ArticlePDF Available

Abstract and Figures

Synthetic modified RNA (modRNA) is a novel vector for gene transfer to the heart and other organs. modRNA can mediate strong, transient protein expression with minimal induction of the innate immune response and risk of genome integration. modRNA is already being used in several human clinical trials and its use in basic and translational science is growing. Due to the complexity of preparing modRNA and the high cost of its reagents, there is a need for an improved, cost-efficient protocol to make modRNA. Here we show that changing the ratio between anti reverse cap analog (ARCA) and N1- methyl-pseudouridine (N1mΨ), favoring ARCA over N1mΨ, significantly increases the yield per reaction, improves modRNA translation and reduces its immunogenicity in vitro. This protocol will make modRNA preparation more accessible and financially affordable for basic and translational research.
Content may be subject to copyright.
Original Article
Optimizing Modified mRNA
In Vitro Synthesis Protocol
for Heart Gene Therapy
Yoav Hadas,
1,2,3,4
Nishat Sultana,
1,2,3,4
Elias Youssef,
1,2,3
Mohammad Tofael Kabir Sharkar,
1,2,3
Keerat Kaur,
1,2,3
Elena Chepurko,
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
Synthetic modied RNA (modRNA) is a novel vector for gene
transfer to the heart and other organs. modRNA can mediate
strong, transient protein expression with minimal induction
of the innate immune response and risk for genome integra-
tion. modRNA is already being used in several human clinical
trials, and its use in basic and translational science is growing.
Due to the complexity of preparing modRNA and the high cost
of its reagents, there is a need for an improved, cost-efcient
protocol to make modRNA. Here we show that changing
the ratio between anti-reverse cap analog (ARCA) and
N1-methyl-pseudouridine (N1mJ), favoring ARCA over
N1mJ, signicantly increases the yield per reaction, improves
modRNA translation, and reduces its immunogenicity in vitro.
This protocol will make modRNA preparation more accessible
and nancially affordable for basic and translational research.
INTRODUCTION
Gene therapy treats disease by using genetic material to change or
correct gene expression to cure disease. The concept of gene therapy
emerged in the early 1970s as a consequence of both increased under-
standing of the role of genes in human disease and the development of
genetic engineering techniques.
1
First attempts to treat human pa-
tients with exogenous genes were conducted in the 1980s and
1990s; however, safety issues hampered their progress.
2,3
Extensive
research in recent years has yielded better and safer gene delivery stra-
tegies that reduce vector immunogenicity; consequently, the gene
therapy eld is ourishing again, with exciting successes in treating
rare genetic diseases, including a rst US Food and Drug Administra-
tion (FDA)-approved gene therapy for inherited retinal dystrophy,
4
and thousands of gene therapy clinical trials worldwide.
5
In pursuit
of improved treatments for cardiovascular disease, including compli-
cations of myocardial infarction (MI) and heart failure (HF), exten-
sive gene therapy preclinical research resulted in numerous clinical
trials aiming to promote angiogenesis
6
and improve calcium homeo-
stasis.
7
Although proving safety, all cardiac gene therapy trials failed
to demonstrate signicant therapeutic efcacy.
7
It has been suggested
that inefcient gene transfer that resulted in poor expression of the
target gene is a potential reason for these neutral results.
7
Gene transfer can be achieved by using viral vectors including
adenovirus, retrovirus, adeno-associated virus (AAV), lentivirus,
vaccinia virus, poxvirus, and herpes simplex virus or non-viral vectors
including naked plasmid DNA (pDNA) or lipofection.
5
Modied
RNA (modRNA) is a novel vector for gene transfer to both dividing
and non-dividing mammalian cells that mediate fast, robust, transient
expression of proteins in the targeted cells or tissue.
8
Pre-clinical at-
tempts to use mRNA as a gene transfer vector showed successful
expression of the encoded proteins
9
and improved symptoms in a
rodent model of diabetes insipidus.
10
Yet the high immunogenicity
of exogenous mRNA
1115
and high susceptibility to degradation by
RNase have hampered progress in developing mRNA as a vector
for treating humans.
16,17
Since the rst description of a biologically
active and translatable in vitro transcription (IVT) product by Krieg
and Melton
18
in 1984, however, immense technological advances
have led to synthetic mRNAs emergence as a promising vector for
gene delivery.
19
Two important milestones in developing IVT tech-
nology turned synthetic mRNA from a scientic tool to a potential
platform for gene therapy. First, pioneering work by Karikó et al.
20
demonstrated that incorporating chemically modied nucleotides
in the synthetic mRNA results in a more stable and less immunogenic
mRNA that mediates rapid, high expression of the encoded
protein.
21,22
Second, the discovery of a stable cap analog, anti-
reverse cap analog (ARCA), increases synthetic mRNAs stability
and translatability.
23
We and others have recently shown that modRNA can be used to
express reporter proteins in rodent and porcine myocardium,
8,24,25
and that functional genes can be used to promote angiogenesis,
26
cardiomyocyte proliferation,
27
and cardiac cell survival.
28
Previously,
we described a detailed protocol for synthesizing modRNA capped
with ARCA 30-O-Me-m7G(50)ppp(50)G and substituting 100%
Received 23 June 2019; accepted 21 July 2019;
https://doi.org/10.1016/j.omtm.2019.07.006.
4
These authors contributed equally to this work.
Correspondence: Lior Zangi, Cardiovascular Research Center, Icahn School of
Medicine at Mount Sinai, New York, NY 10029, USA.
E-mail: lior.zangi@mssm.edu
300 Molecular Therapy: Methods & Clinical Development Vol. 14 September 2019 ª2019 The Authors.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
cytidine (C) with 5-methylcytidine (m5C) and 100% uridine (U) with
pseudouridine (J),
29
or substituting 100% uridine with N1-methyl-
pseudouridine (N1mJ), which has been shown to be superior over
the combination of Jand m5C.
8,30
Here we describe an optimized
protocol that indicates that a change in ARCA and N1mJratios
leads to cost-effective modRNA with higher protein expression and
lower immunogenicity in vitro.
RESULTS
modRNA Yield and Quality
Currently, there are two strategies for mRNA capping in an IVT re-
action: (1) post-transcriptional capping and (2) chemical capping.
In the post-transcriptional approach, the vaccinia virus capping
enzyme is used to cap the enzyme after the mRNA synthesis is
completed.
31,32
Although effective, this procedure is time-consuming
and expensive. In chemical capping, a cap analog is added directly to
the IVT reaction, after which the cap structure is incorporated at the
50end of the mRNA. To achieve a high percentage of capped mRNA
using this method, the ratio between the concentrations of the cap
analog and the guanosine triphosphate (GTP) in the reaction are
kept high. For IVT reactions, the ratio of ARCA to GTP should always
be 1:3.7. In order to reduce the synthetic mRNAs immunogenicity
and increase its translatability, we replace 100% U with N1mJ. Pre-
viously, for the IVT reaction, we used a composition of 5 nM ARCA,
1.35 mM GTP, 7.5 mM N1mJTP, and 7.5 mM MTP (composition 1
in Table 1, hereafter termed ARCA 5 protocol). To achieve our aim of
increasing the translation capacity and lowering the cost of modRNA
production, we reduce the amount of template DNA by 85% (Fig-
ure S1). We then explored different nucleotide compositions and
desalting methods for making and cleaning the modRNA (Table 1).
Out of the various nucleotide stoichiometries tested, composition 5,
comprising higher concentrations of ARCA, GTP, ATP, and cytidine
triphosphate (CTP) and less N1mJTP, another costly element in
modRNA production, resulted in the greatest modRNA yield
compared with the previously used protocol (ARCA 5 protocol),
keeping the amount of T7 RNA Polymerase consistent (Table 1).
To desalt the modRNA in the rst cleaning process, we compared
the MEGAclear RNA purication kit (used previously) with the
more cost-efcient Amicon lter. We found that the nuclear GFP
(nGFP) modRNA yield using the Amicon lter was equivalent to
the amount generated by the MEGAclear RNA purication kit.
Thus, we established that the most cost-effective protocol for IVT re-
action uses nucleotide concentrations of 10 mM ARCA, 2.7 mM GTP,
8.1 mM ATP, 8.1 mM CTP, and 2.7 mM N1mJTP followed by de-
salting and nucleotide clearing using the Amicon lter (this protocol
will now be referred to as the ARCA 10 protocol).
We also evaluated modRNA quality using a bioanalyzer. We found no
differences in the integrity of the modRNA generated using the
ARCA 10 protocol compared with that produced via the ARCA 5
protocol (Figures 1A and 1B). Interestingly, when using different
nucleotide compositions in the reaction, we observed high levels of
uncompleted IVT products (Figure 1F).
Translational Capacity of modRNA In Vitro
Next, we analyzed the translational capacity of modRNA generated
with the ARCA 10 protocol by transfecting several human cell
lines and primary cardiac cells isolated from neonatal rat hearts
and measuring the expression levels of two different reporter
genes: nGFP and rey luciferase (Luc) (Figure 2). We observed
signicantly increased nGFP expression in HeLa cells (data not
shown), primary rat cardiac cells, and neonatal rat cardiomyocytes
Table 1. Nucleotide Composition and Cleaning Methods
Nucleotide
Composition 1
(ARCA 5)
Nucleotide
Composition 2
Nucleotide
Composition 3
Nucleotide
Composition 4
Nucleotide
Composition 5
(ARCA 10)
Nucleotide
Composition 6
ARCA 5 mM 10 mM 10 mM 10 mM 10 mM 7.5 mM
GTP 1.35 mM 2.7 mM 2.7 mM 2.7 mM 2.7 mM 2.25 mM
ATP 7.5 mM 8.1 mM 15 mM 15 mM 8.1 mM 10 mM
CTP 7.5 mM 2.7 mM 15 mM 15 mM 8.1 mM 10 mM
N1mJTP 7.5 mM 2.7 mM 15 mM 2.7 mM 2.7 mM 2.25 mM
Total 28.85 mM 26.2 mM 57.7 mM 45.4 mM 31.6 mM 32 mM
Desalting with MEGAclear
Total mg nGFP modRNA per
1-mL reaction 1.33 3.12 1.19 2.13 2.94 1.93
% of ARCA 5 protocol 100 235 89 160 221 145
Desalting with Amicon Filter
Total mg nGFP modRNA per
1-mL reaction 2.12 1.04 1.92 2.94 1.74
% of ARCA 5 protocol 159 78 144 221 131
Summary of nucleotide composition and desalting (rst cleansing) methods used in this study and their effects on the nal modRNA yield. For modRNA integrity, see Figure 1.
www.moleculartherapy.org
Molecular Therapy: Methods & Clinical Development Vol. 14 September 2019 301
(nrCM) transfected with nGFP modRNA generated with the
ARCA 10 protocol compared with the ARCA 5 protocol (Figures
2C and 2D). Similar results were observed in primary rat cardiac
cells, nrCM, human umbilical vein endothelial cells (HUVECs),
HeLa cells, and HEK293 cells when transfected with Luc modRNA
(Figures 2E2H). We did not observe any difference in cell death
levels post transfection in vitro as measured by DAPI or propi-
dium iodide (PI)-positive cells (data not shown).
Immunogenicity of modRNA
Evaluating immunogenicity is critical to dening the safety prole
of mRNA therapeutics and their use in humans. To evaluate the
innate immune response in vitro, we measured the expression
levels of three innate immune-related genes, retinoic acid-induc-
ible gene I (RIG1), interferon alpha (IFNa), and interferon beta
(IFNb), in nrCM 8 h post transfection with Luc modRNA
(made by using either the ARCA 5 protocol or the ARCA 10
protocol) or unmodied mRNA (Figure 3). Importantly, Luc mod-
RNA made via both the ARCA 5 and ARCA 10 protocols has
signicantly lower levels of RIG1, IFNa,andIFNbwhen compared
with unmodied mRNA (Figures 3B3D). However, Luc made
using the ARCA 5 protocol was signicantly more immunogenic
than Luc modRNA made using the ARCA 10 protocol (Figures
3B3D).
DISCUSSION
Synthetic modRNA is now emerging as a promising vector for gene
delivery.
19
However, synthesizing modRNA in the high quantities
required by pre-clinical and clinical trials is expensive and time-
consuming. In light of recent successes in promoting angiogenesis,
26
CM proliferation,
27
and cardiac cell survival
28
in rodents, there in an
urgent need to reduce the cost and improve the quality of synthesizing
modRNA.
At present, our capping strategy is chemical capping; in this capping
method, the IVT reaction includes a cap analog, resulting in a cap
structure at the mRNAs5
0end. This capping method requires a
high ratio of cap analog and GTP concentrations to produce a high
percentage of capped mRNA.
This study aimed to optimize the protocol for synthesizing mRNA
using ARCA and N1mJ. Previously published protocols
8,26,28,29
maintained the correct ratio between ARCA and GTP by keeping
the GTP concentration low (1.35 mM) and the nucleotides high
(7.5 mM). In this composition, the GTP becomes the limiting factor
in the reaction, leading the reaction to terminate without incorpo-
rating the other nucleotides present in excess. The unincorporated
nucleotides are later washed away during the cleaning process.
Although the ratio between ARCA and GTP must be kept high,
we realized that the composition of the other nucleotides in the re-
action can be modied, leading us to explore the effect of different
nucleotide compositions on the nal yield and quality of the mod-
RNA. Interestingly, we found that when doubling the concentration
of all nucleotides to increase the nal concentration to 57.7 mM, the
nal yield is lower than the original concentration of 28.85 mM. The
optimal nal nucleotide concentration in our study was 31.6 mM
(ARCA 10 mM, GTP 2.7 mM, CTP 8.1 mM, ATP 8.1 mM, and
N1mJ2.7 mM). Using this concentration, we were able to increase
the nal modRNA yield by 290%. Combining this protocol with
improved cleaning methods results in modRNA with increases
translatability in both human cell lines and primary cardiac cells
(Figure 2), as well as reduced immunogenicity (Figure 3). A possible
explanation for this result is that optimizing nucleotide composition
in the IVT reaction raises the percentage of successfully capped
modRNA.
Here we developed an improved protocol for large-scale production
of modRNA. By increasing the nal nucleotide concentrations while
Figure 1. Effect of Nucleotide Composition in IVT Reaction on modRNA Integrity
(A–F) modRNA integrity was evaluated using a bioanalyzer (Tap Station 2200) of compositions 1 (A), 2 (B), 3 (C), 4 (D), 5 (E), and 6 (F).
Molecular Therapy: Methods & Clinical Development
302 Molecular Therapy: Methods & Clinical Development Vol. 14 September 2019
reducing the N1mJmolarity and using the same amount of T7 RNA
polymerase, we established a new method that is more cost-effective
than the previously published approach.
29
Our novel protocol
generated improved modRNA with higher translatability and lower
immunogenicity in vitro, and will allow basic and translational
research labs to overcome the high cost of modRNA preparation
and attain more effective modRNA for their research.
MATERIALS AND METHODS
Synthesizing modRNA
In brief, clean PCR products generated with plasmid templates
purchased from GenScript were used as the template for mRNA.
Figure 2. Effect of Nucleotide Composition in IVT
Reaction on Gene Expression in Human Cell Lines
and Rat Cardiac Cells
(A) GFP expression in rat cardiac cells 18 h post trans-
fection with or without nGFP modRNA generated with the
ARCA 5 protocol and ARCA 10 protocol. (B) Represen-
tative image of Luc activity level in neonatal rat car-
diomyocytes 24 h post transfection with or without Luc
modRNA generated using the ARCA 5 protocol and
ARCA 10 protocol. (C and D) Quantification of GFP
expression in neonatal rat cardiac cells (C) or car-
diomyocytes (D) 18 h post transfection with nGFP
modRNA generated using the ARCA 5 protocol and
ARCA 10 protocol. (E–H) Quantification of Luc activity
level in HUVECs (E), rat neonatal cardiomyocytes (F), HeLa
(G), or Hek293 (H) cells 18 h post transfection with or
without Luc modRNA generated using the ARCA 5
protocol and ARCA 10 protocol. One-way ANOVA,
Tukey’s multiple comparison test; ***p < 0.001, **p < 0.01,
*p < 0.05. n = 5 (A and B); n = 4 (C–F).
modRNAs were generated by transcription
in vitro with a customized ribonucleoside blend
of ARCA; 30-O-Me-m7G(50) ppp(50)G
(catalog no. [cat. #] N-1081; Trilink Biotechnol-
ogies); GTP (cat. #am1334-5; Life Technolo-
gies); ATP (cat. #am1334-5; Life Technologies);
cytidine triphosphate (cat. #am1334-5; Life
Technologies), and N1-methylpseudouridine-
50-triphosphate (cat. #N-1081; Trilink Biotech-
nologies). The mRNA was puried with the
MEGAclear kit (cat. #AM1908; Life Tech-
nologies) according to the manufacturers
instructions or using Amicon Ultra-4 Cen-
trifugal Filter Unit 4 mL,10 kDa (cat.
#UFC801024; MilliporeSigma) and treated
with Antarctic Phosphatase (cat. #M0289L;
NEB). It was then re-puried with the
MEGAclear kit. The mRNA was quantied
using a NanoDrop spectrometer (Thermo
Scientic), precipitated with ethanol and
ammonium acetate, and re-suspended in
10 mM Tris-HCl and 1 mM EDTA. The open reading frame for
the modRNA used is listed in Table S1. This protocol has been
described in detail elsewhere.
29
Desalting and Nucleotide Clearing
Amicon Ultra-4 Centrifugal Filter Unit 4 mL, 10 kDa
(cat. #UFC801024; MilliporeSigma) was washed with 4 mL
nuclease-free water two times. A total of 400 mLIVTreaction
was loaded to the lter and diluted with 3.5 mL nuclease-free
water. The lter was centrifuged at 3,200 gfor 20 min until
thevolumereducesto100mL. A total of 3.9 mL water was
added to the lter and centrifuged at 3,200 gfor 20 min two
www.moleculartherapy.org
Molecular Therapy: Methods & Clinical Development Vol. 14 September 2019 303
times. The puried modRNA was incubated at 70Cfor10min(to
inactivate T7).
Neonatal Rat Cardiac Cells Isolation
Neonatal rat ventricular CMs were isolated from 3- to 4-day-
old Sprague-Dawley rats (Jackson ImmunoResearch Laboratories)
by multiple rounds of digestion with 0.1% collagenase II (In-
vitrogen) in PBS. After each digestion, the supernatant was
collected in horse serum (Invitrogen). The total cell suspension
was centrifuged at 300 gfor 5 min. The supernatants were
discarded and cells were resuspended in DMEM (GIBCO)
supplemented with 0.1 mM ascorbic acid (Sigma), 0.5% insulin-
transferrin-selenium (100), penicillin (100 U/mL), and strepto-
mycin (100 mg/mL). Cells were plated in plastic culture dishes
for 90 min until most of the non-myocytes were attached to
the dish while the myocytes remained in suspension. Myocytes
were then used to seed 24-well plates at a density of 1 10
5
cells/well. nrCMs were incubated for 48 h in DMEM supple-
mented with 5% horse serum before transfection with modRNAs.
Twenty-four hours post isolation, the non-myocytes were used
to seed 24-well plates at a density of 1 10
5
cells/well.
The non-myocytes were transfected with modRNA 24 h post
plating.
In Vitro Transfection with modRNA
Using a 24-well plate, we complexed 2.5 mg/well mRNA with Lipo-
fectamine RNAiMAX Transfection Reagent (cat. #13778030; Life
Technologies) and used the resulting complex to transfect either
neonatal rat cardiac cells or human cell lines according to the manu-
facturers instructions.
Figure 3. Immunogenicity of modRNA Compared
with Unmodified mRNA
(A) Luc mRNA or modRNA transfected into neonatal rat
cardiomyocytes 8 h after transfection cells were collected.
qPCR was performed to evaluate the gene expression
of innate immune response markers. (B–D) Expression of
innate immune response markers: RIG1 (B), IFNa(C), or
IFNb(D) 8 h post transfecting unmodified mRNA, Luc
modRNA (ARCA 5 protocol), or Luc modRNA (ARCA
10 protocol). One-way ANOVA, Tukey’s multiple com-
parison test; ****p < 0.0001. n = 5.
Assessing Number of nGFP-Expressing
Cells
Eighteen hours post transfection, cells were
imaged using a Zeiss Slide Scanner Axio Scan.
Quantication of nGFP-positive cells was per-
formed using ImageJ software.
Detection of Luciferase Activity
Bioluminescence of the transfected cells was
measured 18 h post transfection. Each unit
of Luc signal represents p/s/cm
2
/sr 10
6
.
Luciferin (cat. #L9504; Sigma) (1.5 mg/mL culture media) was added
to cell culture 10 s before imaging. Cells were imaged using an
IVIS100 charge-coupled device imaging system every 20 s until the
Luc signal reached a plateau. Imaging data were analyzed and quan-
tied with Living Image software.
qRT-PCR
Total RNA was reverse transcribed with Superscript III reverse
transcriptase (cat. #LS18080044; Invitrogen), according to the manu-
facturers instructions. qRT-PCR analyses were performed on a
Mastercycler Realplex 4 Sequence Detector (Eppendorf) with the
HotStart-IT SYBR Green qPCR master mix (2) (cat. #75762; Affy-
metrix). Data were normalized relative to GAPDH. Fold-changes in
gene expression were determined by the delta-delta-Ct (ddCT)
method. The PCR primer sequences used are listed in Table S2.
Statistical Analyses
Statistical analyses were performed with GraphPad Prism software.
Values are reported as means ±SD. Two-tailed Students t tests
(*p < 0.05 considered signicant) or one-way ANOVA with Bonfer-
roni correction (*p < 0.05 considered signicant) were used for
comparisons between groups.
SUPPLEMENTAL INFORMATION
Supplemental Information can be found online at https://doi.org/10.
1016/j.omtm.2019.07.006.
AUTHOR CONTRIBUTIONS
Y.H. designed most of the experiments, performed experiments,
analyzed most of the data, and wrote the manuscript. N.S. performed
Molecular Therapy: Methods & Clinical Development
304 Molecular Therapy: Methods & Clinical Development Vol. 14 September 2019
most of the experiments, analyzed data, and revised the manuscript.
E.Y. performed experiments. M.T.K.S. prepared modRNAs and per-
formed experiments. K.K. revised the manuscript. E.E. performed
CM isolation method. L.Z. designed experiments, analyzed data,
and revised the manuscript.
ACKNOWLEDGMENTS
This work was funded by a cardiology start-up grant awarded to the
Zangi laboratory and also by NIH grant R01 HL142768-01.
REFERENCES
1. Friedmann, T., and Roblin, R. (1972). Gene therapy for human genetic disease?
Science 175, 949955.
2. Walters, L. (1984). Human Gene Therapy (DIANE Publishing).
3. Somia, N., and Verma, I.M. (2000). Gene therapy: trials and tribulations. Nat. Rev.
Genet. 1,9199.
4. Smalley, E. (2017). First AAV gene therapy poised for landmark approval. Nat.
Biotechnol. 35,998999.
5. Ginn, S.L., Amaya, A.K., Alexander, I.E., Edelstein, M., and Abedi, M.R. (2018). Gene
therapy clinical trials worldwide to 2017: An update. J. Gene Med. 20, e3015.
6. Gupta, R., Tongers, J., and Losordo, D.W. (2009). Human studies of angiogenic gene
therapy. Circ. Res. 105, 724736.
7. Ishikawa, K., Weber, T., and Hajjar, R.J. (2018). Human Cardiac Gene Th erapy. Circ.
Res. 123, 601613.
8. 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
Delivery of Modied mRNA. Mol. Ther. 25, 13061315.
9. 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.
10. Jirikowski, G.F., Sanna, P.P., Maciejewski-Lenoir, D., and Bloom, F.E. (1992).
Reversal of diabetes insipidus in Brattleboro rats: intrahypothalamic injection of
vasopressin mRNA. Science 255, 996998.
11. Diebold, S.S., Massacrier, C., Akira, S., Paturel, C., Morel, Y., and Reis e Sousa, C.
(2006). Nucleic acid agonists for Toll-like receptor 7 are dened by the presence of
uridine ribonucleotides. Eur. J. Immunol. 36, 32563267.
12. Heil, F., Hemmi, H., Hochrein, H., Ampenberger, F., Kirschning, C., Akira, S.,
Lipford, G., Wagner, H., and Bauer, S. (2004). Species-specic recognition of sin-
gle-stranded RNA via toll-like receptor 7 and 8. Science 303, 15261529.
13. Diebold, S.S., Kaisho, T., Hemmi, H., Akira, S., and Reis e Sousa, C. (2004). Innate
antiviral responses by means of TLR7-mediated recognition of single-stranded
RNA. Science 303, 15291531.
14. Alexopoulou, L., Holt, A.C., Medzhitov, R., and Flavell, R.A. (2001). Recognition of
double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature
413, 732738.
15. Pichlmair, A., Schulz, O., Tan, C.P., Rehwinkel, J., Kato, H., Takeuchi, O., Akira, S.,
Way, M., Schiavo, G., and Reis e Sousa, C. (2009). Activation of MDA5 requires
higher-order RNA structures generated during virus infection. J. Virol. 83, 10761
10769.
16. Dyer, K.D., and Rosenberg, H.F. (2006). The RNase a superfamily: generation of di-
versity and innate host defense. Mol. Divers. 10, 585597.
17. Rigby, R.E., and Rehwinkel, J. (2015). RNA degradation in antiviral immunity and
autoimmunity. Trends Immunol. 36, 179188.
18. Krieg, P.A., and Melton, D.A. (1984). Functional messenger RNAs are produced by
SP6 in vitro transcription of cloned cDNAs. Nucleic Acids Res. 12, 70577070.
19. Hadas, Y., Katz, M.G., Bridges, C.R., and Zangi, L. (2017). Modied mRNA as a
therapeutic tool to induce cardiac regeneration in ischemic heart disease. Wiley
Interdiscip. Rev. Syst. Biol. Med. 9, e1367.
20. Karikó, K., Buckstein, M., Ni, H., and Weissman, D. (2005). Suppression of RNA
recognition by Toll-like receptors: the impact of nucleoside modication and the
evolutionary origin of RNA. Immunity 23, 165175.
21. 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.
22. Anderson, B.R., Muramatsu, H., Nallagatla, S.R., Bevilacqua, P.C., Sansing, L.H.,
Weissman, D., and Karikó, K. (2010). Incorporation of pseudouridine into mRNA
enhances translation by diminishing PKR activation. Nucleic Acids Res. 38, 5884
5892.
23. Jemielity, J., Fowler, T., Zuberek, J., Stepinski, J., Lewdorowicz, M., Niedzwiecka, A.,
Stolarski, R., Darzynkiewicz, E., and Rhoads, R.E. (2003). Novel anti-reversecap an-
alogs with superior translational properties. RNA 9, 11081122.
24. 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. (2019). M
3
RNA drives targeted gene de-
livery in acute myocardial infarction. Tissue Eng. Part A 25, 145158.
25. 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.
26. 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.
27. 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
Cardiomyocyte Proliferation and Cardiac Regeneration. Mol. Ther. Nucleic Acids
13, 133143.
28. 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.
29. Kondrat, J., Sultana, N., and Zangi, L. (2017). Synthesis of Modied mRNA for
Myocardial Delivery. Methods Mol. Biol. 1521, 127138.
30. 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.
31. Fuchs, A.L., Neu, A., and Sprangers, R. (2016). A general method for rapid and cost-
efcient large-scale production of 50capped RNA. RNA 22, 14541466.
32. Paterson, B.M., and Rosenberg, M. (1979). Efcient translation of prokaryotic
mRNAs in a eukaryotic cell-free system requires addition of a cap structure.
Nature 279,692696.
www.moleculartherapy.org
Molecular Therapy: Methods & Clinical Development Vol. 14 September 2019 305
... Typical plasmid vectors contain a T7 promoter sequence upstream of multiple cloning sites. Furthermore, a poly(A) tail sequence, 5′ and 3′ untranslated regions are included in the DNA template design (Hadas et al., 2019;Tusup et al., 2019). In addition, a DNA template can be generated using a PCR product consisting of a T7 promoter at the 5′ end (Hadas et al., 2019). ...
... Furthermore, a poly(A) tail sequence, 5′ and 3′ untranslated regions are included in the DNA template design (Hadas et al., 2019;Tusup et al., 2019). In addition, a DNA template can be generated using a PCR product consisting of a T7 promoter at the 5′ end (Hadas et al., 2019). Typically, the poly(A) tail is incorporated in the initial DNA plasmid to be transcribed (Grier et al., 2016;To and Cho, 2021). ...
Article
Full-text available
mRNA technology has recently demonstrated the ability to significantly change the timeline for developing and delivering a new vaccine from years to months. The potential of mRNA technology for rapid vaccine development has recently been highlighted by the successful development and approval of two mRNA vaccines for COVID-19. Importantly, this RNA-based approach holds promise for treatments beyond vaccines and infectious diseases, e.g., treatments for cancer, metabolic disorders, cardiovascular conditions, and autoimmune diseases. There is currently significant demand for the development of improved manufacturing processes for the production of mRNA therapeutics in an effort to increase their yield and quality. The development of suitable analytical methods for the analysis of mRNA therapeutics is critical to underpin manufacturing development and the characterisation of the drug product and drug substance. In this study we have developed a high-throughput, high-performance liquid chromatography (HPLC) workflow for the rapid analysis of mRNA generated using in vitro transcription (IVT). We have optimised anion exchange (AEX) HPLC for the analysis of mRNA directly from IVT. Chromatography was performed in under 6 min enabling separation of all of the key components in the IVT, including nucleoside triphosphates (NTPs), Cap analogue, plasmid DNA and mRNA product. Moreover, baseline separation of the NTPs was achieved, which facilitates accurate quantification of each NTP such that their consumption may be determined during IVT reactions. Furthermore, the HPLC method was used to rapidly assess the purification of the mRNA product, including removal of NTPs/ Cap analogue and other contaminants after downstream purification, including solid phase extraction (SPE), oligo deoxythymidine (oligo-dT) affinity chromatography and tangential flow filtration (TFF). Using the developed method excellent precision was obtained with calibration curves for an external mRNA standard and NTPs giving correlation coefficients of 0.98 and 1.0 respectively. Intra-and inter-day studies on retention time stability of NTPs, showed a relative standard deviation ≤ 0.3% and ≤1.5% respectively. The mRNA retention time variability was ≤0.13%. This method was then utilised to monitor
... These plasmid vectors typically contain a T7 promoter sequence upstream of multiple cloning sites. The DNA template design also includes a poly(A) tail sequence, as well as 5' and 3' untranslated regions [17,18]. DNA template can also be produced from PCR products to include a T7 promoter sequence at the 5' end. ...
Article
Full-text available
mRNA technology has been successfully deployed to rapidly develop and mass-manufacture vaccines. Beyond vaccines, RNA-based therapeutics have potential for treatments for infectious diseases, cancer, metabolic disorders, cardiovascular conditions and autoimmune diseases. mRNA based vaccines and therapeutics work by translating exogenous mRNA into the target protein. Analytical methods for mRNA characterisation, lot release and stability testing of mRNA drug substance and drug product must be developed and performed to monitor critical quality attributes (CQAs). mRNA is a highly polar molecule due to its extensive negatively charged phosphodiester backbone. Its single stranded nature forms dynamic alternative secondary structures that can generate potential sample heterogeneity, creating challenges for the analysis and characterisation of this large biomolecule. In this review, we describe current analytical methods, focussing on high performance liquid chromatography in conjunction with both UV detection and mass spectrometry for the analysis and characterisation of mRNA. In particular, we describe recent developments covering a wide range of methods centred on liquid chromatography for the analysis of important CQAs including mRNA identity, mRNA integrity, 5′ capping efficiency and poly(A) tail length and heterogeneity.
... In vitro-transcribed (IVT) synthetic messenger RNA incorporates chemically modified nucleoside analogues to promote translation and limit innate immune responses. This technology has been reported to offer a rapid, dosable gene transfer mode in the heart with substantially reduced innate immune response [23][24][25][26][27] . Earlier efforts to deliver synthetic mRNA in vivo have relied upon cationic lipid carriers, which have potential complications of infusion-related hypersensitivity reactions, tissue injury and local innate immune activation [28][29][30] . ...
Article
Full-text available
The adenovirus-mediated somatic transfer of the embryonic T-box transcription factor 18 (TBX18) gene can convert chamber cardiomyocytes into induced pacemaker cells. However, the translation of therapeutic TBX18-induced cardiac pacing faces safety challenges. Here we show that the myocardial expression of synthetic TBX18 mRNA in animals generates de novo pacing and limits innate and inflammatory immune responses. In rats, intramyocardially injected mRNA remained localized, whereas direct myocardial injection of an adenovirus carrying a reporter gene resulted in diffuse expression and in substantial spillover to the liver, spleen and lungs. Transient expression of TBX18 mRNA in rats led to de novo automaticity and pacemaker properties and, compared with the injection of adenovirus, to substantial reductions in the expression of inflammatory genes and in activated macrophage populations. In rodent and clinically relevant porcine models of complete heart block, intramyocardially injected TBX18 mRNA provided rate-adaptive cardiac pacing for one month that strongly correlated with the animal’s sinus rhythm and physical activity. TBX18 mRNA may aid the development of biological pacemakers.
... Growing numbers of outbreaks of various diseases such as COVID-19, 1 monkeypox, 2 polio 3 and many others 46 gave a kick-start to usage of mRNA gene-therapies as vaccines against them. 7 These vaccines are also seen as potential treatments against cardiovascular diseases, 8,9 Alzheimer's disease, 10,11 cancer 12,13 etc. ...
Article
The efficacies of modern gene-therapies strongly depend on their contents. At the same time the most potent formulations might not contain the best compounds. In this work we investigated the effect of phospholipids and their saturation on the binding ability of (6Z,9Z,28Z,31Z)-heptatriacont-6,9,28,31-tetraene-19-yl 4-(dimethylamino) butanoate (DLin-MC3-DMA) to model membranes at the neutral pH. We discovered that DLin-MC3-DMA has affinity to the most saturated monocomponent lipid bilayer 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) and an aversion to the unsaturated one 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC). The preference to a certain membrane was also well-correlated to the phase transition temperatures of phospholipid bilayers, and to their structural and dynamical properties. Additionally, in the case of the presence of DLin-MC3-DMA in the membrane with DOPC the ionizable lipid penetrated it, which indicates possible synergistic effects. Comparisons with other ionizable lipids were performed using a model lipid bilayer of 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (POPC). Particularly, the lipids heptadecan-9-yl 8-[2-hydroxyethyl-(6-oxo-6-undecoxyhexyl)amino]octanoate (SM-102) and [(4-Hydroxybutyl) azanediyl] di(hexane-6,1-diyl) bis(2-hexyldecanoate) (ALC-0315) from modern mRNA-vaccines against COVID-19 were investigated and force fields parameters were derived for those new lipids. It was discovered that ALC-0315 binds strongest to the membrane, while DLin-MC3-DMA is not able to reside in the bilayer center. The ability to penetrate the membrane POPC by SM-102 and ALC-0315 can be related to their saturation, comparing to DLin-MC3-DMA.
Article
Full-text available
Heart failure remains the leading cause of human death worldwide. After a heart attack, the formation of scar tissue due to the massive death of cardiomyocytes leads to heart failure and sudden death in most cases. In addition, the regenerative ability of the adult heart is limited after injury, partly due to cell-cycle arrest in cardiomyocytes. In the current post-COVID-19 era, urgently authorized modified mRNA (modRNA) vaccines have been widely used to prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Therefore, modRNA-based protein replacement may act as an alternative strategy for improving heart disease. It is a safe, effective, transient, low-immunogenic, and integration-free strategy for in vivo protein expression, in addition to recombinant protein and stem-cell regenerative therapies. In this review, we provide a summary of various cardiac factors that have been utilized with the modRNA method to enhance cardiovascular regeneration, cardiomyocyte proliferation, fibrosis inhibition, and apoptosis inhibition. We further discuss other cardiac factors, modRNA delivery methods, and injection methods using the modRNA approach to explore their application potential in heart disease. Factors for promoting cardiomyocyte proliferation such as a cocktail of three genes comprising FoxM1, Id1, and Jnk3-shRNA (FIJs), gp130, and melatonin have potential to be applied in the modRNA approach. We also discuss the current challenges with respect to modRNA-based cardiac regenerative medicine that need to be overcome to apply this approach to heart disease. This review provides a short description for investigators interested in the development of alternative cardiac regenerative medicines using the modRNA platform.
Article
Full-text available
The discovery of nucleic acids stands as a paramount achievement in the history of scientific endeavors. By applying trans-formative advancements in the fields of chemistry and physics to biological systems, researchers unveiled the enigmatic nature of life. Notably, messenger RNA (mRNA) emerged as a crucial player in this profound revelation, serving as a transient intermediary for genetic information transfer between genes and proteins. Groundbreaking investigations carried out from 1944 to 1961 led to the initial identification of this pivotal molecule, captivating scientific interest for the past three decades. The field of mRNA research has witnessed a transformative shift owing to the development of cap analogs and nucleotide modifications. This revolutionary progress has fostered a new generation of potent therapeutics. Prior to the advent of the coronavirus pandemic, numerous scientists had already begun exploring the unique properties of mRNA. However, with the onset of the pandemic, mRNA catapulted into the limelight as a heroic agent, providing the foundation for highly effective vaccines that have played a crucial role in mitigating the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The successive generations of cap analogs have significantly enhanced the translation efficacy of mRNA, while the discovery of suitable purification, packaging, and delivery methods has paved the way for groundbreaking medical breakthroughs. Pioneers in the field such as Katalin Karikó, Drew Weissman, Edward Darzynkiewicz, Robert Rhodes, Ugur Sahin, and Ozlem Tureci have made significant contributions during the early stages of mRNA research, warranting acknowledgement for their visionary endeavors. The narrative of mRNA represents a remarkable journey marked by a succession of breakthroughs in a discipline that holds immense promise for the future of medicine. Thanks to the pioneering work of these exceptional scientists, we are well-positioned to unlock the full potential of this extraordinary molecule, ushering in a new era of medical advancements.
Article
Full-text available
Base editing is a revolutionary gene-editing technique enabling the introduction of point mutations into the genome without generating detrimental DNA double-stranded breaks. Base-editing enzymes are commonly delivered in the form of modified linear messenger RNA (mRNA) that is costly to produce. Here, we address this problem by developing a simple protocol for manufacturing base-edited cells using circular RNA (circRNA), which is less expensive to synthesize. Compared with linear mRNA, higher editing efficiencies were achieved with circRNA, enabling an 8-fold reduction in the amount of RNA required. We used this protocol to manufacture a clinical dose (1 × 10⁸ cells) of base-edited chimeric antigen receptor (CAR) T cells lacking expression of the inhibitory receptor, PD-1. Editing efficiencies of up to 86% were obtained using 0.25 μg circRNA/1 × 10⁶ cells. Increased editing efficiencies with circRNA were attributed to more efficient translation. These results suggest that circRNA, which is less expensive to produce than linear mRNA, is a viable option for reducing the cost of manufacturing base-edited cells at scale.
Article
Background: Experiments in mammalian models of cardiac injury suggest that the cardiomyocyte-specific overexpression of CCND2 (cyclin D2, in humans) improves recovery from myocardial infarction (MI). The primary objective of this investigation was to demonstrate that our specific modified mRNA translation system (SMRTs) can induce CCND2 expression in Cardiomyocytes and replicate the benefits observed in other studies of cardiomyocyte-specific CCND2 overexpression for myocardial repair. Methods: The CCND2-cardiomyocyte-specific modified mRNA translation system (cardiomyocyte SMRTs) consists of 2 modRNA constructs: 1 code for CCND2 and contains a binding site for L7Ae, and the other codes for L7Ae and contains recognition elements for the cardiomyocyte-specific microRNAs miR-1 and miR-208. Thus, L7Ae suppresses CCND2 translation in noncardiomyocytes but is itself suppressed by endogenous miR-1 and -208 in cardiomyocytes, thereby facilitating cardiomyocyte-specific CCND2 expression. Experiments were conducted in both mouse and pig models of MI, and control assessments were performed in animals treated with an SMRTs coding for the cardiomyocyte-specific expression of luciferase or GFP, in animals treated with L7Ae modRNA alone or with the delivery vehicle, and in Sham-operated animals. Results: CCND2 was abundantly expressed in cultured, postmitotic cardiomyocytes 2 days after transfection with the CCND2-cardiomyocyte SMRTs, and the increase was accompanied by the upregulation of markers for cell-cycle activation and proliferation (eg, Ki67 and Aurora B kinase). When the GFP-cardiomyocyte SMRTs were intramyocardially injected into infarcted mouse hearts, the GFP signal was observed in cardiomyocytes but no other cell type. In both MI models, cardiomyocyte proliferation (on day 7 and day 3 after treatment administration in mice and pigs, respectively) was significantly greater, left-ventricular ejection fractions (days 7 and 28 in mice, days 10 and 28 in pigs) were significantly higher, and infarcts (day 28 in both species) were significantly smaller in animals treated with the CCND2-cardiomyocyte SMRTs than in any other group that underwent MI induction. Conclusions: Intramyocardial injections of the CCND2-cardiomyocyte SMRTs promoted cardiomyocyte proliferation, reduced infarct size, and improved cardiac performance in small and large mammalian hearts with MI.
Article
Full-text available
Recent advances in mRNA therapeutics demand efficient toolkits for the incorporation of nucleoside analogues into mRNA suitable for downstream applications. Herein, we report the application of a versatile enzyme cascade for the triphosphorylation of a broad range of nucleoside analogues, including unprotected nucleobases containing chemically labile moieties. Our biomimetic system was suitable for the preparation of nucleoside triphosphates containing adenosine, cytidine, guanosine, uridine and non‐canonical core structures, as determined by capillary electrophoresis coupled to mass spectrometry. This enabled us to establish an efficient workflow for transcribing and purifying functional mRNA containing these nucleoside analogues, combined with mass spectrometric verification of analogue incorporation. Our combined methodology allows for analyses of how incorporation of nucleoside analogues that are commercially unavailable as triphosphates affect mRNA properties: The translational fidelity of the produced mRNA was demonstrated in analyses of how incorporated adenosine analogues impact translational recoding. For the SARS‐CoV‐2 frameshifting site, analyses of the mRNA pseudoknot structure using circular dichroism spectroscopy allowed insight into how the pharmacologically active 7‐deazaadenosine destabilises RNA secondary structure, consistent with observed changes in recoding efficiency.
Article
Messenger RNA (mRNA) has become a key focus in the development of therapeutic agents, showing significant potential in preventing and treating a wide range of diseases. The COVID-19 pandemic in 2020 has accelerated the development of mRNA nucleic therapeutics and attracted significant investment from global biopharmaceutical companies. These therapeutics deliver genetic information into cells without altering the host genome, making them a promising treatment option. However, their clinical applications have been limited by issues such as instability, inefficient in vivo delivery, and low translational efficiency. Recent advances in molecular design and nanotechnology have helped overcome these challenges, and several mRNA formulations have demonstrated promising results in both animal and human testing against infectious diseases and cancer. This review provides an overview of the latest research progress in structural optimization strategies and delivery systems, and discusses key considerations for their future clinical use.
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
To date, nearly 2600 gene therapy clinical trials have been completed, are ongoing or have been approved worldwide. Our database brings together global information on gene therapy clinical activity from trial databases, official agency sources, published literature, conference presentations and posters kindly provided to us by individual investigators or trial sponsors. This review presents our analysis of clinical trials that, to the best of our knowledge, have been or are being performed worldwide. As of our November 2017 update, we have entries on 2597 trials undertaken in 38 countries. We have analysed the geographical distribution of trials, the disease indications (or other reasons) for trials, the proportions to which different vector types are used, and which genes have been transferred. Details of the analyses presented, and our searchable database are on The Journal of Gene Medicine Gene Therapy Clinical Trials Worldwide website at http://www.wiley.co.uk/genmed/clinical. We also provide an overview of the progress being made in gene therapy clinical trials around the world, and discuss key trends since the previous review, namely the use of chimeric antigen receptor (CAR) T cells for the treatment of cancer and advancements in genome editing technologies, which have the potential to transform the field moving forward.
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
Chapter
Full-text available
An area of active research in the field of cardiac gene therapy aims to achieve high transfection efficiency without eliciting immune or inflammatory reactions. Nanomedicine offers an attractive alternative to traditional viral delivery vehicles because nanoparticle technology can enable safer and more controlled delivery of therapeutic agents. Here we describe the use of lipidoid nanoparticles for delivery of modified mRNA (modRNA) to the myocardium in vivo, with a focus on rodent models that represent a first step toward preclinical studies. Three major procedures are discussed in this chapter: (1) preparation of lipid modRNA nanoparticles, (2) intramyocardial delivery of the lipid modRNA nanoparticles by direct injection with an open chest technique in rats, and (3) intracoronary delivery of the lipid modRNA nanoparticles with open chest and temporary aortic cross clamping in rats.
Chapter
Full-text available
Cardiac gene therapy shows tremendous promise in combating the growing problem of heart disease. Modified mRNA (modRNA) is a novel gene delivery system used in vitro or in vivo to achieve transient expression of therapeutic proteins in a heterogeneous population of cells. Incorporation of specific modified nucleosides enables modRNA to be translated efficiently without triggering antiviral and innate immune responses. ModRNA has been shown to be effective at delivering short-term robust gene expression to the heart and its use in the field of cardiac gene therapy is expanding. Here, we describe a stepwise protocol for the synthesis of modRNA for in vivo myocardial delivery.
Article
In the past 10 years, there has been tremendous progress made in the field of gene therapy. Effective treatments of Leber congenital amaurosis, hemophilia, and spinal muscular atrophy have been largely based on the efficiency and safety of adeno-associated vectors. Myocardial gene therapy has been tested in patients with heart failure using adeno-associated vectors with no safety concerns but lacking clinical improvements. Cardiac gene therapy is adapting to the new developments in vectors, delivery systems, targets, and clinical end points and is poised for success in the near future.
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.