors, mRNA encoding master regulators of cardiovascular lineage may be used to achieve therapeutic transdifferentiation in a non-integrating manner that raises fewer safety concerns. Indeed, using mRNA to encode these endothelial transcription factors, we have successfully transdifferentiated human fibroblasts to endothelial cells with high transcriptional and functional fidelity to authentic human endothelial cells (Meng S and Cooke JP, unpublished data).

Although optimization of mRNA constructs and delivery vehicles are still necessary to introduce such therapies into the clinical practice, they hold great promise as proof-of-concept studies emerge. For instance, cardiac reprogramming of human mesenchymal stromal cells with an mRNA differentiation cocktail has been demonstrated recently [38]. Similarly, overexpression of human vascular endothelial growth factor A (

VEGFA

) with mRNA promotes endothelial specification of the human 

ISL1+ progenitors as well as their engraftment, proliferation and survival in vivo [39].

progenitors as well as their engraftment, proliferation and survival in vivo [39].

Mesenchymal stromal cells (MSCs) are available from different sources, such as the umbilical cord, bone marrow, liver, adipose tissue and multiple dental tissues [40]. These cells have the ability of self-renewal, differentiate into different cell lineages, migrate into the site of injury and secrete proteins which reduce inflammation and promote angiogenesis and tissue repair. Clinical trials are underway to assess the benefits of autologous MSCs in patients with ischemic syndromes or cardiomyopathy [41,42]. These trials are made more difficult by the heterogeneity of the quality of the biological product. A strategy to increase the therapeutic potency and homogeneity of the cell therapy would be through mRNA enhancement. Indeed, synthetic mRNA is being applied to engineer MSCs. Synthetic mRNA has been used to modulate the migratory properties of MSCs by temporal expression of homing proteins on the cell surface. In this way, mRNA-modified MSCs may be targeted to treat vascular inflammation. Specifically in one case [43], MSCs were transfected with three different synthetic mRNAs, 

Mesenchymal stromal cells (MSCs) are available from different sources, such as the umbilical cord, bone marrow, liver, adipose tissue and multiple dental tissues [40]. These cells have the ability of self-renewal, differentiate into different cell lineages, migrate into the site of injury and secrete proteins which reduce inflammation and promote angiogenesis and tissue repair. Clinical trials are underway to assess the benefits of autologous MSCs in patients with ischemic syndromes or cardiomyopathy [41][42]. These trials are made more difficult by the heterogeneity of the quality of the biological product. A strategy to increase the therapeutic potency and homogeneity of the cell therapy would be through mRNA enhancement. Indeed, synthetic mRNA is being applied to engineer MSCs. Synthetic mRNA has been used to modulate the migratory properties of MSCs by temporal expression of homing proteins on the cell surface. In this way, mRNA-modified MSCs may be targeted to treat vascular inflammation. Specifically in one case [43], MSCs were transfected with three different synthetic mRNAs, 

SELPLG

SLeX

 and 

IL10

, to enhance vascular targeting and the anti-inflammatory effect of MSCs, which were then systemically administered to the mice with LPS-induced vascular inflammation. 

SELPLG

 and 

SLeX

 are the ligands for P- and E-/L-selectin, respectively, and 

IL10

 acts as an immunosuppressive cytokine. The tethering capacity to the site of vascular inflammation was significantly improved in these mRNA-engineered MSCs. Furthermore, 

IL10 levels were temporally increased in the inflamed region (the mouse ear) and were associated with superior local anti-inflammatory effect. In a similar approach [44], the therapeutic outcome of targeted delivery of MSCs triple-engineered with 

levels were temporally increased in the inflamed region (the mouse ear) and were associated with superior local anti-inflammatory effect. In a similar approach [44], the therapeutic outcome of targeted delivery of MSCs triple-engineered with 

SELPLG

/

SLeX

/

IL10

 mRNAs was evaluated in a murine model of multiple sclerosis, a form of neurovascular inflammation. The engineered MSCs showed enhanced migration and adherence to inflamed brain microvascular endothelial cells and homing to the inflamed spinal cord. Additionally, 

IL10

 from these MSCs inhibited the proliferation of 

CD4

+ T lymphocytes. In a model of brain ischemia, treatment of MSCs with mRNA encoding integrin a4 (

ITGA4

) facilitated their adhesion to endothelial 

VCAM1 and improved the migration of MSCs into the ischemic region of the brain [45].

and improved the migration of MSCs into the ischemic region of the brain [45].

The C-X-C motif receptor 4 (

CXCR4

) is a chemokine receptor that binds to stromal derived factor-1 (

CXCL12

) expressed in inflammatory sites with high affinity. MSCs engineered with mRNA 

CXCR4

 showed improved cell migration toward 

CXCL12 in transwell experiments, suggesting that transient initiation of chemotaxis can be triggered by mRNA-mediated chemokine receptor overexpression [46,47]. Thus, improvement of migration of MSCs by synthesized mRNA can enhance the capability for regeneration of damaged tissue.

in transwell experiments, suggesting that transient initiation of chemotaxis can be triggered by mRNA-mediated chemokine receptor overexpression [46][47]. Thus, improvement of migration of MSCs by synthesized mRNA can enhance the capability for regeneration of damaged tissue.

4. Employing mRNA for Cardiovascular Regeneration

In addition to modifying therapeutic cells ex vivo, mRNA may be delivered directly into a tissue to have a therapeutic effect. This was first demonstrated in 1992, when Jirikowski and co-workers [10] injected synthetic mRNA encoding antidiuretic hormone (vasopressin) into hypothalamus of rats with a genetic deficiency of vasopressin. These animals have diabetes insipidus, which is characterized by difficulty concentrating urine, and the excretion of large volumes of diluted urine. Intrahypothalamic injection of vasopressin mRNA in these animals induced the synthesis of vasopressin protein and transiently reversed the disease. Since then, the feasibility of using mRNA to replace defective or missing proteins for therapeutic purposes was demonstrated in multiple studies and a variety of tissues [64,65,66,67,68]. Although the majority of the mRNA-based therapies are still in pre-clinical development, a growing number of candidates is reaching first-in-man trials [69,70,71].

In addition to modifying therapeutic cells ex vivo, mRNA may be delivered directly into a tissue to have a therapeutic effect. This was first demonstrated in 1992, when Jirikowski and co-workers [10] injected synthetic mRNA encoding antidiuretic hormone (vasopressin) into hypothalamus of rats with a genetic deficiency of vasopressin. These animals have diabetes insipidus, which is characterized by difficulty concentrating urine, and the excretion of large volumes of diluted urine. Intrahypothalamic injection of vasopressin mRNA in these animals induced the synthesis of vasopressin protein and transiently reversed the disease. Since then, the feasibility of using mRNA to replace defective or missing proteins for therapeutic purposes was demonstrated in multiple studies and a variety of tissues [48][49][50][51][52]. Although the majority of the mRNA-based therapies are still in pre-clinical development, a growing number of candidates is reaching first-in-man trials [53][54][55].

The feasibility of direct intramyocardial injection was first reported in 2013 by Zangi et al. [72]. In this foundational work, 

The feasibility of direct intramyocardial injection was first reported in 2013 by Zangi et al. [56]. In this foundational work, 

VEGFA

 mRNA was injected into the ischemic region of murine myocardium at the time of coronary artery ligation. The local increase in 

VEGFA

 induced the expansion and directed differentiation of endogenous heart progenitors. Furthermore, this intervention markedly improved heart function and enhanced long-term survival of mice with experimental myocardial infarction (MI). Notably, 

VEGFA

 encoding plasmid DNA, unlike mRNA, significantly reduced survival of animals with MI in this study. The unexpected finding might be explained by the temporal differences in expression of 

VEGFA

 because prolonged exposure to 

VEGFA

 expressed from the plasmid DNA was associated with abnormal vascular permeability and myocardial edema.

The efficacy of 

VEGFA mRNA for heart regeneration after MI was subsequently confirmed in a large animal model by Carlsson at al [73]. In this study, MI was induced by a permanent ligation of the mid-left anterior descending coronary artery of mini pigs, and, 7 days after the initial surgery, naked mRNA was injected into the infarct and peri-infarct areas. Two months after injection, significant improvements were observed in left ventricular ejection fraction, contractility and myocardial compliance. Moreover, increased vessel density in the peri-infarct area and decreased myocardial fibrosis were noted in the hearts treated with 

mRNA for heart regeneration after MI was subsequently confirmed in a large animal model by Carlsson at al [57]. In this study, MI was induced by a permanent ligation of the mid-left anterior descending coronary artery of mini pigs, and, 7 days after the initial surgery, naked mRNA was injected into the infarct and peri-infarct areas. Two months after injection, significant improvements were observed in left ventricular ejection fraction, contractility and myocardial compliance. Moreover, increased vessel density in the peri-infarct area and decreased myocardial fibrosis were noted in the hearts treated with 

VEGFA

 mRNA. Notably, the toxicity of the mRNA was also assessed in this study, and neither intradermal nor intravenous administration of the construct into both rats and cynomolgus monkeys increased serum levels of pro-inflammatory cytokines 24 h after injection.

These encouraging data led to the initiation of the first clinical trial of an mRNA therapeutic for cardiac regeneration, which was conducted by AstraZeneca (AZD8601) in collaboration with Moderna [74]. The EPICCURE study (NCT03370887) is a randomized, placebo-controlled, double-blind, multicenter, phase 2a clinical trial of the safety and efficacy of epicardial injections of 

These encouraging data led to the initiation of the first clinical trial of an mRNA therapeutic for cardiac regeneration, which was conducted by AstraZeneca (AZD8601) in collaboration with Moderna [58]. The EPICCURE study (NCT03370887) is a randomized, placebo-controlled, double-blind, multicenter, phase 2a clinical trial of the safety and efficacy of epicardial injections of 

VEGFA mRNA. The inclusion criteria specify patients with stable coronary artery disease and moderately decreased left ventricular ejection fraction who are undergoing coronary artery bypass grafting surgery [75]. The study is currently enrolling and is estimated to be completed in early 2023 [76]. Enrolled participants are to receive a placebo or either a low or high dose of AZD8601 (8 patients in each group) as 30 epicardial injections in a 10-min extension of cardioplegia. Injections will be targeted to ischemic but viable myocardial regions, which will be identified with positron emission tomography imaging. Improvement in myocardial blood flow will be an exploratory efficacy outcome, together with echocardiographic, clinical, functional and biomarker measures. The initiation of EPICCURE was preceded by a randomized, double-blind, placebo-controlled, phase 1 study in men with type 2 diabetes mellitus, where AZD8601 was given intradermally as a single ascending dose into the forearm skin with safety follow-up for 6 months [77]. The only causally treatment-related adverse event observed in the trial was an injection-site reaction of mild intensity, while local skin blood flow was significantly increased within 7 days after mRNA injection and correlated with amount of 

mRNA. The inclusion criteria specify patients with stable coronary artery disease and moderately decreased left ventricular ejection fraction who are undergoing coronary artery bypass grafting surgery [59]. The study is currently enrolling and is estimated to be completed in early 2023 [60]. Enrolled participants are to receive a placebo or either a low or high dose of AZD8601 (8 patients in each group) as 30 epicardial injections in a 10-min extension of cardioplegia. Injections will be targeted to ischemic but viable myocardial regions, which will be identified with positron emission tomography imaging. Improvement in myocardial blood flow will be an exploratory efficacy outcome, together with echocardiographic, clinical, functional and biomarker measures. The initiation of EPICCURE was preceded by a randomized, double-blind, placebo-controlled, phase 1 study in men with type 2 diabetes mellitus, where AZD8601 was given intradermally as a single ascending dose into the forearm skin with safety follow-up for 6 months [61]. The only causally treatment-related adverse event observed in the trial was an injection-site reaction of mild intensity, while local skin blood flow was significantly increased within 7 days after mRNA injection and correlated with amount of 

VEGFA protein concentration in cutaneous dialysate collected at the area. Similar findings were reported in the pre-clinical studies, where VEGF-A mRNA also facilitated healing of diabetic wounds [78,79].

protein concentration in cutaneous dialysate collected at the area. Similar findings were reported in the pre-clinical studies, where VEGF-A mRNA also facilitated healing of diabetic wounds [62][63].

Although 

VEGFA

 is currently the most advanced mRNA therapeutic candidate for cardiovascular regeneration, promising pre-clinical results have been reported for some other constructs targeting distinct molecular pathways. For instance, Chen at al. demonstrated that transcriptional co-activator yes-associated protein (

YYIAP1) mRNA improved myocardial outcome after ischemia-reperfusion (IR) injury in mice [80]. 

) mRNA improved myocardial outcome after ischemia-reperfusion (IR) injury in mice [64]

YYIAP1

 mRNA significantly reduced the innate immune inflammatory response and cardiomyocyte survival in the injured myocardium, and 4 weeks later, heart function was improved and hypertrophic remodeling was suppressed.

Intramyocardial injection of synthetic mRNA encoding insulin-like growth factor-1 (

Igf1) reduced apoptosis of cardiomyocytes after experimental MI in C57B1/6 mice [81]. The treatment augmented 

) reduced apoptosis of cardiomyocytes after experimental MI in C57B1/6 mice [65]. The treatment augmented 

Akt1

 phosphorylation and decreased 

Casp9

 activity and TUNEL positive cells within the border zone 24 h post-MI. Notably, RNA uptake by the heart slice specimens cultured ex vivo was augmented in the presence of hypoxia compared to normoxic conditions. This may in part relate to cell membrane-enhanced polymer/RNA uptake following hypoxia.

The feasibility and benefits of manipulating IGF-1 signaling pathway with mRNA were also demonstrated by Zangi et al. [82]. However, their work revealed that whereas stimulation of 

The feasibility and benefits of manipulating IGF-1 signaling pathway with mRNA were also demonstrated by Zangi et al. [66]. However, their work revealed that whereas stimulation of 

Igf1

 receptor may enhance survival of cardiomyocytes and cardiac progenitors, it may also promote formation of epicardial adipose tissue in the injured heart. When mRNA encoding dominant-negative mutants of the 

Igf1

 receptor and insulin receptor substrate 1 (

Irs1

) were applied to the heart surface of adult mice as a gel to inhibit 

Igf1

 signaling in epicardial cells, it reduced the expression of adipogenic markers as well as the fraction of hearts with epicardial adipose tissue 28 days after MI.

Follistatin-like 1 (

FSTL1

) is increased in the ischemic myocardium. A modified form of 

FSTL1 mRNA (with replacement of asparagine with glutamine in the N-glycosylation site at position 180) was sufficient and necessary to activate cardiomyocyte proliferation and limit cardiac remodeling post-MI, following a single injection of mRNA into the infarct border zone immediately after LAD ligation in mice [83].

mRNA (with replacement of asparagine with glutamine in the N-glycosylation site at position 180) was sufficient and necessary to activate cardiomyocyte proliferation and limit cardiac remodeling post-MI, following a single injection of mRNA into the infarct border zone immediately after LAD ligation in mice [67].

Increased ceramide levels in mammalian heart during acute MI are associated with higher rates of myocyte death and impaired cardiac function [84,85,86]. Accordingly, mRNA encoding the enzyme acid ceramidase (

Increased ceramide levels in mammalian heart during acute MI are associated with higher rates of myocyte death and impaired cardiac function [68][69][70]. Accordingly, mRNA encoding the enzyme acid ceramidase (

Asah1) has been directly injected into the murine myocardium following MI induction. Such treatment was also associated with improved cardiac function, smaller scar size 28 days post-MI and longer survival [87].

) has been directly injected into the murine myocardium following MI induction. Such treatment was also associated with improved cardiac function, smaller scar size 28 days post-MI and longer survival [71].

Pyruvate kinase muscle isoenzyme 2 (

PKM) is an isoenzyme of the glycolytic enzyme pyruvate kinase that is expressed in cardiomyocytes during development and immediately after birth, but not during adulthood [88]. Magadum et al. have recently discovered that 

) is an isoenzyme of the glycolytic enzyme pyruvate kinase that is expressed in cardiomyocytes during development and immediately after birth, but not during adulthood [72]. Magadum et al. have recently discovered that 

PKM regulates the cardiomyocyte cell cycle and reduces oxidative stress damage through anabolic pathways and β-catenin [88]. In addition, these authors demonstrated that cardiomyocyte-specific 

regulates the cardiomyocyte cell cycle and reduces oxidative stress damage through anabolic pathways and β-catenin [72]. In addition, these authors demonstrated that cardiomyocyte-specific 

PKM mRNA promoted cardiomyocyte cell division, enhanced cardiac function and improved long-term animal survival. To achieve specificity, kink-turn motif, a specific binding site for L7Ae protein [89,90], was added to 5′UTR of 

mRNA promoted cardiomyocyte cell division, enhanced cardiac function and improved long-term animal survival. To achieve specificity, kink-turn motif, a specific binding site for L7Ae protein [73][74], was added to 5′UTR of 

PKM

 mRNA. Subsequently, the modified 

PKM mRNA was co-delivered with mRNA encoding L7Ae which included cardiomyocyte-specific microRNA recognition elements (miR1-1 and miR-208a) within 3′UTR [91,92]. Because of these elements, translation of L7Ae was blocked specifically in cardiomyocytes, thereby allowing 

mRNA was co-delivered with mRNA encoding L7Ae which included cardiomyocyte-specific microRNA recognition elements (miR1-1 and miR-208a) within 3′UTR [75][76]. Because of these elements, translation of L7Ae was blocked specifically in cardiomyocytes, thereby allowing 

PKM

 expression (otherwise, L7Ae would interact with kink-turn motif on 

PKM

 mRNA and suppress its translation).

References

  1. COVID-19 Treatment and Vaccine Tracker. Available online: https://covid-19tracker.milkeninstitute.org/#vaccine_RNA-based-vaccine (accessed on 30 November 2020).
  2. Wu, F.; Zhao, S.; Yu, B.; Chen, Y.; Wang, W.; Song, Z.; Hu, Y.; Tao, Z.; Tian, J.; Pei, Y.; et al. A new coronavirus associated with human respiratory disease in China. Nature 2020, 579, 265–269.
  3. Jackson, L.A.; Anderson, E.J.; Rouphael, N.G.; Roberts, P.C.; Makhene, M.; Coler, R.N.; McCullough, M.P.; Chappell, J.D.; Denison, M.R.; Stevens, L.J.; et al. mRNA Vaccine against SARS-CoV-2–Preliminary report. N. Engl. J. Med. 2020, 383, 1920–1931.
  4. Mulligan, M.J.; Lyke, K.E.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Neuzil, K.; Raabe, V.; Bailey, R.; Swanson, K.A.; et al. Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults. Nature 2020, 586, 589–593.
  5. Hopkins, A.L.; Groom, C.R. The druggable genome. Nat. Rev. Drug Discov. 2002, 1, 727–730.
  6. Ezkurdia, I.; Juan, D.; Rodriguez, J.M.; Frankish, A.; Diekhans, M.; Harrow, J.; Vazquez, J.; Valencia, A.; Tress, M.L. Multiple evidence strands suggest that there may be as few as 19 000 human protein-coding genes. Hum. Mol. Genet. 2014, 23, 5866–5878.
  7. Sanghvi, Y.S. A status update of modified oligonucleotides for chemotherapeutics applications. Curr. Protoc. Nucleic Acid. Chem. 2011, 46, 4.1.1–4.1.22.
  8. Shukla, S.; Sumaria, C.S.; Pradeepkumar, P.I. Exploring chemical modifications for siRNA therapeutics: A structural and functional outlook. Chem. Med. Chem. 2010, 5, 328–349.
  9. Zou, S.; Scarfo, K.; Nantz, M.H.; Hecker, J.G. Lipid-mediated delivery of RNA is more efficient than delivery of DNA in non-dividing cells. Int. J. Pharm. 2010, 389, 232–243.
  10. Jirikowski, G.F.; Sanna, P.P.; Maciejewski-Lenoir, D.; Bloom, F.E. Reversal of diabetes insipidus in Brattleboro rats: Intrahypothalamic injection of vasopressin mRNA. Science 1992, 255, 996–998.
  11. Shin, H.; Salzano, G.; Torchilin, V.P. Recent advances in RNA therapeutics and RNA delivery systems based on nanoparticles. Advan. Ther. 2018, 1, 1800065.
  12. Conry, R.M.; LoBuglio, A.F.; Wright, M.; Sumerel, L.; Pike, M.J.; Johanning, F.; Benjamin, R.; Lu, D.; Curiel, D.T. Characterization of a messenger RNA polynucleotide vaccine vector. Cancer Res. 1995, 55, 1397–1400.
  13. Kariko, K.; Muramatsu, H.; Welsh, F.A.; Ludwig, J.; Kato, H.; Akira, S.; Weissman, D. Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Mol. Ther. 2008, 16, 1833–1840.
  14. Presnyak, V.; Alhusaini, N.; Chen, Y.; Martin, S.; Morris, N.; Kline, N.; Olson, S.; Weinberg, D.; Baker, K.E.; Graveley, B.R.; et al. Codon optimality is a major determinant of mRNA stability. Cell 2015, 160, 1111–1124.
  15. Verma, M.; Choi, J.; Cottrell, K.A.; Lavagnino, Z.; Thomas, E.N.; Pavlovic-Djuranovic, S.; Szczesny, P.; Piston, D.W.; Zaher, H.S.; Puglisi, J.D.; et al. A short translational ramp determines the efficiency of protein synthesis. Nat. Comm. 2019, 10, 5774.
  16. Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. mRNA vaccines—A new era in vaccinology. Nat. Rev. Drug Discov. 2018, 17, 261–279.
  17. Gay, C.L.; DeBenedette, M.A.; Tcherepanova, I.Y.; Gamble, A.; Lewis, W.E.; Cope, A.B.; Kuruc, J.D.; McGee, K.S.; Kearney, M.F.; Coffin, J.M.; et al. Immunogenicity of AGS-004 dendritic cell therapy in patients treated during acute HIV infection. AIDS Res. Hum. Retrovir. 2018, 34, 111–122.
  18. Van Hoecke, L.; Roose, K. How mRNA therapeutics are entering the monoclonal antibody field. J. Trans. Med. 2019, 17, 54.
  19. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A Programmable Dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821.
  20. Zhou, L.Y.; Qin, Z.; Zhu, Y.; He, Z.; Xu, T. Current RNA-based therapeutics in clinical trials. Curr. Gene Ther. 2019, 19, 172–196.
  21. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676.
  22. Filareto, A.; Parker, S.; Darabi, R.; Borges, L.; Iacovino, M.; Schaaf, T.; Mayerhofer, T.; Chamberlain, J.S.; Ervasti, J.M.; McIvor, R.S.; et al. An ex vivo gene therapy approach to treat muscular dystrophy using inducible pluripotent stem cells. Nat. Commun. 2013, 4, 1549.
  23. Lee, A.S.; Tang, C.; Rao, M.S.; Weissman, I.L.; Wu, J.C. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat. Med. 2013, 19, 998–1004.
  24. Fusaki, N.; Ban, H.; Nishiyama, A.; Saeki, K.; Hasegawa, M. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2009, 85, 348–362.
  25. Kim, D.; Kim, C.; Moon, J.; Chung, Y.; Chang, M.; Han, B.; Ko, S.; Yang, E.; Cha, K.Y.; Lanza, R.; et al. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell. Stem. Cell 2009, 4, 472–476.
  26. Jia, F.; Wilson, K.D.; Sun, N.; Gupta, D.M.; Huang, M.; Li, Z.; Panetta, N.J.; Chen, Z.Y.; Robbins, R.C.; Kay, M.A.; et al. A nonviral minicircle vector for deriving human iPS cells. Nat. Methods 2010, 7, 197–199.
  27. Yu, J.; Chau, K.F.; Vodyanik, M.A.; Jiang, J.; Jiang, Y. Efficient feeder-free episomal reprogramming with small molecules. PLoS ONE 2011, 6, e17557.
  28. Yakubov, E.; Rechavi, G.; Rozenblatt, S.; Givol, D. Reprogramming of human fibroblasts to pluripotent stem cells using mRNA of four transcription factors. Biochem. Biophys. Res. Comm. 2010, 394, 189–193.
  29. Warren, L.; Manos, P.D.; Ahfeldt, T.; Loh, Y.; Li, H.; Lau, F.; Ebina, W.; Mandal, P.K.; Smith, Z.D.; Meissner, A.; et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 2010, 7, 618–630.
  30. Steichen, C.; Luce, E.; Maluenda, J.; Tosca, L.; Moreno-Gimeno, I.; Desterke, C.; Dianat, N.; Goulinet-Mainot, S.; Awan-Toor, S.; Burks, D.; et al. Messenger RNA–versus retrovirus-based induced pluripotent stem cell reprogramming strategies: Analysis of genomic integrity. Stem Cells Transl. Med. 2014, 3, 686–691.
  31. Mandal, P.K.; Rossi, D.J. Reprogramming human fibroblasts to pluripotency using modified mRNA. Nat. Protoc. 2013, 8, 568–582.
  32. Papapetrou, E.P.; Tomishima, M.J.; Chambers, S.M.; Mica, Y.; Reed, E.; Menon, J.; Tabar, V.; Mo, Q.; Studer, L.; Sadelain, M. Stoichiometric and temporal requirements of Oct4, Sox2, Klf4, and c-Myc expression for efficient human iPSC induction and differentiation. Proc. Natl. Acad. Sci. USA 2009, 106, 12759–12764.
  33. Yoshida, Y.; Takahashi, K.; Okita, K.; Ichisaka, T.; Yamanaka, S. Hypoxia enhances the generation of induced pluripotent stem cells. Cell Stem Cell 2009, 5, 237–241.
  34. Mehta, A.; Verma, V.; Nandihalli, M.; Ramachandra, C.J.A.; Sequiera, G.L.; Sudibyo, Y.; Chung, Y.; Sun, W.; Shim, W. A Systemic evaluation of cardiac differentiation from mRNA reprogrammed human induced pluripotent stem cells. PLoS ONE 2014, 9, e103485.
  35. Wang, K.; Lin, R.; Hong, X.; Ng, A.H.; Lee, C.N.; Neumeyer, J.; Wang, G.; Wang, X.; Ma, M.; Pu, W.T.; et al. Robust differentiation of human pluripotent stem cells into endothelial cells via temporal modulation of ETV2 with modified mRNA. BioRxiv 2020, 6.
  36. Ieda, M.; Fu, J.; Delgado-Olguin, P.; Vedantham, V.; Hayashi, Y.; Bruneau, B.G.; Srivastava, D. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 2010, 142, 375–386.
  37. Wong, W.T.; Cooke, J.P. Therapeutic transdifferentiation of human fibroblasts into endothelial cells using forced expression of lineage-specific transcription factors. J. Tissue Eng. 2016, 7.
  38. Mueller, P.; Wolfien, M.; Ekat, K.; Lang, C.I.; Koczan, D.; Wolkenhauer, O.; Hahn, O.; Peters, K.; Lang, H.; David, R.; et al. RNA-based strategies for cardiac reprogramming of human mesenchymal stromal cells. Cells 2020, 9, 504.
  39. Lui, K.O.; Zangi, L.; Silva, E.A.; Bu, L.; Sahara, M.; Li, R.A.; Mooney, D.J.; Chien, K.R. Driving vascular endothelial cell fate of human multipotent Isl1+ heart progenitors with VEGF modified mRNA. Cell Res. 2013, 23, 1172–1186.
  40. Park, J.S.; Suryaprakash, S.; Lao, Y.; Leong, K.W. Engineering mesenchymal stem cells for regenerative medicine and drug delivery. Methods 2015, 84, 3–16.
  41. Bolli, R.; Hare, J.M.; March, K.L.; Pepine, C.J.; Willerson, J.T.; Perin, E.C.; Yang, P.C.; Henry, T.D.; Traverse, J.H.; Mitrani, R.D.; et al. Rationale and design of the Concert-HF trial (combination of mesenchymal and c-kit(+) cardiac stem cells as regenerative therapy for heart failure). Circ. Res. 2018, 122, 1703–1715.
  42. Wang, S.K.; Green, L.A.; Drucker, N.A.; Motaganahalli, R.L.; Fajardo, A.; Murphy, M.P. Rationale and design of the Clinical and Histologic Analysis of Mesenchymal Stromal Cells in AmPutations (CHAMP) trial investigating the therapeutic mechanism of mesenchymal stromal cells in the treatment of critical limb ischemia. J. Vasc. Surg. 2018, 68, 176–181.e1.
  43. Levy, O.; Zhao, W.; Mortensen, L.J.; Leblanc, S.; Tsang, K.; Fu, M.; Phillips, J.A.; Sagar, V.; Anandakumaran, P.; Ngai, J.; et al. mRNA-engineered mesenchymal stem cells for targeted delivery of interleukin-10 to sites of inflammation. Blood 2013, 122, e23–e32.
  44. Liao, W.; Pham, V.; Liu, L.; Riazifar, M.; Pone, E.J.; Zhang, S.X.; Ma, F.; Lu, M.; Walsh, C.M.; Zhao, W. Mesenchymal stem cells engineered to express selectin ligands and IL-10 exert enhanced therapeutic efficacy in murine experimental autoimmune encephalomyelitis. Biomaterials 2016, 77, 87–97.
  45. Nowakowski, A.; Andrzejewska, A.; Boltze, J.; Nitzsche, F.; Cui, L.; Jolkkonen, J.; Walczak, P.; Lukomska, B.; Janowski, M. Translation, but not transfection limits clinically relevant, exogenous mRNA based induction of alpha-4 integrin expression on human mesenchymal stem cells. Sci. Rep. 2017, 7, 1103.
  46. Ryser, M.F.; Ugarte, F.; Thieme, S.; Bornhäuser, M.; Roesen-Wolff, A.; Brenner, S. mRNA transfection of CXCR4-GFP fusion—Simply generated by PCR-results in efficient migration of primary human mesenchymal stem cells. Tissue Eng. Part C Methods 2008, 14, 179–184.
  47. Wiehe, J.M.; Kaya, Z.; Homann, J.M.; Wöhrle, J.; Vogt, K.; Nguyen, T.; Rottbauer, W.; Torzewski, J.; Fekete, N.; Rojewski, M.; et al. GMP-adapted overexpression of CXCR4 in human mesenchymal stem cells for cardiac repair. Int. J. Cardiol. 2013, 167, 2073–2081.
  48. Kim, Y.K. RNA therapy: Current status and future potential. Chonnam. Med. J. 2020, 56, 87–93.
  49. Connolly, B.; Isaacs, C.; Cheng, L.; Asrani, K.H.; Subramanian, R.R. SERPINA1 mRNA as a treatment for alpha-1 antitrypsin deficiency. J. Nucleic Acids 2018, 2018, 8247935.
  50. DeRosa, F.; Smith, L.; Shen, Y.; Huang, Y.; Pan, J.; Xie, H.; Yahalom, B.; Heartlein, M.W. Improved efficacy in a fabry disease model. Using a systemic mRNA liver depot system as compared to enzyme replacement therapy. Mol. Ther. 2019, 27, 878–889.
  51. Kaczmarek, J.C.; Kowalski, P.S.; Anderson, D.G. Advances in the delivery of RNA therapeutics: From concept to clinical reality. Genome Med. 2017, 9, 60.
  52. Damase, T.R.; Sukhovershin, R.; Boada, C.; Taraballi, F.; Pettigrew, R.; Cooke, J.P. The limitless future of RNA therapeutics. Under review. 2020.
  53. BIONTECH. Available online: https://biontech.de/science/pipeline (accessed on 17 November 2020).
  54. BIONTECH. Available online: https://www.curevac.com/en/pipeline (accessed on 17 November 2020).
  55. Moderna. Available online: https://www.modernatx.com/pipeline (accessed on 17 November 2020).
  56. Zangi, L.; Lui, K.O.; von Gise, A.; Ma, Q.; Ebina, W.; Ptaszek, L.M.; Später, D.; Xu, H.; Tabebordbar, M.; Gorbatov, R.; et al. Modified mRNA directs the fate of heart progenitor cells and induces vascular regeneration after myocardial infarction. Nat. Biotechnol. 2013, 31, 898–907.
  57. Carlsson, L.; Clarke, J.C.; Yen, C.; Gregoire, F.; Albery, T.; Billger, M.; Egnell, A.; Gan, L.; Jennbacken, K.; Johansson, E.; et al. Biocompatible, purified VEGF-A mRNA improves cardiac function after intracardiac injection 1 week post-myocardial infarction in swine. Mol. Ther. Methods Clin. Dev. 2018, 9, 330–346.
  58. Available online: https://www.fiercebiotech.com/biotech/moderna-raises-500m-to-move-mrna-drugs-deeper-into-human-tests (accessed on 17 November 2020).
  59. Anttila, V.; Saraste, A.; Knuuti, J.; Jaakkola, P.; Hedman, M.; Svedlund, S.; Lagerström-Fermér, M.; Kjaer, M.; Jeppsson, A.; Gan, L. Synthetic mRNA encoding VEGF-A in patients undergoing coronary artery bypass grafting: Design of a phase 2a clinical trial. Mol. Ther. Methods Clin. Dev. 2020, 18, 464–472.
  60. Available online: https://clinicaltrials.gov/ct2/show/NCT03370887 (accessed on 17 November 2020).
  61. Gan, L.M.; Lagerström-Fermér, M.; Carlsson, L.G.; Arfvidsson, C.; Egnell, A.; Rudvik, A.; Kjaer, M.; Collén, A.; Thompson, J.D.; Joyal, J.; et al. Intradermal delivery of modified mRNA encoding VEGF-A in patients with type 2 diabetes. Nat. Commun. 2019, 10, 871.
  62. Sun, N.; Ning, B.; Hansson, K.M.; Bruce, A.C.; Seaman, S.A.; Zhang, C.; Rikard, M.; DeRosa, C.A.; Fraser, C.L.; Wågberg, M.; et al. Modified VEGF-A mRNA induces sustained multifaceted microvascular response and accelerates diabetic wound healing. Sci. Rep. 2018, 8, 17509.
  63. Pehrsson, S.; Hölttä, M.; Linhardt, G.; Danielson, R.F.; Carlsson, L. Rapid production of human VEGF-A following intradermal injection of modified VEGF-A mRNA demonstrated by cutaneous microdialysis in the rabbit and pig in vivo. Biomed. Res. Int. 2019, 2019, 3915851.
  64. Chen, J.; Ma, Q.; King, J.S.; Sun, Y.; Xu, B.; Zhang, X.; Zohrabian, S.; Guo, H.; Cai, W.; Li, G.; et al. aYAP modRNA reduces cardiac inflammation and hypertrophy in a murine ischemia-reperfusion model. Life Sci. Alliance 2020, 3, e201900424.
  65. Huang, C.L.; Leblond, A.; Turner, E.C.; Kumar, A.H.; Martin, K.; Whelan, D.; O’Sullivan, D.M.; Caplice, N.M. Synthetic chemically modified mrna-based delivery of cytoprotective factor promotes early cardiomyocyte survival post-acute myocardial infarction. Mol. Pharm. 2015, 12, 991–996.
  66. Zangi, L.; Oliveira, M.S.; Ye, L.Y.; Ma, Q.; Sultana, N.; Hadas, Y.; Chepurko, E.; Später, D.; Zhou, B.; Chew, W.L.; et al. Insulin-like growth factor 1 receptor-dependent pathway drives epicardial adipose tissue formation after myocardial injury. Circulation 2017, 135, 59–72.
  67. Magadum, A.; Singh, N.; Kurian, A.A.; Sharkar, M.T.K.; Chepurko, E.; Zangi, L. Ablation of a single N-glycosylation site in human FSTL 1 induces cardiomyocyte proliferation and cardiac regeneration. Mol. Ther. Nucleic Acids 2018, 13, 133–143.
  68. Ji, R.; Akashi, H.; Drosatos, K.; Liao, X.; Jiang, H.; Kennel, P.J.; Brunjes, D.L.; Castillero, E.; Zhang, X.; Deng, L.Y.; et al. Increased de novo ceramide synthesis and accumulation in failing myocardium. JCI Insight 2017, 2, e82922.
  69. Reforgiato, M.R.; Milano, G.; Fabriàs, G.; Casas, J.; Gasco, P.; Paroni, R.; Samaja, M.; Ghidoni, R.; Caretti, A.; Signorelli, P. Inhibition of ceramide de novo synthesis as a postischemic strategy to reduce myocardial reperfusion injury. Basic Res. Cardiol. 2016, 111, 12.
  70. Klevstig, M.; Ståhlman, M.; Lundqvist, A.; Täng, M.S.; Fogelstrand, P.; Adiels, M.; Andersson, L.; Kolesnick, R.; Jeppsson, A.; Borén, J.; et al. Targeting acid sphingomyelinase reduces cardiac ceramide accumulation in the post-ischemic heart. J. Mol. Cell. Cardiol. 2016, 93, 69–72.
  71. Hadas, Y.; Vincek, A.S.; Youssef, E.; Żak, M.M.; Chepurko, E.; Sultana, N.; Sharkar, M.T.K.; Guo, N.; Komargodski, R.; Kurian, A.A.; et al. Altering sphingolipid metabolism attenuates cell death and inflammatory response after myocardial infarction. Circulation 2020, 141, 916–930.
  72. Magadum, A.; Singh, N.; Kurian, A.A.; Munir, I.; Mehmood, T.; Brown, K.; Sharkar, M.T.K.; Chepurko, E.; Sassi, Y.; Oh, J.G.; et al. Pkm2 regulates cardiomyocyte cell cycle and promotes cardiac regeneration. Circulation 2020, 141, 1249–1265.
  73. Hamma, T.; Ferre-D’Amare, A.R. Structure of protein L7Ae bound to a K-turn derived from an archaeal box H/ACA sRNA at 1.8 A resolution. Structure 2004, 12, 893–903.
  74. Wroblewska, L.; Kitada, T.; Endo, K.; Siciliano, V.; Stillo, B.; Saito, H.; Weiss, R. Mammalian synthetic circuits with RNA binding proteins for RNA-only delivery. Nat. Biotechnol. 2015, 33, 839–841.
  75. Zhao, Y.; Samal, E.; Srivastava, D. Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature 2005, 436, 214–220.
  76. Williams, A.H.; Liu, N.; van Rooij, E.; Olson, E.N. MicroRNA control of muscle development and disease. Curr. Opin. Cell. Biol. 2009, 24, 461–469.
More