ArticlePDF AvailableLiterature Review

Abstract and Figures

Myocardial infarction (MI) and heart failure (HF) are the lead-ing causes of death in the United States and in most other industrialized nations. MI leads to a massive loss of cardiomyo-cytes (CMs), which are replaced with non-CM cells, leading to scarring and, in most cases, HF. The adult mammalian heart has a low intrinsic regenerative capacity, mainly because of cell-cycle arrest in CMs. No effective treatment promoting heart regeneration is currently available. Recent efforts to useDNA-based or viral gene therapy approaches to induce cardiac regeneration post-MI or in HF conditions have encountered major challenges, mostly because of the poor and uncontrolled delivery of the introduced genes. Modified mRNA (modRNA)is a safe, non-immunogenic, efficient, transient, local, and controlled nucleic acid delivery system that can overcome the obstacles to DNA-based or viral approaches for cardiac gene delivery. We here review the use of modRNA in cardia ctherapy, to induce cardioprotection and vascular or cardiac regeneration after MI. We discuss the current challenges in modRNA-based cardiac treatment, which will need to be over-come for the application of such treatment to ischemic heart disease (PDF) mRNA-Based Protein Replacement Therapy for the Heart. Available from: https://www.researchgate.net/publication/329448176_mRNA-Based_Protein_Replacement_Therapy_for_the_Heart [accessed Jan 02 2019].
Content may be subject to copyright.
Review
mRNA-Based Protein Replacement
Therapy for the Heart
Ajit Magadum,
1,2,3
Keerat Kaur,
1,2,3
and Lior Zangi
1,2,3
1
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
2
Department of Genetics and Genomic Sciences, Icahn School of
Medicine at Mount Sinai, New York, NY 10029, USA;
3
Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
Myocardial infarction (MI) and heart failure (HF) are the lead-
ing causes of death in the United States and in most other
industrialized nations. MI leads to a massive loss of cardiomyo-
cytes (CMs), which are replaced with non-CM cells, leading to
scarring and, in most cases, HF. The adult mammalian heart
has a low intrinsic regenerative capacity, mainly because of
cell-cycle arrest in CMs. No effective treatment promoting
heart regeneration is currently available. Recent efforts to use
DNA-based or viral gene therapy approaches to induce cardiac
regeneration post-MI or in HF conditions have encountered
major challenges, mostly because of the poor and uncontrolled
delivery of the introduced genes. Modied mRNA (modRNA)
is a safe, non-immunogenic, efcient, transient, local, and
controlled nucleic acid delivery system that can overcome the
obstacles to DNA-based or viral approaches for cardiac gene
delivery. We here review the use of modRNA in cardiac
therapy, to induce cardioprotection and vascular or cardiac
regeneration after MI. We discuss the current challenges in
modRNA-based cardiac treatment, which will need to be over-
come for the application of such treatment to ischemic heart
disease.
Ischemic Heart Disease
Despite advances in curative and preventive medicine, heart failure
(HF) remains the leading cause of mortality and hospitalization
worldwide.
1,2
Almost 300,000 individuals each year experience recur-
rent heart attacks in the United States alone,
3,4
and the prevalence of
ischemic heart disease is projected to rise to about 40.5% of the USA
population by 2030.
4
Traditional approaches for dealing with end-
stage HF are often not feasible, due to the limited number of hearts
available for transplantation. Preclinical trials have reported improve-
ments in patient outcomes,
5,6
but prognosis remains poor, and there
is, therefore, an urgent need for new approaches to the prevention and
treatment of HF.
During HF, billions of cardiomyocytes (CMs) are progressively lost,
and brotic non-functional scar tissue develops, signicantly reducing
the pumping capacity of the heart muscle. The remaining CMs have a
limited intrinsic regenerative capacity and cannot, therefore, replace
the lost CMs. Cardiac regeneration studies have shown that dividing
CMs are abundant in the fetus, but rapidly lost after birth.
7
Cell-based
therapies with exogenous cells, such as bone marrow cells, cardiac pro-
genitor cells, and other self-renewing stem cells, have been developed
to improve heart function. However, little meaningful improvement
has been reported for these treatments, owing to the limited interac-
tion between the various progenitor cells and the myocardium envi-
ronment during myocardial infarction (MI).
In the last two decades, our understanding of the molecular pathways
and genes involved in the disease has improved, and gene therapy has
emerged as a possible treatment for HF. Given the limited site spec-
icity of pharmacological inhibitors, gene therapy is an exciting pros-
pect for more precise targeting of the signaling pathways involved in
disease progression. The gene therapy approaches currently being
developed for HF aim: (1) to increase the proliferation or contractility
of endogenous CMs; (2) to reprogram cardiac broblasts to develop
into benecial cardiac cell types, such as endothelial cells (ECs) or
CMs; and (3) to increase capillary density by activating endogenous
ECs or progenitors. Recent studies have reported reactivation of the
CM cell cycle following protein delivery to the myocardium, either
by direct injection or via patch delivering the protein to the epicar-
dium. CM proliferation has been reported following the delivery of
NRG1 protein via intraperitoneal (i.p.) injections,
8
intramyocardial
(IM) injections of agrin,
9
or the delivery of follistatin-like 1 to the
epicardium.
10
Furthermore, the proliferation of adult CMs has been
observed following transfection with an adenovirus encoding a domi-
nant-negative p38 mitogen-activated protein kinase (MAPK)
11
or an
extracellular matrix component, periostin.
12
Also, Hajjar and co-
workers
13,14
have shown that the adeno-associated virus (AAV)-
mediated delivery of Serca2a
13
or SUMO
14
improves cardiac function
post-MI and in HF condition via elevation of endogenous CMs
contractility.
Another successful avenue of gene therapy for heart repair is the ge-
netic in situ reprogramming of cardiac broblasts into CMs. Pioneer-
ing work by Srivastava and coworkers
15
showed that broblasts could
undergo cardiac reprogramming to become beating CMs following
direct virus-mediated IM delivery and overexpression of the cardiac
myocyte transcription factors Gata4, Mef2c, and Tbx5 (GMT). This
approach is promising as an alternative to cell-based regeneration
https://doi.org/10.1016/j.ymthe.2018.11.018.
Correspondence: Lior Zangi, Department of Genetics and Genomic Sciences,
Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030,
New York, NY 10029, USA.
E-mail: lior.zangi@mssm.edu
Molecular Therapy Vol. 27 No 4 April 2019 ª2018 The Author(s). 1
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Please cite this article in press as: Magadum et al., mRNA-Based Protein Replacement Therapy for the Heart, Molecular Therapy (2018), https://doi.org/
10.1016/j.ymthe.2018.11.018
therapies, but the efciency of in vivo reprogramming remained low,
and there is also a potential risk of viral genome insertions associated
with the use of viral vectors. Olson and coworkers
16
improved in vivo
reprograming by using Hand2 as an additional reprogramming fac-
tor, together with GMT. In addition to its use to stimulate CM prolif-
eration and reprogramming, gene therapy has also been used to
induce myocardial repair by enhancing angiogenesis and inducing
cardiovascular regeneration. Over the last decade, preclinical studies
have reported revascularization in ischemic heart following the direct
delivery of vascular endothelial growth factors (VEGFs) by various
methods.
1723
Widely used vectors provided robust and consistent
gene expression leading to neovascularization, but this expression
was often accompanied by undesirable effects, such as the induction
of edema or angioma due to the prolonged expression of VEGF, the
elicitation of an immune response against the vector, or a potential
risk of genomic integration.
2022
Nevertheless, these studies have pro-
vided useful insight and support for the use of gene therapy to repair
the injured myocardium, although a number of hurdles remain to be
overcome for this therapeutic approach to be considered successful.
Current Approaches in Cardiac Gene Therapy
Gene therapy can be dened as the transplantation of normal genes
into cells to replace missing or defective genes, with the aim of correct-
ing genetic disorders or promoting inactive benecial mechanisms or
pathways. With improvements in our understanding of cardiac dis-
ease, interest is growing in the use of gene therapies to treat coronary
heart disease. The ultimate goal of gene therapies is the expression of
protein of interest, and the most feasible way of achieving this goal is to
introduce the corresponding protein directly into the myocardium.
Direct protein delivery overcomes the difculties of translation within
the cell, thereby offering a potential advantage in terms of higher levels
of expression, dose regulation, and control over a viral-based gene
therapy approach. However, the short half-life and instability of
injected proteins, the lack of use of this approach for intracellular
proteins (e.g., transcription factors), and possible immunogenicity
due to minor histocompatibility antigens are problems that must be
overcome for this approach to be therapeutically successful.
However, approaches based on the insertion of nucleic acids, which
can be translated into proteins within the cardiac cells, can circum-
vent the challenges of protein therapy. In recent decades, considerable
advances have been made toward the delivery of nucleic acids into the
heart by viral and non-viral vectors. Lentiviral vectors are favored for
cardiac gene therapy by many researchers because they can transduce
non-dividing CMs. However, their chief advantagetheir integration
into the host genome, ensuring sustained gene expressionalso en-
tails a risk of compromising the genome and tumorigenesis. The
exceptional transduction efciencies of adenoviruses and AAVs
have resulted in these vectors being the most widely used for cardio-
vascular applications. Adenoviruses transfer genes efciently into the
myocardium in large animals,
24
but expression is transient, and these
viruses trigger a strong immune response.
25
AAVs have low immuno-
genicity and are a widely used alternative for gene delivery to the
heart. These nonpathogenic vectors ensure cardiac tropism without
integration into the host genome and have been used in a recent
study
26
in which persistent Yap-associated protein expression re-
sulted in CM proliferation and regeneration post-MI. AAV gene
delivery peak levels of expression are reached about 4 weeks after de-
livery
27
and continue for up to 11 months.
28
Despite the predomi-
nance of AAVs over other gene delivery vectors, the production of
neutralizing antibodies against the AAV capsid, delayed pharmacoki-
netics, and limited gene packaging capacity of these vectors restrict
their use in cardiac gene therapy.
29
The delivery of naked plasmid DNA overcomes the risk of immune
responses and oncogenesis, because of the absence of the viral vector.
Plasmid DNA displays impressive organ specicity, but transfection
efciency is low. The recent elucidation of the role of microRNAs
(miRNAs) and long noncoding RNAs in cardiac repair and regener-
ation has provided new hope for innovative therapy.
30
A recent re-
view by Hermans-Beijnsberger et al.
31
summarized newly found
long non-coding RNAs involved in the cellular process during devel-
opment of cardiovascular disease (CVD). These non-coding RNAs
can efciently suppress the target mRNA post-transcriptionally by
promoting mRNA degradation or inhibiting translation. Despite
the successful results obtained in vitro, systems for delivering them
to the heart in vivo have yet to be optimized. Furthermore, therapeutic
miRNAs may have off-target effects, resulting in potential risk of
oncogenesis.
32,33
There is, therefore, an urgent need to explore clini-
cally relevant approaches for enhancing cardiac regeneration and
maintaining correct heart function both during and immediately after
ischemic injury.
Modified mRNA Therapy
Different gene therapies have proved inefcient due to a short half-
life, production of neutralizing antibodies, or a poor transduction
capacity. By contrast, mRNA-based therapies are highly promising
for the treatment of various human disorders. The delivery of
mRNA to the cell has signicant advantages over the use of protein
or DNA-based delivery systems: (1) the use of mRNA transfection
overcomes the need for nuclear localization or for transcription of
the gene of interest in the patients cells; (2) the introduction of
mRNA into cells is safe under physiological conditions, because
mRNA does not integrate into the host genome; and (3) the effect
of the mRNA is transient, minimizing the risk of mutagenesis after
mRNA therapy (Figure 1).
Successful direct mRNA transfer was rst reported about three
decades ago, when Wolff et al.
34
demonstrated the delivery of
mRNA and its translation into protein in mouse skeletal muscle. After
a few initial successes with mRNA therapy, research into mRNA
structure and delivery methods continued, but the use of mRNA ther-
apy was limited to vaccine development, because of problems of insta-
bility and immunogenicity. Within cells, mRNA is prone to cleavage
by RNase and can trigger the innate immune system via Toll-like
receptors (TLRs) 7 and 8 (which recognize single-stranded RNA)
or TLR3 (which recognizes double-stranded RNA), leading to an in-
crease in cytokine levels and associated toxicity. In 2008, pioneering
2 Molecular Therapy Vol. 27 No 4 April 2019
www.moleculartherapy.org
Review
Please cite this article in press as: Magadum et al., mRNA-Based Protein Replacement Therapy for the Heart, Molecular Therapy (2018), https://doi.org/
10.1016/j.ymthe.2018.11.018
work by Karikó et al.
35
addressed these issues and provided a platform
for mRNA therapy in genetic, regenerative medicine, immunothera-
peutics, and cancer. The study showed that replacing the uridine
residues in mRNA with the naturally occurring modied nucleoside
pseudouridine (hence the name modied mRNA [modRNA])
enhanced translation, due to changes in the secondary structure of
the mRNA limiting its recognition by the TLRs and nucleases.
35,36
The use of modRNA has since been on the increase in genetic
medicine, for protein-replacement therapies and the treatment of ge-
netic diseases. The efciency of modRNA delivery in vivo has been
increased by enhancing the stability of the mRNA and increasing
translational efciency by capping the molecule with the 30-O-Me-
m7G(50)ppp(50)G Anti Reverse Cap Analog (ARCA) at its 50
end.
37,38
The uses of modRNA technology as a model for cardiac
repair are listed in Table 1.
Immediately after MI, CMs and other cardiac cells such as ECs are
lost due to occlusion of the coronary artery. A chain of events down-
stream leads to oxidative stress and inammation, resulting in
impaired pump function and, ultimately, HF. The remaining CMs
in the heart have a very limited proliferative potential and are there-
fore unable to replace the lost cells. The damaged coronary vascula-
ture also creates a hostile environment in which it is difcult for
the remaining CMs to survive. Various strategies have been developed
Figure 1. Methods of Gene Delivery to the Heart
In vivo gene expression profiles for various methods of gene delivery to the heart. (A) Recombinant protein. (B) Modified mRNA (modRNA). (C) Adeno-associated viruses
(AAVs).
Molecular Therapy Vol. 27 No 4 April 2019 3
www.moleculartherapy.org
Review
Please cite this article in press as: Magadum et al., mRNA-Based Protein Replacement Therapy for the Heart, Molecular Therapy (2018), https://doi.org/
10.1016/j.ymthe.2018.11.018
to try to reverse the situation by inducing regeneration of cardiac neo-
vasculature and encouraging CMs to proliferate.
A few independent clinical trials over the last 20 years have assessed
the therapeutic potential of a potent angiogenic factor, VEGF-A,
after ischemic injury. VEGF-A was delivered by intracoronary,
intravenous, or IM injection, in the form of a recombinant
protein,
1719
adenoviral plasmid,
20,22
naked cDNA, or non-viral
plasmid.
21,23
A moderate improvement in ejection fraction and left
ventricular (LV) function was reported, but ndings differed between
trials.
3941
These differences can be explained by the short half-life of
VEGF-A in plasma (about 3045 min in humans), degradation by
proteases, off-target effects associated with systemic delivery,
and the lack of an efcient delivery platform. Attempts to retain
VEGF-A in the infarcted heart for therapeutic purposes have been
made, based on the implantation of biodegradable scaffolds including
hydrogel,
42
collagen,
43
or self-assembling peptide nanobers,
44
but
the success of these approaches was limited.
4244
Zangi et al.
45
introduced a modRNA encoding VEGF-A into mouse
hearts, and reported a decrease in infarct size and an improved
myocardial outcome with higher survival rates (80% survival with
VEGF-A modRNA versus only 20% for the group receiving DNA).
They observed that VEGF-A protein secretion levels were much
higher following treatment with modRNA than with unmodied
mRNA, with no reported apoptosis or increase in the expression of
immune response genes, such as retinoic acid-inducible gene
(RIG)-1, interferon (IFN)-a, and IFN-b. Both VEGF-A modRNA
and VEGF-A DNA increased capillary density and reduced infarct
size and apoptotic cell frequency in MI mice, but the prolonged expo-
sure to VEGF-A in DNA-treated hearts increased vessel permeability,
a sign of abnormal vessel function. The study also showed that the
favorable outcome achieved with pulse-like VEGF-A overexpression
was associated with better vessel formation in the peri-infarct area
because of the presence of larger numbers of WT1 epicardial progen-
itor cells activated via the kinase insert domain receptor (KDR) under
stress conditions. These activated progenitor cells remain conned to
the epicardial layer in the absence of VEGF-A, which induces their
mobilization to the myocardial layer. Stimulation of the endogenous
epicardial progenitor pool by the right paracrine factor (VEGF-A),
time, and place enhances the differentiation of these cells into ECs
and, to some extent, into CMs. Therefore, VEGF-A modRNA is an
Table 1. Published Studies for the Use of modRNA Technology as a Model of Cardiac Repair: modRNA as a Therapeutic Strategy for Cardiac Vascularization
and Regeneration
No. Publication Gene(s) Role Cellular Process or Disease Delivery Material Animal
1 Zangi et al.
45
VEGFa
directs the fate of heart progenitor
cells and induces vascular regeneration
after MI
cellular fate switch post-MI RNAiMAX mice
2 Lui et al.
46
VEGFa
VEGF-A promotes not only the endothelial
specication but also engraftment,
proliferation, and survival (reduced
apoptosis) of the human Isl1
+
progenitors
in vivo
VEGFa promotes Isl1
+
to
endothelial cell fate, proliferation
and survival of Isl1
+
progenitors
RNAiMAX mice
3 Huang et al.
52
IGF-1
anti-apoptosis, cardiomyocyte survival,
augmented Akt phosphorylation, and
decreased caspase-9 activity
anti-apoptosis, cardiomyocyte
survival post-MI
polyethylenimine-based
nanoparticle mice
4 Turnbull et al.
58
EGFP
modRNA delivery (direct myocardial or
intracoronary administration) into rat
and pig heart
modRNA expression in heart formulated lipidoid
nanoparticles (FLNP) rat and pig
5 Turnbull et al.
59
EGFP protocol lipidoid mRNA nanoparticles
protocol
formulated lipidoid
nanoparticles (FLNP) rodents
6 Kondrat et al.
37
protocol modied mRNA synthesis RNAiMAX mice
7 Sultana et al.
51
modRNA delivery
optimization
modRNA delivery optimization, modRNA
amount and time optimization optimal modRNA expression sucrose-citrate buffer mice
8 Zangi et al.
53
DN-IGF-1R, IGFR inhibition of adipogenic differentiation
post-MI
inhibition of adipogenic
differentiation post-MI RNAiMAX mice
9 Carlsson et al.
47
VEGFa increased angiogenesis, improved heart
function post-MI, reduced brosis
increased angiogenesis, improved
heart function post-MI, reduced
brosis
sucrose-citrate buffer pig, monkey
10 Singh et al.
57
EGFP, mCherry, Fluc modRNA delivery optimization optimal modRNA expression in
heart
alginate, nanomaterial
encapsulated mice and pig
11 Magadum et al.
48
mutated FSTL1
ablation of N180Q, N-glycosylation site of
hFSTL1 by modRNA delivery increased
CM proliferation, improved cardiac
output, and reduced scar size post-MI
CM proliferation, decreased scar
size, improved heart function sucrose-citrate buffer mice
4 Molecular Therapy Vol. 27 No 4 April 2019
www.moleculartherapy.org
Review
Please cite this article in press as: Magadum et al., mRNA-Based Protein Replacement Therapy for the Heart, Molecular Therapy (2018), https://doi.org/
10.1016/j.ymthe.2018.11.018
excellent clinical approach to repair of the damaged vasculature and
can further improve myocardial outcome and survival after injury.
Moreover, VEGF-A modRNA also can promote the engraftment,
proliferation, and survival (reduced apoptosis) of transplanted
human Isl1-positive cells.
46
Carlsson et al.
47
recently reported ef-
cient intracardiac transfection and protein expression from a
VEGF-A modRNA in a pig model of MI. They reported improve-
ments in % ejection fraction, inotropic function and compliance,
border zone capillary and arteriole density, and a decrease in myocar-
dial brosis 2 months after the treatment of MI with VEGF-A mod-
RNA. These improvements in cardiac systolic parameters were
observed following the delivery of 1 or 10 mg modRNA via intracar-
diac injections post-MI.
Several attempts have been made to use conventional viral proteins to
upregulate cell-cycle promoters or to downregulate the brakes on the
cell cycle, with the aim of promoting the re-entry of post-mitotic CMs
into the cell cycle. However, the long-term uncontrolled expression of
pro-proliferative genes can be detrimental to heart function. For this
reason, modRNA technology has been tested in the eld of cardiac
regeneration. Magadum et al.
48
recently investigated the role of
hFSTL1 glycosylation in CM proliferation and showed that the
myocardial injection of a mutated hFSTL1 modRNA with a single
asparagine-to-arginine (N-Q) substitution in the glycosylation site
(N180Q) was necessary and sufcient to increase the proliferation
of neonatal rat or mouse adult CMs in vitro or after MI, respectively,
with no signs of cardiac hypertrophy. This nding can be explained
by changes in the glycosylation pattern of hFSTL1 upon N180 site
ablation, leading to activation of CM proliferation and regeneration.
Interestingly, a single administration of N180Q modRNA in the
mouse MI model was sufcient to increase cardiac function signi-
cantly, with a decrease in scar size and an increase in capillary density
28 days post-MI, showing modRNA to be an efcient tool for induc-
tion of control CM proliferation and cardiac regeneration post-MI.
Our studies of the use of modRNA technology have yielded promising
results, showing that it is possible to create mutated constructs or pro-
teins for investigations of their role in heart disease and, potentially, to
introduce therapeutic constructs for cardiological treatments.
Cardioprotective Role of modRNA
modRNA-based gene delivery has several advantages over other
intracardiac therapies. Viral vectors and plasmid DNA delivery
methods have spatiotemporal shortcomings, whereas modRNA al-
lows rapid, transient, and efcient gene expression to a specic
time window after cardiac injury. In this respect, modRNA is an ideal
tool for delivering factors targeting the signaling pathways altered in
the rst few hours of infarction.
A series of events takes place after MI, leading to a massive sudden
loss of CMs, beginning within an hour of occlusion. The stress to
which CMs are subjected post-MI leads to the induction of pro-
inammatory cytokines and chemokines, and an accumulation of
inammatory cells in the heart.
49
This chain of events occurs rapidly,
within 23 days of ischemia injury. These days thus constitute the
time frame in which desirable gene combinations could be delivered
to prevent CM apoptosis.
50
Sultana et al.
51
have shown that luciferase
modRNA can be detected in the heart 10 min after injection, and that
its expression peaks at 24 hr but remains detectable for up to 10 days.
Thus, based on its expression dynamics, modRNA has been used in
various studies to deliver genes or gene combinations for cytoprotec-
tion and to induce cellular reprogramming in a desired time frame
after cardiac injury.
Consistent with this approach, Huang et al.
52
delivered insulin growth
factor 1 (IGF-1) modRNA to the area of mouse hearts at high risk af-
ter injury, and extended the temporal window for the cytoprotection
of CMs against apoptosis after hypoxia and MI. The delivery of IGF-1
modRNA, with a polyethylenimine-based nanoparticle, resulted in
efcient transient protein expression within cells. IGF-1 was ex-
pressed rapidly, within 2 hr of injection, and its levels peaked 24 hr
post-injection, decreasing thereafter to 48 hr, and about 25% of cells
in the border zone were transfected. The delivery of IGF-1 modRNA
promoted CM survival and decreased cell apoptosis by more than
50% post-hypoxia in vitro and post-MI. The increase in IGF-1 levels
was shown to be associated with CM survival and a decrease in the
number of TUNEL-positive cells post-hypoxia. The decrease in
apoptosis rates was accompanied by higher levels of Akt and Erk
phosphorylation and a downregulation of IGF-1-specic miRNAs.
Despite this demonstration of the cardioprotective role of IGF-1,
Zangi et al.
53
found that the activation of IGF-1 signaling pathways
in the heart post-MI also had negative consequences. In addition to
its cardioprotective action, IGF-1 expression can lead to the forma-
tion of epicardial adipose tissue (EAT) post-MI. EAT is an active
tissue located between the myocardium and the visceral pericardium,
and contributes to the pathological mechanisms of coronary artery
disease. Excessive epicardial fat deposition around the heart may
trigger the production of several adipocytokines and chemokines
through the activation of various paracrine and vasocrine signaling
pathways, resulting in the development of atherosclerotic plaques in
the coronary vessels.
54
Hence the group evaluated the role of para-
crine contributors in the development of EAT under normal and
pathological conditions.
The study showed that IGF-1 delivered to post-MI stressed hearts by
modRNA contributed to the differentiation of epicardial progenitor
cells into adipogenic cells and the formation of EAT.
53
It was demon-
strated that WT1 expression was essential for epicardium-derived cells
(EPDCs) differentiation into adipocytes, by delivering a Cre modRNA
by gel application onto the surface of WT1
x/x
;Rosa26
Tomato
hearts
for local WT1 inactivation and EPDC labeling. This study provided
unique insight into the modRNA gene delivery method, in which
a gene can be delivered locally through the application of a biocom-
patible gel directly onto the cardiac tissue in situations in which the
development of knockout animals is not possible. This mode of gene
transfection was also used to deliver dominant-negative IGF-1 recep-
tor antagonists to the injury-exposed epicardial cells shortly after MI,
Molecular Therapy Vol. 27 No 4 April 2019 5
www.moleculartherapy.org
Review
Please cite this article in press as: Magadum et al., mRNA-Based Protein Replacement Therapy for the Heart, Molecular Therapy (2018), https://doi.org/
10.1016/j.ymthe.2018.11.018
during the brief time window in which the IGF-1-induced differenti-
ation of progenitor cells into adipocytes appears to occur. The tran-
sient inhibition of IGF-1 receptors signicantly decreased EAT
formation, conrming our hypothesis that IGF-1 receptor signaling
is required to stimulate the adipogenic differentiation of EPDCs in
the context of MI. This study provides an illustration of the ability
of modRNA techniques to deliver a gene transiently at the appropriate
time and place to block an undesired signaling pathway in one cell type
(EPDCs), but not on another (CMs).
Challenges in the Cardiac Delivery of modRNA
Improvements in our understanding of the pathology of HF over time
have led to novel gene therapy targets being identied, although inef-
cient delivery to the target tissue remains a substantial problem. For
efcient gene therapy in the heart, the delivery systems carrying the
nucleic acid must ensure: (1) the uptake of the nucleic acid by cardiac
cells; (2) escape from the immune response; and (3) efcient transla-
tion and biodistribution of the genes in the post-ischemic, peri-
ischemic, or non-ischemic areas of the myocardium.
Cells typically take up modRNA via endocytosis, a process in which
foreign molecules or ligands (in this case, modRNA) are engulfed
by an area of plasma membrane, which then buds off intracellularly,
leading to the formation of modRNA-containing endosomes. These
endosomes later disassemble to deliver the mRNA to the cytoplasm,
in which it is immediately translated into protein. However, human
TLR8 (hTLR8) and mouse TLR7 (mTLR7), which are expressed
only on endosomal membranes, recognize single-stranded RNA
[particularly poly(U) and poly(U/G) motifs in the case of hTLR8],
and this recognition triggers the innate immune response. TLR3 is
also expressed on endosomal membranes and can elicit an innate
immune response following its recognition of unmethylated CpG
motifs in double-stranded RNA.
55
Another obstacle to the translation
of the imported mRNA is its degradation by RNase. Over a decade
ago, a revolutionary study by Karikó et al.
35
demonstrated that the
replacement of uridine residues in the mRNA with naturally pro-
duced pseudouridine resulted in much lower levels of TLR-mediated
immunogenicity and prevented degradation by RNase. Subsequent
studies, including studies by our group, have used such modied
RNA to achieve high translation efciencies without immunogenicity
in non-cardiac tissues.
45,51
In 2015, Andries et al.
56
showed that natu-
rally produced 1-mJU incorporation into mRNA reduced immuno-
genicity in mammalian cells lines by preventing endosomal TLR3
activation and downstream innate immune signaling. Consistent
with these ndings, our modRNA, containing 1-mJU in place of uri-
dine residues, resulted in signicantly lower levels of activation for
innate immunity genes, such as those encoding IFN-aor IFN-b
and RIG, in cardiac cells and tissues than were observed with unmod-
ied mRNA. We were also able to show that modRNA with 1-mJU
was less likely to be degraded by RNase compared with unmodied
mRNA (Figure 2). Thus, the choice of an appropriate delivery method
is critical for transfection with a large modRNA that cannot simply
diffuse into the negatively charged CMs.
Maximum transfection efciency and modRNA stability in vitro can
be ensured by complexing modRNA with transfection reagents, to
Figure 2. Comparing Uptake of RNA and modRNA by the Cell
(Left) modRNA delivery does not cause any activation of immune response and escapes RNase degradation. (Right) mRNA triggers activation of TLR7/8 and is prone to
degradation by RNase.
6 Molecular Therapy Vol. 27 No 4 April 2019
www.moleculartherapy.org
Review
Please cite this article in press as: Magadum et al., mRNA-Based Protein Replacement Therapy for the Heart, Molecular Therapy (2018), https://doi.org/
10.1016/j.ymthe.2018.11.018
encapsulate the modRNA with positively charged polymers or lipids.
These spherical vesicles containing polar head groups and nonpolar
tails promote the electromagnetic attachment and subsequent endo-
cytosis of the complex. The efcient transfection of isolated CMs can
be achieved by delivering 0.013 mg/mm
3
modRNA in complex with
the positively charged transfection reagent RNAiMAX. However,
despite the efcient delivery of modRNA to cardiomyocytes in vitro
reported with RNAiMAX, the use of this agent is associated with
higher rates of cell death around the injection site in the myocardium,
suggesting it may not be an ideal vehicle for in vivo transfection.
51
Microencapsulated modRNA in nanoparticles was recently tested as a
way of delivering modRNA to the heart. Expression of the protein was
observed in multiple cell lines and primary CMs, within 24hrof
transfection, and persisted for up to 7 days without altering the struc-
tural and functional properties of the cells.
57
This study demonstrated
the simultaneous delivery of multiple genes to mouse hearts and
showed that the reporter gene was efciently delivered by an alginate
gel in the pig MI model. Similarly, Turnbull et al.
58,59
used formulated
lipidoid nanoparticles
59
(FLNPs) and assessed modRNA transfer into
the heart. They demonstrated that FLNPs delivered mRNA much
more efciently, within 20 min, to rat and pig myocardium than sa-
line containing naked modRNA. In contrast, Sultana et al.
51
found
that encapsulating the modRNA with nanoparticles hindered its
effective translation, whereas naked modRNA in sucrose-citrate
buffer was translated very efciently, with the protein corresponding
to the reporter gene detected within 10 min in cardiac muscle. These
ndings were recently conrmed by Carlsson et al.,
47
who reported
efcient intracardiac transfection and protein expression following
the delivery of modRNA in saline citrate buffer. Translation efciency
in mouse heart was highest for 100 mg of naked modRNA delivered in
sucrose-citrate buffer.
51
Achieving the desired biodistribution of the
therapeutic gene in the heart remains one of the largest challenges fac-
ing us, but the modRNA used in this case was expressed in more than
20% of the LV, demonstrating the potential utility of this approach for
delivering disease-specic genes to the heart in cases of injury. The
use, in most cases, of intracardiac injection to deliver modRNA to
the heart may limit the biodistribution of the modRNA. A better, sys-
temic, non-invasive cardiac delivery method is therefore required.
With the increasing use of modRNA in preclinical studies (Table 1),
many researchers are now trying to increase its translational capacity
Figure 3. Use of Modified mRNA Therapy in
Prevention of Cardiac Remodeling
modRNA can be used to improve the condition of ischemic
injury by inducing cardiac and cardiovascular regeneration
and cardiac proliferation.
of modRNA in vivo. Conventional mRNA, con-
taining uridine, is associated with low translation
rates due to activation of the RNA-dependent
protein kinase (PKR), which then phosphorylates
translation initiation factor 2-alpha (elF2a). The phosphorylated
form, elf-2, binds to elF2B with a higher afnity, preventing the for-
mation of the elF2,GTP,Met-tRNA
i
tertiary complex required to
deliver mRNA to the ribosome, limiting the translation capacity of
the mRNA. However, we have shown that this process can be altered
by the complete replacement of uridine with 1-mJU, which results in
maximal translation and optimal expression kinetics for modRNA in
the heart. The 1-mJU modRNA displayed signicantly higher levels
of reporter mRNA expression in rat CMs in vitro and in mouse hearts
in vivo than the modRNAs used in previous studies. In a complemen-
tary study, Svitkin et al.
60
showed that mRNA with the 1-mJU modi-
cation resulted in much higher levels of reporter protein production,
due to attenuation of the elF2 phosphorylation-dependent inhibition
of translation and an increase in ribosome density on the mRNA.
Given the large amounts of modRNA needed to transfect large-size
heart, such as human heart, and the detrimental nature of the trans-
fection achieved by IM injection, the use of modRNA as a therapeutic
option in cardiac disease patients would require improvements in
modRNA translation and non-invasive transfection methods.
Future Directions in Cardiac modRNA Therapy
modRNA is a promising approach for the treatment of cardiovascular
disorders because it circumvents the key difculties presented by con-
ventional protein- and DNA-based gene therapy. Figure 3 summa-
rizes the ideal use of modRNA in prevention of cardiac remodeling.
Transfection with modRNA results in transient protein expression
and is, therefore, an attractive tool for therapeutic purposes for the
correction of cellular processes that do not require long-term protein
expression, such as cardiac regeneration, CM proliferation, and re-
programming. However, current modRNA approaches have no
inherent tissue- or cell-type-specic targeting capability in vivo,
whereas AAV gene therapy vectors can include tissue-specic pro-
moters.
6163
Improvements in targeting are, therefore, required,
because the activation of intracellular genes (e.g., transcription fac-
tors) in the wrong cell type can be detrimental. In addition, because
the IM injections may be stressful to the tissue, further research is
needed in development of non-invasive delivery methods. To ensure
targeted and non-invasive delivery of RNA, RNA aptamers, which
have great afnity to bind specic cell markers and are widely used
in cell-type-specic delivery of other RNA therapeutics like small
Molecular Therapy Vol. 27 No 4 April 2019 7
www.moleculartherapy.org
Review
Please cite this article in press as: Magadum et al., mRNA-Based Protein Replacement Therapy for the Heart, Molecular Therapy (2018), https://doi.org/
10.1016/j.ymthe.2018.11.018
interfering RNA (siRNA), can be used in conjunction with modRNA
to ensure its target-specic delivery.
64,65
Moreover, the transient
expression of modRNA may have made this tool ideal for approaches
targeting regeneration, but it is also the principal obstacle to the
replacement of long-term protein therapy by modRNA therapy in
the heart. Long-term controlled protein expression, with a method
of repeated systemic modRNA delivery, would make it possible to
use the modRNA delivery system to promote cardiac function in pre-
clinical or in clinical HF settings.
In our view, as research effects increase safety and scalability, and lead
to the development of cost-effective clinical-grade materials, robust
delivery methods, and lower treatment costs, modRNA technology
will become an excellent therapeutic agent to address experimental
and clinical needs to induce cardiac regeneration and promote car-
diac function in ischemic heart disease.
REFERENCES
1. Savarese, G., and Lund, L.H. (2017). Global public health burden of heart failure.
Card. Fail. Rev. 3,711.
2. Bui, A.L., Horwich, T.B., and Fonarow, G.C. (2011). Epidemiology and risk prole of
heart failure. Nat. Rev. Cardiol. 8,3041.
3. Dargie, H. (2005). Heart failure post-myocardial infarction: a review of the issues.
Heart 91 (Suppl 2 ), ii3ii6, discussion ii31, ii43ii48.
4. Go, A.S., Mozaffarian, D., Roger, V.L., Benjamin, E.J., Berry, J.D., Blaha, M.J., Dai, S.,
Ford, E.S., Fox, C.S., Franco, S., et al.; American Heart Association Statistics
Committee and Stroke Statistics Subcommittee (2014). Heart disease and stroke sta-
tistics2014 update: a report from the American Heart Association. Circulation 129,
e28e292.
5. Hajjar, R.J., Zsebo, K., Deckelbaum, L., Thompson, C., Rudy, J., Yaroshinsky, A., Ly,
H., Kawase, Y., Wagner, K., Borow, K., et al. (2008). Design of a phase 1/2 trial of
intracoronary administration of AAV1/SERCA2a in patients with heart failure.
J. Card. Fail. 14, 355367.
6. Greenberg, B., Butler, J., Felker, G.M., Ponikowski, P., Voors, A.A., Desai, A.S.,
Barnard, D., Bouchard, A., Jaski, B., Lyon, A.R., et al. (2016). Calcium upregulation
by percutaneous administration of gene therapy in patients with cardiac disease
(CUPID 2): a randomised, multinational, double-blind, placebo-controlled, phase
2b trial. Lancet 387, 11781186.
7. Porrello, E.R., Mahmoud, A.I., Simpson, E., Hill, J.A., Richardson, J.A., Olson, E.N.,
and Sadek, H.A. (2011). Transient regenerative potential of the neonatal mouse heart.
Science 331, 10781080.
8. Engel, F.B., Schebesta, M., Duong, M.T., Lu, G., Ren, S., Madwed, J.B., Jiang, H.,
Wang, Y., and Keating, M.T. (2005). p38 MAP kinase inhibition enables proliferation
of adult mammalian cardiomyocytes. Genes Dev. 19, 11751187.
9. Bassat, E., Mutlak, Y.E., Genzelinakh, A., Shadrin, I.Y., Baruch Umansky, K. , Yifa, O.,
Kain, D., Rajchman, D., Leach, J., Riabov Bassat, D., et al. (2017). The extracellular
matrix protein agrin promotes heart regeneration in mice. Nature 547, 179184.
10. Wei, K., Serpooshan, V., Hurtado, C., Diez-Cuñado, M., Zhao, M., Maruyama, S.,
Zhu, W., Fajardo, G., Noseda, M., Nakamura, K., et al. (2015). Epicardial FSTL1
reconstitution regenerates the adult mammalian heart. Nature 525, 479485.
11. Engel, F.B., Hsieh, P.C., Lee, R.T., and Keating, M.T. (2006). FGF1/p38 MAP kinase
inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and rescues func-
tion after myocardial infarction. Proc. Natl. Acad. Sci. USA 103, 1554615551.
12. Kühn, B., del Monte, F., Hajjar, R.J., Chang, Y.S., Lebeche, D., Arab, S., and Keating,
M.T. (2007). Periostin induces proliferation of differentiated cardiomyocytes and
promotes cardiac repair. Nat. Med. 13, 962969.
13. del Monte, F., Hajjar, R.J., and Harding, S.E. (2001). Overwhelming evidence of the
benecial effects of SERCA gene transfer in heart failure. Circ. Res. 88, E66E67.
14. Kho, C., Lee, A., Jeong, D., Oh, J.G., Chaanine, A.H., Kizana, E., Park, W.J., and
Hajjar, R.J. (2011). SUMO1-dependent modulation of SERCA2a in heart failure.
Nature 477,601605.
15. Ieda, M., Fu, J.D., Delgado-Olguin, P., Vedantham, V., Hayashi, Y., Bruneau, B.G.,
and Srivastava, D. (2010). Direct reprogramming of broblasts into functional cardi-
omyocytes by dened factors. Cell 142,375386.
16. Song, K., Nam, Y.J., Luo, X., Qi, X., Tan, W., Huang, G.N., Acharya, A., Smith, C.L.,
Tallquist, M.D., Neilson, E.G., et al. (2012). Heart repair by reprogramming non-
myocytes with cardiac transcription factors. Nature 485, 599604.
17. Henry, T.D., Annex, B.H., McKendall, G.R., Azrin, M.A., Lopez, J.J., Giordano, F.J.,
Shah, P.K., Willerson, J.T., Benza, R.L., Berman, D.S., et al.; VIVA Investigators
(2003). The VIVA trial: vascular endothelial growth factor in ischemia for vascular
angiogenesis. Circulation 107, 13591365.
18. Hendel, R.C., Henry, T.D., Rocha-Singh, K., Isner, J.M., Kereiakes, D.J., Giordano,
F.J., Simons, M., and Bonow, R.O. (2000). Effect of intracoronary recombinant
human vascular endothelial growth factor on myocardial perfusion: evidence for a
dose-dependent effect. Circulation 101, 118121.
19. Sato, K., Wu, T., Laham, R.J., Johnson, R.B., Douglas, P., Li, J., Sellke, F.W., Bunting,
S., Simons, M., and Post, M.J. (2001). Efcacy of intracoronary or intravenous
VEGF165 in a pig model of chronic myocardial ischemia. J. Am. Coll. Cardiol. 37,
616623.
20. Hedman, M., Hartikainen, J., Syvänne, M., Stjernvall, J., Hedman, A., Kivelä, A.,
Vanninen, E., Mussalo, H., Kauppila, E., Simula, S., et al. (2003). Safety and feasibility
of catheter-based local intracoronary vascular endothelial growth factor gene transfer
in the prevention of postangioplasty and in-stent restenosis and in the treatment of
chronic myocardial ischemia: phase II results of the Kuopio Angiogenesis Trial
(KAT). Circulation 107, 26772683.
21. Stewart, D.J., Kutryk, M.J., Fitchett, D., Freeman, M., Camack, N., Su, Y., Della Siega,
A., Bilodeau, L., Burton, J.R., Proulx, G., and Radhakrishnan, S.; NORTHERN Trial
Investigators (2009). VEGF gene therapy fails to improve perfusion of ischemic
myocardium in patients with advanced coronary disease: results of the
NORTHERN trial. Mol. Ther. 17, 11091115.
22. Stewart, D.J., Hilton, J.D., Arnold, J.M., Gregoire, J., Rivard, A., Archer, S.L.,
Charbonneau, F., Cohen, E., Curtis, M., Buller, C.E., et al. (2006). Angiogenic gene
therapy in patients with nonrevascularizable ischemic heart disease: a phase 2 ran-
domized, controlled trial of AdVEGF(121) (AdVEGF121) versus maximum medical
treatment. Gene Ther. 13, 15031511.
23. Losordo, D.W., Vale, P.R., Symes, J.F., Dunnington, C.H., Esakof, D.D., Maysky, M.,
Ashare, A.B., Lathi, K., and Isner, J.M. (1998). Gene therapy for myocardial angiogen-
esis: initial clinical results with direct myocardial injection of phVEGF165 as sole
therapy for myocardial ischemia. Circulation 98, 28002804.
24. French, B.A., Mazur, W., Geske, R.S., and Bolli, R. (1994). Direct in vivo gene transfer
into porcine myocardium using replication-decient adenoviral vectors. Circulation
90, 24142424.
25. Muruve, D.A. (2004). The innate immune response to adenovirus vectors. Hum.
Gene Ther. 15, 11571166.
26. Lin, Z., von Gise, A., Zhou, P., Gu, F., Ma, Q., Jiang, J., Yau, A.L., Buck, J.N., Gouin,
K.A., van Gorp, P.R., et al. (2014). Cardiac-specic YAP activation improves cardiac
function and survival in an experimental murine MI model. Circ. Res. 115, 354363.
27. Chu, D., Sullivan, C.C., Weitzman, M.D., Du, L., Wolf, P.L., Jamieson, S.W., and
Thistlethwaite, P.A. (2003). Direct comparison of efciency and stability of gene
transfer into the mammalian heart using adeno-associated virus versus adenovirus
vectors. J. Thorac. Cardiovasc. Surg. 126, 671679.
28. Vassalli, G., Büeler, H., Dudler, J., von Segesser, L.K., and Kappenberger, L. (2003).
Adeno-associated virus (AAV) vectors achieve prolonged transgene expression in
mouse myocardium and arteries in vivo: a comparative study with adenovirus
vectors. Int. J. Cardiol. 90, 229238.
29. Mingozzi, F., and High, K.A. (2013). Immune responses to AAV vectors: overcoming
barriers to successful gene therapy. Blood 122,2336.
30. Eulalio, A., Mano, M., Dal Ferro, M., Zentilin, L., Sinagra, G., Zacchigna, S., and
Giacca, M. (2012). Functional screening identies miRNAs inducing cardiac regen-
eration. Nature 492, 376381.
8 Molecular Therapy Vol. 27 No 4 April 2019
www.moleculartherapy.org
Review
Please cite this article in press as: Magadum et al., mRNA-Based Protein Replacement Therapy for the Heart, Molecular Therapy (2018), https://doi.org/
10.1016/j.ymthe.2018.11.018
31. Hermans-Beijnsberger, S., van Bilsen, M., and Schroen, B. (2018). Long non-coding
RNAs in the failing heart and vasculature. Noncoding RNA Res. 3, 118130.
32. Rane, S., He, M., Sayed, D., Vashistha, H., Malhotra, A., Sadoshima, J., Vatner, D.E.,
Vatner, S.F., and Abdellatif, M. (2009). Downregulation of miR-199a derepresses
hypoxia-inducible factor-1alpha and Sirtuin 1 and recapitulates hypoxia precondi-
tioning in cardiac myocytes. Circ. Res. 104, 879886.
33. Lin, Z., and Pu, W.T. (2014). Strategies for cardiac regeneration and repair. Sci.
Transl. Med. 6, 239rv1.
34. Wolff, J.A., Malone, R.W., Williams, P., Chong, W., Acsadi, G., Jani, A., and Felgner,
P.L. (1990). Direct gene transfer into mouse muscle in vivo. Science 247, 14651468.
35. Karikó, K., Muramatsu, H., Welsh, F.A., Ludwig, J., Kato, H., Akira, S., and
Weissman, D. (2008). Incorporation of pseudouridine into mRNA yields superior
nonimmunogenic vector with increased translational capacity and biological stability.
Mol. Ther. 16, 18331840.
36. Kormann, M.S., Hasenpusch, G., Aneja, M.K., Nica, G., Flemmer, A.W., Herber-
Jonat, S., Huppmann, M., Mays, L.E., Illenyi, M., Schams, A., et al. (2011).
Expression of therapeutic proteins after delivery of chemically modied mRNA in
mice. Nat. Biotechnol. 29, 154157.
37. Kondrat, J., Sultana, N., and Zangi, L. (2017). Synthesis of modied mRNA for
myocardial delivery. Methods Mol. Biol. 1521, 127138.
38. Mockey, M., Gonçalves, C., Dupuy, F.P., Lemoine, F.M., Pichon, C., and Midoux, P.
(2006). mRNA transfection of dendritic cells: synergistic effect of ARCA mRNA
capping with Poly(A) chains in cis and in trans for a high protein expression level.
Biochem. Biophys. Res. Commun. 340, 10621068.
39. Formiga, F.R., Tamayo, E., Simón-Yarza, T., Pelacho, B., Prósper, F., and Blanco-
Prieto, M.J. (2012). Angiogenic therapy for cardiac repair based on protein delivery
systems. Heart Fail. Rev. 17, 449473.
40. Hinkel, R., Trenkwalder, T., and Kupatt, C. (2011). Gene therapy for ischemic heart
disease. Expert Opin. Biol. Ther. 11, 723737.
41. Simón-Yarza, T., Formiga, F.R., Tamayo, E., Pelacho, B., Prosper, F., and Blanco-
Prieto, M.J. (2012). Vascular endothelial growth factor-delivery systems for cardiac
repair: an overview. Theranostics 2, 541552.
42. Wu, J., Zeng, F., Huang, X.P., Chung, J.C., Konecny, F., Weisel, R.D., and Li, R.K.
(2011). Infarct stabilization and cardiac repair with a VEGF-conjugated, injectable
hydrogel. Biomaterials 32, 579586.
43. Gao, J., Liu, J., Gao, Y., Wang, C., Zhao, Y., Chen, B., Xiao, Z., Miao, Q., and Dai, J.
(2011). A myocardial patch made of collagen membranes loaded with collagen-bind-
ing human vascular endothelial growth factor accelerates healing of the injured rabbit
heart. Tissue Eng. Part A 17, 27392747.
44. Lin, Y.D., Luo, C.Y., Hu, Y.N., Yeh, M.L., Hsueh, Y.C., Chang, M.Y., Tsai, D.C.,
Wang, J.N., Tang, M.J., Wei, E.I., et al. (2012). Instructive nanober scaffolds with
VEGF create a microenvironment for arteriogenesis and cardiac repair. Sci. Transl.
Med. 4, 146ra109.
45. 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. (2013). Modied mRNA directs the fate of heart
progenitor cells and induces vascular regeneration after myocardial infarction. Nat.
Biotechnol. 31, 898907.
46. Lui, K.O., Zangi, L., Silva, E.A., Bu, L., Sahara, M., Li, R.A., Mooney, D.J., and Chien,
K.R. (2013). Driving vascular endothelial cell fate of human multipotent Isl1+ heart
progenitors with VEGF modied mRNA. Cell Res. 23, 11721186.
47. Carlsson, L., Clarke, J.C., Yen, C., Gregoire, F., Albery, T., Billger, M., Egnell, A.C.,
Gan, L.M., Jennbacken, K., Johansson, E., et al. (2018). Biocompatible, puried
VEGF-A mRNA improves cardiac function after intracardiac injection 1 week
post-myocardial infarction in Swine. Mol. Ther. Methods Clin. Dev. 9, 330346.
48. Magadum, A., Singh, N., Kurian, A.A., Sharkar, M.T.K., Chepurko, E., and Zangi, L.
(2018). Ablation of a single N-glycosylation site in human FSTL 1 induces cardio-
myocyte proliferation and cardiac regeneration. Mol. Ther. Nucleic Acids 13,
133143.
49. Ren, G., Dewald, O., and Frangogiannis, N.G. (2003). Inammatory mechanisms in
myocardial infarction. Curr. Drug Targets Inamm. Allergy 2, 242256.
50. Hadas, Y., Katz, M.G., Bridges, C.R., and Zangi, L. (2017). Modied mRNA as a ther-
apeutic tool to induce cardiac regeneration in ischemic heart disease. Wiley
Interdiscip. Rev. Syst. Biol. Med. 9, e1367.
51. Sultana, N., Magadum, A., Hadas, Y., Kondrat, J., Singh, N., Youssef, E., Calderon, D.,
Chepurko, E., Dubois, N., Hajjar, R.J., and Zangi, L. (2017). Optimizing cardiac de-
livery of modied mRNA. Mol. Ther. 25, 13061315.
52. Huang, C.L., Leblond, A.L., Turner, E.C., Kumar, A.H., Martin, K., Whelan, D.,
OSullivan, D.M., and Caplice, N.M. (2015). Synthetic chemically modied mrna-
based delivery of cytoprotective factor promotes early cardiomyocyte survival post-
acute myocardial infarction. Mol. Pharm. 12, 991996.
53. 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. (2017). Insulin-like growth factor 1 receptor-depen-
dent pathway drives epicardial adipose tissue formation after myocardial injury.
Circulation 135,5972.
54. Nagy, E., Jermendy, A.L., Merkely, B., and Maurovich-Horvat, P. (2017). Clinical
importance of epicardial adipose tissue. Arch. Med. Sci. 13, 864874.
55. Crozat, K., and Beutler, B. (2004). TLR7: a new sensor of viral infection. Proc. Natl.
Acad. Sci. USA 101, 68356836.
56. Andries, O., Mc Cafferty, S., De Smedt, S.C., Weiss, R., Sanders, N.N., and Kitada, T.
(2015). N(1)-methylpseudouridine-incorporated mRNA outperforms pseudouri-
dine-incorporated mRNA by providing enhanced protein expression and reduced
immunogenicity in mammalian cell lines and mice. J. Control. Release 217, 337344.
57. Singh, R.D., Hillestad, M.L., Livia, C., Li, M., Alekseev, A.E., Witt, T.A., Stalboerger,
P.G., Yamada, S., Terzic, A., and Behfar, A. (2018). M
3
RNA drives targeted gene de-
livery in acute myocardial infarction. Tissue Eng. Part A. , Published online
September 21, 2018. https://doi.org/10.1089/ten.tea.2017.0445.
58. Turnbull, I.C., Eltoukhy, A.A., Fish, K.M., Nonnenmacher, M., Ishikaw a, K., Chen, J.,
Hajjar, R.J., Anderson, D.G., and Costa, K.D. (2016). Myocardial delivery of lipidoid
nanoparticle carrying modRNA induces rapid and transient expression. Mol. Ther.
24,6675.
59. Turnbull, I.C., Eltoukhy, A.A., Anderson, D.G., and Costa, K.D. (2017). Lipidoid
mRNA nanoparticles for myocardial delivery in rodents. Methods Mol. Biol. 1521,
153166.
60. Svitkin, Y.V., Cheng, Y.M., Chakraborty, T., Presnyak, V., John, M., and Sonenberg,
N. (2017). N1-methyl-pseudouridine in mRNA enhances translation through
eIF2a-dependent and independent mechanisms by increasing ribosome density.
Nucleic Acids Res. 45, 60236036.
61. Papadakis, E.D., Nicklin, S.A., Baker, A.H., and White, S.J. (2004). Promoters and
control elements: designing expression cassettes for gene therapy. Curr. Gene Ther.
4,89113.
62. Chen, S.J., Johnston, J., Sandhu, A., Bish, L.T., Hovhannisyan, R., Jno-Charles, O.,
Sweeney, H.L., and Wilson, J.M. (2013). Enhancing the utility of adeno-associated
virus gene transfer through inducible tissue-specic expression. Hum. Gene Ther.
Methods 24, 270278.
63. Zacchigna, S., Zentilin, L., and Giacca, M. (2014). Adeno-associated virus vectors as
therapeutic and investigational tools in the cardiovascular system. Circ. Res. 114,
18271846.
64. Li, J., Youse, K., Ding, W., Singh, J., and Shehadeh, L.A. (2017). Osteopontin RNA
aptamer can prevent and reverse pressure overload-induced heart failure. Cardiovasc.
Res. 113, 633643.
65. Zhou, J., and Rossi, J.J. (2014). Cell-type-specic, aptamer-functionalized agents for
targeted disease therapy. Mol. Ther. Nucleic Acids 3, e169.
Molecular Therapy Vol. 27 No 4 April 2019 9
www.moleculartherapy.org
Review
Please cite this article in press as: Magadum et al., mRNA-Based Protein Replacement Therapy for the Heart, Molecular Therapy (2018), https://doi.org/
10.1016/j.ymthe.2018.11.018
... Em células cardíacas, o modRNA exibe tradução rápida, pico de expressão entre 12 e 48 h e cinética transitória. Sua expressão controlada e incapacidade de integração ao genoma tornam o modRNA uma plataforma promissora de terapia genética para várias doenças, incluindo condições cardíacas 41,42 . ...
Chapter
As doenças cardiovasculares (DCVs) continuam sendo a principal causa de morbidade e mortalidade no mundo, motivando a busca por terapias mais eficazes e personalizadas. Nos últimos anos, as terapias baseadas em RNA têm emergido como uma estratégia promissora para o tratamento de diversas condições cardiovasculares. Essas abordagens envolvem o uso de diferentes tipos de RNA, como RNA mensageiro (mRNA), RNA de interferência (RNAi), microRNA (miRNA) e oligonucleotídeos antisense, para modular a expressão gênica de maneira precisa. O avanço da biotecnologia e o desenvolvimento de sistemas de entrega mais seguros e eficientes permitiram a aplicação clínica de algumas dessas terapias, com resultados iniciais encorajadores. Entre os alvos terapêuticos estudados estão genes relacionados à dislipidemia, hipertensão arterial, insuficiência cardíaca e aterosclerose. Um exemplo notável é o uso de RNAi para inibir a expressão da proteína PCSK9, envolvida na regulação dos níveis de colesterol LDL, demonstrando eficácia na redução de eventos cardiovasculares. Além disso, terapias com miRNA têm mostrado potencial para regular processos inflamatórios, remodelamento cardíaco e angiogênese, fundamentais na fisiopatologia das DCVs. Apesar do progresso, desafios como a imunogenicidade, a estabilidade das moléculas de RNA e a especificidade da entrega ainda limitam a aplicação generalizada dessas terapias. Ensaios clínicos em andamento buscam validar a segurança e a eficácia dessas abordagens em populações maiores e em diferentes contextos clínicos. A perspectiva futura inclui o desenvolvimento de terapias combinadas, uso de RNA em edição gênica e estratégias personalizadas com base no perfil genético dos pacientes. Em resumo, as terapias com RNA representam uma fronteira inovadora na cardiologia, com o potencial de transformar a abordagem terapêutica das DCVs. A compreensão aprofundada dos mecanismos moleculares envolvidos e a superação dos desafios técnicos são fundamentais para a consolidação dessas terapias como parte do arsenal clínico na prática médica cardiovascular.
... The transient nature of gene expression achieved with modRNA administration makes it an ideal candidate for treating myocardial infarction (MI) and IHD. modRNA can provide temporary, high-level expression of therapeutic proteins that promote cardiomyocyte proliferation, reduce post-injury remodeling, and enhance cardiovascular regeneration by increasing capillary density after a heart attack [16]. Despite its potential, the short expression period and high production cost of modRNA necessitate the identification of optimized 5 ′ UTRs to enhance translation efficiency and therapeutic effects in cardiac tissue. ...
Article
Full-text available
Background/Objectives: Modified messenger RNA (modRNA) is a promising gene delivery method used to upregulate genes in cardiac tissue, with applications in both clinical and preclinical settings to prevent cardiac remodeling after ischemic injury. The 5′ untranslated region (5′UTR) plays a crucial role in regulating the translation efficiency of mRNA into functional proteins. Due to the high production cost and short half-life of modRNA, it is essential to identify novel 5′UTR designs that enhance modRNA translation in the heart. Methods: Here, we present an artificial 5′UTR, termed “Top Heart 5′UTR”, designed based on ribonucleotide frequency analyses of 1000 genes highly expressed in the heart. This novel artificial 5′UTR contains a unique 20-nucleotide sequence, consisting of 11 previously uncharacterized nucleotides (CCCCCGCCCCC) and 9 well-described nucleotides from the Kozak sequence upstream of the start codon (ATG). Results: This design significantly improves modRNA translation efficiency in cardiomyocytes (CMs) and heart cells both in vitro and in vivo. Specifically, the Top Heart 5′UTR increases translation efficiency by approximately 30–60% in both mouse and human CMs compared to a standard 5′UTR control. Moreover, the artificial 5′UTR induces a 2–2.5 times higher translation of modRNA in the mouse heart 24 and 48 h post-delivery. Conclusions: Our findings may contribute to the development of a superior modRNA platform for use in preclinical and clinical studies, potentially allowing reduced dosages or increased gene expression at the same dosage level. This approach can be extended to identify optimized 5′UTRs for various cell types or organs, including applications in cancer therapies.
... This impairment in cardiac function further compromises renal perfusion, creating a vicious cycle that can trigger or worsen CKD, thereby exacerbating CKM syndrome [81,82]. Currently, the ability to regenerate lost cardiomyocytes remains elusive, highlighting the critical need for innovative therapeutic strategies [83]. RNA-based therapies have emerged as a promising avenue, targeting various pathways involved in cell death, revascularization, and cardiomyocyte generation [4]. ...
Article
Full-text available
Cardio-Kidney-Metabolic (CKM) Syndrome involves metabolic, kidney, and cardiovascular dysfunction, disproportionately affecting disadvantaged groups. Its staging (0–4) highlights the importance of early intervention. While current management targets hypertension, heart failure, dyslipidemia, and diabetes, RNA-based therapies offer innovative solutions by addressing molecular mechanisms of CKM Syndrome. Emerging RNA treatments, including antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs), show promise in slowing disease progression across CKM stages. For example, ASOs and siRNAs targeting ApoC-III and ANGPTL3 reduce triglycerides and LDL cholesterol, while siRNAs improve blood pressure control by targeting the renin–angiotensin–aldosterone system. Obesity treatments leveraging miRNAs and circRNAs tackle a key CKM risk factor. In heart failure and diabetes, RNA-based therapies improve cardiac function and glucose control, while early kidney disease trials show potential for RNAi in acute injury. Further research is essential to refine these therapies and ensure equitable access.
... Messenger RNA (mRNA)-based therapy has recently emerged as a secure and highly efficient approach with broad applications ranging from vaccination and protein replacement to gene editing [23,24]. This therapeutic modality boasts several conceptual advantages over protein or other nucleic acid-based methods, including the absence of insertional mutagenesis, sustained protein production for extended periods, and the lack of a requirement for nuclear entry of mRNA molecules. ...
Article
Full-text available
Spinal cord injury (SCI), characterized by the disruption of neural pathways and an increase in inflammatory cell infiltration, leads to profound and lasting neurological deficits, with a high risk of resulting in permanent disability. Currently, the therapeutic landscape for SCI is notably sparse, with limited effective treatment options available. Methylprednisolone (MP), a widely used clinical anti-inflammatory agent for SCI, requires administration in high doses that are associated with significant adverse effects. In this study, we introduce an innovative approach by substituting cholesterol with MP to engineer a novel Lipid Nanoparticle (MP-LNP). This strategy aims to enhance the localization and concentration of MP at the injury site, thereby amplifying its therapeutic efficacy while mitigating systemic side effects. Furthermore, we explore the integration of C3 transferase mRNA into MP-LNPs. C3 transferase, a potent inhibitor of the RhoA pathway, has shown promise in facilitating neurological recovery in animal models of SCI and is currently being evaluated in clinical trials. The novel formulation, MP-LNP-C3, is designed for direct administration to the injury site during decompression surgery, offering a targeted therapeutic modality for SCI. Our findings reveal several significant advantages of this approach: Firstly, the incorporation of C3 transferase mRNA into MP-LNPs does not compromise the structural integrity of the nanoparticles, ensuring efficient mRNA expression within the spinal cord. Secondly, the MP-LNP formulation effectively attenuates inflammation and reduces the adverse effects associated with high-dose MP treatment in the acute phase of SCI. Lastly, MP-LNP-C3 demonstrates notable neuroprotective properties and promotes enhanced recovery of motor function in SCI mouse models. Together, these results underscore the potential of this innovative LNP-based therapy as a promising avenue for advancing the treatment of clinical SCI. Graphical Abstract
Chapter
Messenger RNA (mRNA) plays a pivotal role in the central dogma of molecular biology, serving as an intermediary between DNA and protein synthesis. Within cells, DNA encodes genetic information that is transcribed into mRNA. This mRNA then carries this genetic code from the nucleus to the cytoplasm, where it directs the synthesis of proteins through a process known as translation. mRNA molecules are thus essential in translating the genetic instructions stored in DNA into functional proteins that perform various vital functions within organisms. Initially, mRNA faced scepticism as a viable therapeutic due to concerns about its stability and challenges in scaling up production. Recent advancements in technology and a deeper understanding of biomolecular processes have largely dispelled these doubts. Current research is heavily focused on overcoming these initial obstacles. Consequently, mRNA is now increasingly recognized for its potential to revolutionize therapeutic strategies such as immunotherapy, regenerative medicine, vaccination, and gene editing. However, realizing the full therapeutic promise of mRNA hinges on improving its production efficiency, stability, and precise delivery to target cells. To address these critical issues, substantial research efforts are underway to explore innovative solutions. These efforts aim to enhance mRNA stability and refine delivery mechanisms, thereby maximizing its therapeutic efficacy.
Article
Myocardial infarction (MI) is a pathological state characterized by persistent ischemia of the heart. Following MI, the structural and functional remodeling of the myocardium and vasculature involves oxidative stress and mitochondrial dysfunction, which exacerbate myocardial injury. Currently, there are limited effective treatments available to alleviate MI‐induced damage. Vascular endothelial growth factor‐mimetic (QK) peptides and mitochondria‐targeted Szeto–Schiller (SS31) peptides have been extensively investigated for their therapeutic potential in various ischemic cardiomyopathies. However, traditional topical agents used in myocardial ischemia treatment suffer from limitations such as transient retention or undesirable diffusion of the drug. Consequently, a controlled drug delivery system capable of delivering QK and SS31 has gained significant attention for repair. In this study, we constructed self‐assembled nanofibrous hydrogels incorporating QK and SS31 with customizable peptide amphiphilic (PA) molecules, resulting in PA1‐QK and PA2‐SS31 formulations. In vitro experiments demonstrated that both QK and SS31 effectively inhibited mitochondrial damage and apoptosis in a cellular hypoxia/reoxygenation (H/R) model. In vivo studies using a mouse MI model revealed that PA1‐QK and PA2‐SS31 significantly promoted vascular regeneration, attenuated mitochondrial dysfunction and apoptosis, and facilitated the recovery of cardiac structure and function. These results suggest that PA1‐QK and PA2‐SS31‐loaded self‐assembled nanofiber hydrogels represent an effective drug delivery system for promoting regenerative repair of myocardium and blood vessels following MI.
Article
Full-text available
Developing new lipid nanoparticle (LNP) formulations typically involves reconstruction from separate elements followed by rigorous purification steps, contributing to drawn‐out drug discovery processes. Membrane‐intercalating conjugated oligoelectrolytes (COEs) are water‐soluble molecules featuring a conjugated backbone and peripheral ionic groups, specifically designed to spontaneously integrate into lipid bilayers. Herein, an ex situ strategy to “dope” the representative COE‐S6 into pre‐formed messenger RNA‐LNPs (mRNA‐LNPs) is presented, exploiting its spontaneous membrane intercalation property through a straightforward add‐and‐mix procedure. Incorporating 0.2% COE‐S6 into mRNA‐LNPs relative to lipid content reduced particle size from 84.5 ± 1 to 67.9 ± 0.8 nm, elevated cellular uptake, and improved endosomal escape. These traits culminate in an increase in in cellula transfection from 24.2 ± 1.6% to 98.7 ± 0.6%. When injected intravenously into healthy BALB/c mice, the optimized COE‐S6‐doped mRNA‐LNPs boost in vivo luciferase expression by 1.75‐fold. Additionally, COE‐S6‐doped mRNA‐LNPs exhibit fluorogenic properties, enabling intracellular mechanistic studies via confocal microscopy. This simple method enhances the properties of mRNA‐LNPs with minimal COE quantities, offering a novel strategy to improve existing LNP formulations and provide optical reporting capabilities, essential for expediting drug discovery and delivery.
Article
Full-text available
Adult mammalian hearts have a very limited regeneration capacity, due largely to a lack of cardiomyocyte (CM) proliferation. It was recently reported that epicardial, but not myocardial, follistatin-like 1 (Fstl1) activates CM proliferation and cardiac regeneration after myocardial infarction (MI). Furthermore, bacterially synthesized human FSTL 1 (hFSTL1) was found to induce CM proliferation, whereas hFSTL1 synthesized in mammals did not, suggesting that post-translational modifications (e.g., glycosylation) of the hFSTL1 protein affect its regenerative activity. We used modified mRNA (modRNA) technology to investigate the possible role of specific hFSTL1 N-glycosylation sites in the induction, by hFSTL1, of CM proliferation and cardiac regeneration. We found that the mutation of a single site (N180Q) was sufficient and necessary to increase the proliferation of rat neonatal and mouse adult CMs in vitro and after MI in vivo, respectively. A single administration of the modRNA construct encoding the N180Q mutant significantly increased cardiac function, decreased scar size, and increased capillary density 28 days post-MI. Overall, our data suggest that the delivery of N180Q hFSTL1 modRNA to the myocardium can mimic the beneficial effect of epicardial hFSTL1, triggering marked CM proliferation and cardiac regeneration in a mouse MI model.
Article
Full-text available
Following completion of the human genome, it became evident that the majority of our DNA is transcribed into non-coding RNAs (ncRNAs) instead of protein-coding messenger RNA. Deciphering the function of these ncRNAs, including both small- and long non-coding RNAs (lncRNAs), is an emerging field of research. LncRNAs have been associated with many disorders and a number have been identified as key regulators in the development and progression of disease, including cardiovascular disease (CVD). CVD causes millions of deaths worldwide, annually. Risk factors include coronary artery disease, high blood pressure and ageing. In this review, we will focus on the roles of lncRNAs in the cellular and molecular processes that underlie the development of CVD: cardiomyocyte hypertrophy, fibrosis, inflammation, vascular disease and ageing. Finally, we discuss the biomarker and therapeutic potential of lncRNAs.
Article
Full-text available
mRNA can direct dose-dependent protein expression in cardiac muscle without genome integration, but to date has not been shown to improve cardiac function in a safe, clinically applicable way. Herein, we report that a purified and optimized mRNA in a biocompatible citrate-saline formulation is tissue specific, long acting, and does not stimulate an immune response. In small- and large-animal, permanent occlusion myocardial infarction models, VEGF-A 165 mRNA improves systolic ventricular function and limits myocardial damage. Following a single administration a week post-infarction in mini pigs, left ventricular ejection fraction, inotropy, and ventricular compliance improved, border zone arteriolar and capillary density increased, and myocardial fibrosis decreased at 2 months post-treatment. Purified VEGF-A mRNA establishes the feasibility of improving cardiac function in the sub-acute therapeutic window and may represent a new class of therapies for ischemic injury.
Article
Full-text available
The adult mammalian heart is non-regenerative due to the post-mitotic nature of cardiomyocytes. The neonatal mouse heart can regenerate, but only for the first week of life. Here we show that changes in the composition of the extracellular matrix (ECM) during this week can affect cardiomyocyte growth and differentiation in mice. We identify Agrin, a component of neonatal ECM, as required for the full regenerative capacity of neonatal mouse hearts. In vitro, recombinant Agrin promotes the division of mouse and human iPSC-derived cardiomyocytes via a mechanism that involves the disassembly of the dystrophin glycoprotein complex and Yap and ERK-mediated signaling. In vivo, a single administration of Agrin promotes cardiac regeneration in adult mice after myocardial infarction, although the degree of cardiomyocyte proliferation observed in this model suggests additional therapeutic mechanisms. Collectively, we uncover a new inducer of mammalian heart regeneration, highlighting fundamental roles of the ECM in cardiac repair.
Article
Full-text available
Modified mRNA (modRNA) is a new technology in the field of somatic gene transfer that has been used for the delivery of genes into different tissues, including the heart. Our group and others have shown that modRNAs injected into the heart are robustly translated into the encoded protein and can potentially improve outcome in heart injury models. However, the optimal compositions of the modRNA and the reagents necessary to achieve optimal expression in the heart have not been characterized yet. In this study, our aim was to elucidate those parameters by testing different nucleotide modifications, modRNA doses, and transfection reagents both in vitro and in vivo in cardiac cells and tissue. Our results indicate that optimal cardiac delivery of modRNA is with N1-Methylpseudouridine-5'-Triphosphate nucleotide modification and achieved using 0.013 μg modRNA/mm(2)/500 cardiomyocytes (CMs) transfected with positively charged transfection reagent in vitro and 100 μg/mouse heart (1.6 μg modRNA/μL in 60 μL total) sucrose-citrate buffer in vivo. We have optimized the conditions for cardiac delivery of modRNA in vitro and in vivo. Using the described methods and conditions may allow for successful gene delivery using modRNA in various models of cardiovascular disease.
Article
Full-text available
Certain chemical modifications confer increased stability and low immunogenicity to in vitro transcribed mRNAs, thereby facilitating expression of therapeutically important proteins. Here, we demonstrate that N1-methyl-pseudouridine (N1mΨ) outperforms several other nucleoside modifications and their combinations in terms of translation capacity. Through extensive analysis of various modified transcripts in cell-free translation systems, we deconvolute the different components of the effect on protein expression independent of mRNA stability mechanisms. We show that in addition to turning off the immune/eIF2α phosphorylation-dependent inhibition of translation, the incorporated N1mΨ nucleotides dramatically alter the dynamics of the translation process by increasing ribosome pausing and density on the mRNA. Our results indicate that the increased ribosome loading of modified mRNAs renders them more permissive for initiation by favoring either ribosome recycling on the same mRNA or de novo ribosome recruitment.
Article
Full-text available
Ischemic heart disease ( IHD ) is a leading cause of morbidity and mortality in developed countries. Current pharmacological and interventional therapies provide significant improvement in the life quality of patient; however, they are mostly symptom‐oriented and not curative. A high disease and economic burden of IHD requires the search for new therapeutic strategies to significantly improve patients’ prognosis and quality of life. One of the main challenges during IHD is the massive loss of cardiomyocytes that possess minimal regenerative capacity. Recent understanding of the pathophysiological mechanisms underlying IHD , as well as new therapeutic approaches provide new hope for patients suffering from IHD . Synthetic modified mRNA ( modRNA ) is a new gene delivery vector that is increasingly used in in vivo applications. modRNA is a relatively stable, non‐immunogenic, highly‐expressed molecule that has been shown to mediate high and transient expression of proteins in different type of cells and tissues including cardiomyocytes. modRNA properties, together with its expression kinetics in the heart make it an attractive option for the treatment of IHD , especially after myocardial infarction. In this review we discuss the role of gene therapy in cardiac regeneration as an approach to treat IHD ; traditional and innovative gene delivery methods; and focus specifically on modRNA structure, mode of delivery, and its use for the induction of endogenous regenerative capacity, mainly in the context of IHD . WIREs Syst Biol Med 2017, 9:e1367. doi: 10.1002/wsbm.1367 This article is categorized under: Developmental Biology > Stem Cell Biology and Regeneration Translational, Genomic, and Systems Medicine > Therapeutic Methods Translational, Genomic, and Systems Medicine > Translational Medicine
Article
Impact statement: The M3RNA (microencapsulated modified messenger RNA) platform is an approach to deliver messenger RNA (mRNA) in vivo, achieving a nonintegrating and viral-free approach to gene therapy. This technology was, in this study, tested for its utility in the myocardium, providing a unique avenue for targeted gene delivery into the freshly infarcted myocardial tissue. This study provides the evidentiary basis for the use of M3RNA in the heart through depiction of its performance in cultured cells, healthy rodent myocardium, and acutely injured porcine hearts. By testing the technology in large animal models of infarction, compatibility of M3RNA with current coronary intervention procedures was verified.
Article
Heart failure (HF) is a global pandemic affecting at least 26 million people worldwide and is increasing in prevalence. HF health expenditures are considerable and will increase dramatically with an ageing population. Despite the significant advances in therapies and prevention, mortality and morbidity are still high and quality of life poor. The prevalence, incidence, mortality and morbidity rates reported show geographic variations, depending on the different aetiologies and clinical characteristics observed among patients with HF. In this review we focus on the global epidemiology of HF, providing data about prevalence, incidence, mortality and morbidity worldwide.
Article
Aims Cardiac myocyte hypertrophy, the main compensatory response to chronic stress in the heart often progresses to a state of decompensation that can lead to heart failure. Osteopontin (OPN) is an effector for extracellular signalling that induces myocyte growth and fibrosis. Although increased OPN activity has been observed in stressed myocytes and fibroblasts, the detailed and long term effects of blocking OPN signalling on the heart remain poorly defined. Targeting cardiac OPN protein by an RNA aptamer may be beneficial for tuning down OPN pathologic signalling. We aimed to demonstrate the therapeutic effects of an OPN RNA aptamer on cardiac dysfunction. Methods and results In vivo, we show that in a mouse model of pressure overload, treating at the time of surgeries with an OPN aptamer prevented cardiomyocyte hypertrophy and cardiac fibrosis, blocked OPN downstream signalling (PI3K and Akt phosphorylation), reduced expression of extracellular matrix (Lum, Col3a1, Fn1) and hypertrophy (Nppa, Nppb) genes, and prevented cardiac dysfunction. Treating at two months post-surgeries with the OPN aptamer reversed cardiac dysfunction and fibrosis and myocyte hypertrophy. While genetic homozygous deletion of OPN reduced myocardial wall thickness, surprisingly cardiac function and myocardial fibrosis, specifically collagen deposition and myofibroblast infiltration, were worse compared with wild type mice at three months of pressure overload. Conclusion Taken together, these data demonstrate that tuning down cardiac OPN signalling by an OPN RNA aptamer is a novel and effective approach for preventing cardiac hypertrophy and fibrosis, improving cardiac function, and reversing pressure overload-induced heart failure.