ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 6, pp. 725-753 © Pleiades Publishing, Ltd., 2025.
Published in Russian in Biokhimiya, 2025, Vol. 90, No. 6, pp. 781-811.
725
REVIEW
Selection of UTRs
in mRNA-Based Gene Therapy and Vaccines
Ilya A. Volkhin
1#
, Anastasia Iu. Paremskaia
2,3#
, Maria A. Dashian
3
,
Darya S. Smeshnova
2
, Roman E. Pavlov
2
, Olga N. Mityaeva
2,4
,
Pavel Yu. Volchkov
2,5
, and Andrei A. Deviatkin
2,6,7,a
*
1
Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University,
119234 Moscow, Russia
2
Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies,
125315 Moscow, Russia
3
Department of Biomedicine, Pirogov Medical University, 117997 Moscow, Russia
4
Moscow Center for Advanced Studies, 123592 Moscow, Russia
5
Center for Personalized Medicine, Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
6
Laboratory of Postgenomic Technologies, Izmerov Research Institute of Occupational Health,
105275 Moscow, Russia
7
Federal State Budgetary Institution ‘Centre for Strategic Planning and Management
of Biomedical Health Risks’ of the Federal Medical and Biological Agency,
119121 Moscow, Russia
a
e-mail: devyatkin_aa@academpharm.ru
Received December 25, 2024
Revised May 19, 2025
Accepted May 28, 2025
AbstractThe untranslated regions (UTRs) of messenger RNAs (mRNAs) play a crucial role in regulating
translational efficiency, stability, and tissue-specific expression. The review describes various applications
and challenges of UTR design in the development of gene therapy and mRNA-based therapeutics. UTRs af-
fect critical biological functions, such as mRNA stability, modulation of protein synthesis, and attenuation of
immune response. Incorporating tissue-specific microRNA (miRNA)-binding sites into 3′ UTRs might improve
precise targeting of transgene expression and minimize off-target effects. Nucleotide modifications (pseudou-
ridine, N1-methyladenosine, and N4-acetylcytidine) in mRNA and UTRs in particular, improve mRNA stability
and translational efficiency. At the same time, several challenges remain, such as lack of consensus on UTRs
best suited for certain biomedical applications. Current efforts are focused on integrating high-throughput
screening, computational modeling, and experimental validation to refine UTR-based therapeutic strategies.
The review presents current information on the design of UTRs and their role in therapeutic applications,
with special focus on the possibilities and limitations of existing approaches.
DOI: 10.1134/S0006297924604659
Keywords: gene therapy, mRNA vaccine, UTR, rational design, optimization, RNA secondary structure
* To whom correspondence should be addressed.
# These authors contributed equally to this study.
INTRODUCTION
The emergence of mRNA-based therapeutics has
revolutionized gene therapy and vaccine development
by offering promising solutions in the treatment of
a broad range of diseases, from genetic disorders to
infectious diseases [1, 2]. Currently, there are sever-
al approaches to delivering transgenes into human
cells for the following protein translation on the de-
livered mRNA. mRNA vaccines are typically used for
a short-term transgene expression, while viral vectors
[adeno-associated viruses (AAVs), adenoviruses, len-
tiviruses, etc.] ensure a long-term expression of the
VOLKHIN et al.726
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
transgene. After virus entry to the cell, transcription
of the viral mRNA is regulated by the same processes
as transcription of exogenous mRNAs. However, the
differences between such mRNAs should not be ig-
nored. For example, modifications of ribonucleotides
aimed to reduce the immunogenicity [3] and increase
the translational efficiency [4] of artificial RNAs can
disrupt their translation [5]. Numerous details in the
process of optimizing the structure of viral vectors
has been described in several studies [6-9]. Howev-
er, the issue of UTR design has been rarely discussed
[10-12]. At the same time, the studies describing novel
approaches to the UTR design [13-16] do not address
the challenge of extending such improved UTR design
to viral vectors (e.g., AAV vectors). The introduction
of best practices in the mRNA design phase of AAV
vectors and mRNA vaccines can significantly improve
both technologies.
mRNA vaccine encodes a protein (e.g., surface an-
tigen of a pathogen or cancer epitope) that can trigger
the human immune response. mRNAs can be encapsu-
lated in lipid nanoparticles, lipid-like materials, poly-
meric nanoparticles, hybrid systems, and nanoemul-
sions [1, 17, 18]. These artificial carriers are positively
charged and form polyplexes with negatively charged
mRNAs, which facilitates their release from the par-
ticle into the cell’s cytoplasm for further translation
[19, 20]. It should be noted that mRNAs are relatively
rapidly degraded in the cell. However, their transla-
tion yields heterologous proteins that might induce
the immune response against the target antigen. The
synthesized protein is further processed into smaller
peptide epitopes that are presented on the cell surface
by the major histocompatibility complexes (MHCs)
class  I and class  II to CD8
+
and CD4
+
T  cells, leading
to the development of cellular and humoral immu-
nity, respectively, against the antigen encoded in the
mRNA [17,  21]. Due to its high immunogenicity and
short lifespan, mRNA is considered to be the optimal
vaccine against infectious diseases or cancer [17,  20,
22]. In some cases, mRNA is used as a template for
the synthesis of a functional protein that is necessary
for the treatment of a specific disease. For example,
mRNA-based therapeutics against protein deficien-
cy-related metabolic diseases (propionic acidemia,
methylmalonic acidemia, and phenylketonuria) have
recently been successfully tested in preclinical studies
[23]. Hence, the range of possible applications for mR-
NA-based drugs can be extended to the development
of protein replacement therapies.
Viral vectors (adenoviruses, lentiviruses, retro-
viruses, AAVs, and others) are typically used for the
long-term transgene production a with a direct ther-
apeutic effect [24] which is achieved by maintaining
the viral genome through its constant transcription.
This process results in the intracellular synthesis of
mRNA utilized for the production of a desired pro-
tein. The precise dosage of the virus is determined,
in particular, by the efficiency of protein synthesis,
as higher efficiency of transcription and translation
allows to use a lower virus dose, which is safer and
more cost-effective [25]. Due to the high efficacy, AAV
vectors have been used much more commonly than
other viral vectors [26, 27]. For this reason, this re-
view focused mostly on AAV-based gene therapeutics.
Recombinant AAV (rAAV) vectors, which lack the viral
rep and cap genes, carry an expression cassette con-
sisting of the transcription promoter, single-stranded
DNA coding for the transgene and flanked by inverted
terminal repeats, and polyadenylation signal (polyA)
within an icosahedral capsid that can allow tissue-spe-
cific targeting. After endocytosis, viral single-stranded
DNA of the forms episomes in the nucleus and then
converted into the double-stranded DNA transcribed
by the cellular machinery. The mRNA is exported to
the cytosol for the translation of the therapeutic trans-
gene [27, 28]. The lifespan of the transgene introduced
to the cell by this approach is limited only by the lifes-
pan of the cell itself [21  29]. For instance, in tissues
with a low cell turnover (e.g., muscles), expression of
the introduced transgene (factor IX) has been detected
for at least 10 years [30]. Consequently, the use of AAV
vectors is currently the preferred method for produc-
ing intracellular functional proteins to compensate for
defective endogenous proteins in the cytoplasm. The
application of AAVs as delivery vectors for the gene
replacement therapy has been supported by clinical
trials and FDA approvals for their use in the treatment
of ocular, neurological, metabolic, and hematological
disorders [27, 31-35].
The above therapies rely directly (mRNA vaccines)
or indirectly (AAV vectors) on mRNA as a template
for protein synthesis. It should be noted that mRNAs
transcribed on the AAV DNA in the nucleus and then
exported to the cytoplasm are structurally similar
to heterologous mRNAs that enter the cell from the
external environment (Fig.  1). Both types of mRNAs
have similar structural domains that contain all essen-
tial elements required for their translation, namely,
the 5′  cap, 5′  untranslated region (UTR), open read-
ing frame (ORF), 3′ UTR, and poly(A) tail [1, 20, 36].
Atthe same time, the mechanism for exogenous RNA
recognition by the cellular machinery can be differ-
ent from that for endogenous RNAs. RNA synthesized
in vitro may not have the nucleotide composition and
secondary structure of natural RNAs. As a result, ex-
ternal RNAs can trigger the mechanisms of intracellu-
lar immunity after recognition by the corresponding
RNA-binding proteins [37]. Moreover, the process of
RNA synthesis in vitro is not precise and can lead
to the formation of double-stranded RNAs [38] and
truncated RNAs (aborted transcription products) [39].
SELECTION OF UTRs 727
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig. 1. mRNA-based therapeutics: mRNA vaccine (left) and rAAV vector-based drug (right). The mRNA vaccine contains
ready-for-translation mRNA encapsulated in a liposomal particle. Upon release from the liposome inside the cell, the mRNA
is translated by the cell ribosomes, and the synthesized polypeptide serves as an antigen that stimulates the host’s im-
mune response or as a therapeutic protein compensating for a defective cellular protein. The rAAV vector consists of a
single-stranded DNA carrying a transgene inside a viral capsid. After the vector enters the cell via endocytosis, the rAAV
genome is transcribed in the nucleus. The resulting mRNA is exported to the cytoplasm, where it is translated into the
therapeutic protein.
Numerous efforts have been made to minimize these
factors (see review [37]).
After mRNA entry to the cytoplasm, its 3′ poly(A)
tail and 5′ cap interact with the translation initiation
factors (eIFs), leading to the mRNA circularization and
recruitment of the preinitiation complex (PIC) formed
by the 40S small ribosomal subunit, eIFs, initiating me-
thionyl-tRNA, and GTP. The 40S small ribosomal sub-
unit scans the 5′  UTR for the start codon in the Kozak
sequence. After the start codon is found, the eIFs are
released and the 60S ribosomal subunit is recruited
to form the 80S ribosome capable of protein synthe-
sis. The elongation of the amino acid chain continues
until the stop codon is reached. Finally, the nascent
protein gains functionality through post-translational
modifications [1,  17,  40]. It should be noted that trans-
lation requires the presence of all mRNA structural
domains (5′  cap, 5′  UTR, Kozak sequence, ORF, and
3′  UTR). Therefore, altering any mRNA domain can
directly affect the stability, immunogenicity, and trans-
lation of mRNA therapeutics [20, 41]. Currently, there
are a variety of approaches for optimizing the 5′  cap,
nucleotide context of the start codon, and poly(A) tail
[42-58]. For example, the length of the poly(A) tail
can influence the efficiency of mRNA translation and
mRNA degradation rate [45, 50-52]. Interestingly, the
effect of the poly(A) tail on translation depends on the
length of the 3′ UTR [59, 60]. The stability of mRNA
can also be affected by the chemical modifications of
the poly(A) tail [61, 62]. However, this review primar-
ily focuses on the strategies for optimizing the UTRs.
Despite a significant progress in the development
of mRNA-based therapies and vaccines, there is a lack
of consensus on the optimal UTRs to be used in spe-
cific biomedical applications. UTRs, which flank the
coding sequence in mRNA, are crucial for regulating
the translational efficiency and stability of mRNA.
Commonly, the UTRs selected for therapeutic applica-
tions (e.g., UTRs of human β-globin mRNA) provide a
high translational efficiency and mRNA stability. Con-
sequently, the optimization of UTRs has emerged as
an area of focus in the development of mRNA-based
therapeutics [63, 64].
In this review, we analyzed in detail the UTRs
that have shown promise in preclinical and clinical
studies in order to identify the strategies for selection
VOLKHIN et al.728
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig.  2. Regulatory structures in the 5′ UTR and 3′ UTR that influence translational efficiency and mRNA stability. Abbrevi-
ations: uORF, upstream open reading frame; lncRNA, long non-coding RNA; miRBS, miRNA-binding site; miRISC, miRNA-
induced silencing complex; 4E-T, eukaryotic translation initiation factor 4E transporter; CCR4-NOT, carbon catabolite re-
pression – negative on TATA-less; STAU1, staufen double-stranded RNA binding protein 1; ARE, AU-rich element; WPRE,
woodchuck hepatitis virus posttranscriptional regulatory element; RBPs, RNA-binding proteins.
of most effective UTRs for therapeutic applications
and to guide researchers in choosing the optimal UTRs
for their specific needs.
THE EFFECT OF UTRs
ON THE PROTEIN YIELD
The translational efficiency (translation rate) is
often defined as the number of protein molecules
produced per mRNA molecule per unit of time [65].
The mRNA degradation rate is defined as the number
of mRNA molecules degraded per unit of time, reflect-
ing the speed rate at which mRNA is eliminated from
the cell and can be influenced by multiple factors,
such as mRNA sequence elements, RNA-binding pro-
teins (RBPs), microRNAs (miRNAs), and various decay
mechanisms.
The goal behind the generation of synthetic
mRNAs is usually to achieve a higher protein yield,
which depends on both mRNA translation efficiency
and degradation rate. The half-live time differs for dif-
ferent mRNAs, with a median value of approximately
7  h  [66]. There is an inherent trade-off between the
translation rate and mRNA stability  [67]. Although
high ribosome loading increases the translation rate,
it can also make mRNA more susceptible to hydrolysis.
The reasons for this phenomenon remain unknown,
but might involve ribosome crowding and consequent
mRNA degradation. In fact, stable mRNAs, even if they
have a lower translational efficiency, can ultimately
produce more protein over time due to a longer lifes-
pan [67]. Therefore, balancing translational efficiency
and mRNA stability is critical, and the choice of UTRs
plays a pivotal role in modulating these factors.
The efficiency of translation is influenced by the
stability and position of the secondary structure ele-
ments (e.g., hairpins) within the 5′ UTR (Fig. 2). The
rate-limiting step of translation initiation is the bind-
ing of the 43S PIC to mRNA [68]. Since stable second-
ary structures in the 5′ UTR can prevent this process,
it is generally assumed that they suppress translation.
As the stability of the hairpin increases, the transla-
tional efficiency typically decreases. For example, an
increase in the hairpin stability from –25 to –35  kcal/
mol can almost completely halt the translation [68].
It has also been shown that hairpins located close to
the 5′  cap strongly repress translation [69], likely by
interfering with the mRNA recognition or PIC bind-
ing. Furthermore, the guanine–cytosine (GC) content
of the hairpin influences the translational efficien-
cy independently of the hairpin thermal stability or
position [68]. The simplest explanation is that the ri-
bosome does not melt the entire hairpin at once but
unwinds it gradually. The GC pairs are more difficult
to break apart than the AU pairs because of their high-
er stability due to an additional hydrogen bond and,
therefore, can slow down or delay the progress of the
ribosome. The hairpins with the same stability but a
higher GC content have a stronger inhibitory effect
on the translational efficiency. Even though secondary
structures typically reduce the efficiency of transla-
tion, some 5′  UTRs with stable secondary structure el-
ements can support efficient initiation of translation,
for example, the 5′  UTR of dengue virus (DEN2) [67],
presumably, due to the promotion of efficient cap-in-
dependent translation initiation by the secondary
structure [70]. Another probable explanation is that
the 5′  UTR of DEN2 interacts with the cellular protein
La, which acts as an RNA helicase and thus facilitates
translation [71]. In summary, it is usually better to
avoid formation of stable secondary structures in the
5′ UTRs of artificial mRNAs, especially near the start
codon and the cap.
Generally, stable elements of secondary struc-
tures increase RNA stability in solution by protecting
it from hydrolysis [72]. mRNA molecules in the in-
tracellular environment are generally less structured
SELECTION OF UTRs 729
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
than in vitro, due to the interactions with RBPs, RNA
helicases, and ribosomes [73], which highlights the
importance of cellular context when evaluating the
structural features of RNA.
G-quadruplexes (RG4s) are a specific type of tran-
sient four-stranded structures in RNA that can impact
translation [74]. The exact mechanism of their influ-
ence remains unknown, but it has been hypothesized
that RG4s inhibit translation by preventing the bind-
ing of the 43S PIC to mRNA or by slowing down the
scanning [73]. However, there is also evidence of a
positive influence of RG4s on translation (e.g., in the
case of human clAP1 mRNA) [75]. Therefore, the im-
pact of RG4s on translation appears to be multidirec-
tional, and its underlying mechanisms still have to be
elucidated in future studies.
Upstream open reading frames (uORFs) are short
reading frames located upstream of the major coding
sequence; they are common regulatory sequences in
human 5′  UTRs [76-80]. Although ribosomes can reini-
tiate translation downstream of uORFs, these elements
are thought to have a strong inhibitory effect on trans-
lation [81, 82]. The simplest mechanism of such inhibi-
tion is recognition of the uORF instead of the canoni-
cal ORF, which decreases the probability of translation
initiation for the canonical protein. Another mecha-
nism is ribosome stalling of by a peptide encoded by
such uORF, as was shown for human cytomegalovirus
UL4 mRNA  [83]. Beside decrease the translational ef-
ficiency, uORFs cause mRNA degradation via the non-
sense-mediated mRNA decay mechanism [84, 85] initi-
ated by the presence of a premature stop codon [86].
It was demonstrated that an uORF within the 5′ UTR
of mRNA can activate the process, ultimately result-
ing in mRNA degradation [86, 87]. At the same time
some UTRs contain signals that protects mRNA from
degradation despite the presence of uORFs, as it was
shown for yeast mRNAs [88]. The upstream start co-
dons (uAUGs) that do not form complete uORFs within
the 5′  UTR (i.e., lack the in-frame stop codon before
the main AUG) have an even stronger inhibitory effect
on translation than complete uORFs. The reason for
this might be the inability of ribosomes to reinitiate
translation from the main ORF downstream of such
uAUGs [76]. Since uORFs and uAUGs usually have a
strong negative impact on translation, they should be
absent in artificial mRNAs.
The presence of binding sites for proteins and
long non-coding RNAs (lncRNAs) in the UTRs can
also affect translation due to the interaction of these
lncRNAs and RBPs with the translation machinery
[73,  89]. For example, the antisense Uchl1 lncRNA
promotes the synthesis of mouse ubiquitin carboxy-
terminal hydrolase L1 (UCHL1) due to the enhanced
ribosome binding [90]. The RBP-binding sites can ei-
ther enhance or inhibit translation. An example of the
RBP-mediated translational regulation is the role of
iron regulatory proteins  1 and 2 (IRP1 and 2). These
proteins can bind to specific regions (known as iron
response elements) in the 5′  UTRs of mRNAs coding
for iron metabolism proteins, thus inhibiting their
translation [91]. Another example is regulation of the
msl-2 mRNA translation by the sex-lethal (SXL) protein
in Drosophila, which was found to bind both the 5′
and 3′ UTRs of msl-2 mRNA and to inhibit its trans-
lation. However, the mechanisms of inhibition differ
for the 5′ and 3′  UTRs. In the 3′  UTR, SXL recruits the
UNR (upstream of N-Ras) repressor protein and pre-
vents the formation of PIC. The binding of SXL to the
5′  UTR requires the presence of the binding site and
the uORF and promotes the recognition of uAUG, thus
inhibiting translation of the main ORF [92]. An exam-
ple of the positive effect of RBPs is activation of the
cap-independent translation by the YB-1 protein [93].
Some GC-enriched clusters present in particular hu-
man mRNAs can recruit YB-1 and promote translation.
In summary, the interaction of UTRs with lncRNAs and
RBPs may also play a role in the translational regula-
tion. Therefore, potential interactions of the binding
motifs present in the UTRs should be taken into ac-
count when selecting the UTRs for the use in mRNA
vaccines and mRNA-based agents.
In contrast to the 5′ UTR, the 3′  UTR are believed
to have a lesser influence on the ribosome loading
and, therefore, translation rate [67]. However, the
3′  UTRs may contain some regulatory sequences capa-
ble of interacting with RBPs or microRNAs (miRNAs)
and influencing the efficiency of mRNA translation
and lifespan [66, 94]. Their effect could be explained
by several mechanisms. For example, the CCR4-NOT
exonuclease can inhibit translation and cause mRNA
decay. It is recruited by RBPs and miRNAs interacting
with specific sequence motifs in the 3′  UTR. Hence,
a 3′ UTR lacking these sites will not be degraded by
CCR4-NOT. One of the mechanisms of CCR4-NOT action
is physical displacement of poly(A)-binding proteins
(PABPs) and prevention of translation initiation [95].
Another mechanism is the recruitment of special in-
hibitor proteins, such as eukaryotic translation initia-
tion factor 4E transporter (4E-T), which interacts with
the translation initiation factors and inhibits transla-
tion initiation [96]. CCR4-NOT has also been demon-
strated to induce mRNA decapping and deadenylation,
leading to mRNA degradation.
Another mechanism through which 3′  UTRs ex-
ert their influence on mRNA decay is the presence
of adenylate/uridylate (AU)-rich elements (AREs) in
the 3′  UTRs. These sequences are considered to be
mRNA decay signals [94]. It has been demonstrated
that many RBPs, including tristetraprolin and butyrate
response factors 1 and 2, bind to AREs, resulting in
mRNA destabilization and degradation [97]. However,
VOLKHIN et al.730
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
not all AU-rich elements cause translational repres-
sion. Thus, the human antigen R protein (HuR) binds
AU-rich sequences in the 3′ UTRs, thereby increasing
the stability of the corresponding mRNAs by protecting
them from miRNA binding through steric hindrances
[98]. The precise mechanism of ARE influence on the
mRNA stability depends on the stoichiometry between
AREs and RBPs, as well as the 3′  UTR structure and
arrangement of regulatory elements. Furthermore,
mRNA cleavage can be facilitated by lncRNAs. For in-
stance, 3′  UTRs of some mRNAs contain Alu elements
that can pair with certain lncRNAs, resulting in the
formation of double-stranded RNAs that undergo deg-
radation via the STAU1 (staufen double-stranded RNA
binding protein 1)-mediated mechanism [99].
Other molecules that can promote mRNA degra-
dation are miRNAs which guide the miRNA-induced
silencing complex (miRISC) composed of the miRNA
and Argonaute protein [100]. miRISC binds to special-
ized sites on mRNAs, known as miRNA-binding sites
(miRBSs), and recruits effector protein complexes, such
as PAN2-PAN3 and CCR4-NOT, that inhibit translation
and cause mRNA degradation. Therefore, to ensure a
greater mRNA stability, it is important to avoid the
degradation signals (AREs, Alu elements, and miRBSs)
in the 3′  UTRs. However, the effect of such signals de-
pends on the intricate cellular signaling network and
can be almost unpredictable. Interestingly, Leppek
etal. [67] have shown that the 5′  UTRs have a greater
effect on the mRNA stability than the 3′  UTR. This is
surprising because there is a plethora of mechanisms
by which the 3′ UTR can influence the mRNA lifespan.
To the best of our knowledge, the reason for this phe-
nomenon is unknown. The 3′  UTRs can either inhibit
or promote translation. For instance, the 3′  UTRs of
mRNAs coding for some nuclear-encoded mitochondri-
al proteins (e.g., ATPase subunits) promote translation
due to the interactions with the translation machinery
[60, 101]. Another example is the woodchuck hepatitis
virus post-transcriptional regulatory element (WPRE),
which is frequently used in gene therapy to increase
protein production. While its main function is to pro-
mote the nuclear export of mRNAs [102], Loeb et al.
[103] suggested that WPRE can also activate transla-
tion via an unknown mechanism.
UTR DESIGN FOR TISSUE-SPECIFIC EXPRESSION
Approximately half of human genes produce mul-
tiple mRNA isoforms that can vary in their 3′  UTR
sequences [104]. Most mRNAs with a single isoform
are transcribed in a tissue-restricted manner, whereas
ubiquitously expressed genes are typically transcribed
into multiple mRNA isoforms, depending on the tis-
sue context. This tissue-dependent diversity of 3′  UTRs
can be potentially exploited to develop tissue-specific
gene therapies. Limiting expression of transgenes in
non-target tissues is critical for preventing the toxic
effects of protein overexpression and immunotoxicity.
Theoretically, this can be achieved by incorporating
tissue-specific miRBSs into 3′  UTRs in order to restrict
transgene expression in the cells enriched in the cor-
responding miRNA (Fig.  3). For example, it was re-
ported that adding the binding sites for miR-122 and
miR-1 to rAAV9 promoted protein synthesis in the CNS
while reducing its expression in the liver, heart, and
skeletal muscles. The incorporation of miR-204 and
miR-124 targeted transgene expression to the photo-
receptors and retinal pigment epithelium, respectively
[27, 105-108].
miRNA targeting is an effective way to restrict
the transgene expression to specific tissues based on
different miRNA profiles of different cell lineages.
Ludwig et al. [109] created a comprehensive human
miRNA tissue atlas by determining the abundance of
1997 miRNAs in 61 tissue biopsies of different organs.
One thousand three hundred sixty-four miRNAs were
discovered in at least one tissue, 143 were present in
each tissue. The majority of miRNAs displayed inter-
mediate tissue specificity, with some expressed pre-
dominantly in specific tissues, such as the brain [109].
Recent studies have shown that the tissue tropism of
certain viruses could be eliminated by the incorpora-
tion of miRNA target sequences into their genomes to
prevent viral RNA translation in the tissues expressing
tissue-specific miRNAs. Vesicular stomatitis virus is
considered as a promising recombinant vaccine plat-
form and oncolytic agent. However, it has not yet been
tested in humans, as it was found to cause enceph-
alomyelitis in rodents and primates by almost any
route of administration. Kelly et al. [110] attempted
to eliminate this effect by creating an attenuated vi-
ral strain by inserting neuron-specific miRBSs into the
virus 3′  UTR. The new viral strain retained its original
oncolytic properties, but lost the neurotropic effect,
which allows its use in clinical trials. This approach
has been successfully applied to the poliovirus [111]
and mosquito-borne Japanese encephalitis virus [112].
The modified strains retained their ability to replicate
in non-neuronal tissues, but their neurotropism de-
creased significantly, leading to the development of
safer and more efficient vaccines.
5′ UTRs may display tissue specificity due to the
alternative splicing and formation of multiple vari-
ants of the first exon encoding different 5′  UTRs [113,
114], the use of multiple promoters [115,  116], or a
combination of these mechanisms. While the result-
ing mRNAs have identical coding regions, the pres-
ence of different 5′ UTRs affects the translational ef-
ficiency, leading to the tissue-specific expression of
the same gene. However, this regulatory mechanism
SELECTION OF UTRs 731
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig. 3. Principles of tissue-specific 3′ UTR design. Although this approach has been successfully demonstrated for AAV
vectors, it can also be applied to mRNA therapies. a) The AAV vector expression cassette contains the binding sites for
tissue-specific miRNAs in the 3′ UTR. Incorporation of miRBSs for miRNAs present in the non-target tissues or cells allows
to reduce off-target transgene expression. b) The AAV vector is internalized into the cell and enters the nucleus, where
the transgene mRNA is transcribed. After mRNA transcription and nuclear export, tissue-specific miRNA in the content of
miRISC binds to miRBSs in the synthesized mRNA, resulting in silencing due to the translational repression and/or mRNA
degradation in the off-target cells.
is not directly applicable to the development of mRNA
vaccines or gene therapies, as the 5′ UTRs in such con-
structs are synthetic and are not the subjects to en-
dogenous transcriptional regulation. Once an mRNA
enters the cell, transcription no longer occurs and
translation depends on the cellular environment and
properties of mRNA itself. Although this mechanism
is informative from a biological point of view, it can-
not be used for tissue targeting in the development
of mRNA-based therapeutics.
The major challenge in protein replacement ther-
apy is the immune response triggered by heterologous
proteins, which can undermine the efficacy of thera-
py. For example, AAV vectors can deliver transgenes
not only to target cells, but also to antigen-presenting
cells (APCs). APCs process foreign proteins and ini-
tiate the immune response. A promising strategy to
mitigate this issue involves incorporation of binding
sites for APC-specific miRNAs, such as miR-142 and
miR-652, into the rAAV expression cassette to redirect
the transgene expression away from the APCs. The
miRBSs engineered for the transgene can interact with
cellular miRNAs, leading to the transcript degradation.
In APCs, such constructs can prevent the presentation
of transgene peptides to T cells, thus reducing immu-
nity against the transgene protein [117-119]. PARADE
(Prediction and RAtional DEsign of mRNA UTRs) is a
recently developed promising tool [120] for designing
UTRs regions with a tailored cell type-specific activ-
ity. To develop this generative artificial intelligence
framework, the regulatory activity of 60,000 5′ and
3′  UTRs was measured in six cell types. The PARADE
tool was validated by testing 15,800 de novo designed
sequences in six cell types. Many sequences showed
superior specificity and activity compared to existing
RNA therapeutics. The PARADE tool can potentially
VOLKHIN et al.732
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
facilitate the design of tissue-specific mRNAs for var-
ious therapies by mitigating the negative effects of
protein expression in non-target organs.
THE IMPACT OF MODIFIED
NUCLEOTIDES IN THE UTRs
The dynamic regulation of post-transcriptional
modifications (PTMs) is mediated by a complex net-
work of ‘writer’, ‘reader’, and ‘eraser’ proteins [121].
PTMs play a critical role in RNA preprocessing in the
nucleus, including alternative splicing and nuclear
export. In the cytoplasm, PTMs modulate important
aspects of the transcript life cycle, such as intracellu-
lar localization, translational regulation, stability, and
degradation [122].
Current studies on the epigenetic regulation of
RNAs by PTMs primarily focus on their impact in
cancer therapy [123] and involvement in the devel-
opment of complex neurological, cardiovascular, and
metabolic diseases [122, 124]. However, PTMs may
also hold significance for the future development of
mRNA-based therapeutics. Thus, Pfizer and Moderna
used PTMs in the development of the COVID-19 mRNA
vaccine [125]. The mRNA component of the vaccine
was composed of the 5′ UTR, the coding sequence for
the spike protein with two consecutive stop codons,
and the 3′ UTR in which all uridines have been re-
placed by N1-methylpseudouridine.
The distribution of nucleotide modifications across
different regions of mRNA varies significantly. For ex-
ample, pseudouridines are more prevalent in the 3′
UTRs and coding regions than in the 5′ UTRs [126].
In  vivo isomerization of uridine to pseudouridine is
carried out by pseudouridine synthases (PUSs). PUSs
catalyze the formation of pseudouridine by breaking
the N1–C1′ glycosidic bond and forming a new C5–
C1′ bond, with the N1 atom becoming an additional
hydrogen bond donor. The resulting conformational
changes contribute to the stabilization of the polynu-
cleotide chain [127]. mRNAs containing pseudouridine
and 1-methylpseudouridine have a reduced ability to
activate the innate immune system because they are
less likely to be detected by receptors, such as the
Toll-like receptor  3 (TLR3) and retinoic acid-inducible
gene  1 (RIG-I) [3,  128,  129]. According to predictions
from the convolutional neural network trained on
unmodified sequences, the incorporation of pseudou-
ridine and 1-methylpseudouridine into the 5′ UTRs
correlates positively with the ribosomal loading due
to the increase in the minimum free energy and alter-
ations in the RNA secondary structure [130]. However,
N1-methylpseudouridine induces ribosome pausing
and leads to a +1 frameshift [5]. Pseudouridylation
is associated with the enhanced mRNA translational
efficiency. Thus, it was found that PUS7 promotes the
translation of the tumor suppressor ALKBH3 in gastric
cancer [131]. The translational efficiency of modified
transcripts depends on the extent of modification:
the transcripts containing over 50% N1‐meth-
ylpseudouridine substitutions yielded less protein than
those with lesser percentage of modification [129]. As
demonstrated in a recent study, N1-methylpseudouri-
dine in a context-dependent manner affected the de-
coding of the mRNA sequence by altering the strength
and pattern of the mRNA–tRNA interactions in the
ribosome  A site [132]. 5-Methoxyuridine (5moU), an
alternative modification of uridine, increased the sta-
bility, reduced immunogenicity, and enhanced protein
yield for in vitro transcribed (IVT) mRNA [133].
When located in different transcript regions, the
same modification can perform different biological
functions. For example, the enrichment of the start
codon, stop codon, and 5′  UTR (particularly at the crit-
ical cap+1 and cap+2 positions) with N
1
-methylade-
nosine (m
1
A) enhances the translational efficiency,
whereas modification of the coding sequence and the
3′  UTR reduced the efficiency of translation [134-136].
It was also found that m1A in the 3′  UTR impacts the
interaction between mRNA and miRNAs; however, the
underlying mechanisms of this influence are yet poor-
ly understood [137]. The m
1
A modification enhances
the effect of N
6
-methyladenosine (m
6
A) by inducing
the RNA degradation pathway [138]. The m1A sites at-
tract HRSP12 (heat-responsive protein 12), which facil-
itates the binding of the m
6
A reader protein YTHDF2
(YTH domain family  2) to m
6
A sites on the same RNA
molecule. The HRSP12–YTHDF2 complex recruits the
P/MRP endoribonuclease that cleaves mRNA thus sup-
pressing translation [139, 140]. The m
6
A modification
promotes destabilization of the RNA secondary struc-
ture [141]. Early studies have indicated that its pres-
ence in the 5′  UTR near the start codon facilitates the
cap-independent translation. However, recent findings
show that the thermodynamic changes resulting from
a single modification are insufficient to stabilize the
initiation complex and do not affect the translational
efficiency [142]. The effect of the m
6
A modification
on the translation initiation may be mediated by the
reader protein YTHDF1. YTHDF1 binds to m
6
A sites
near the 3′ UTR proximal region and, through loop
formation, interacts with eIF3. YTHDF3 interacts
with YTHDF1 to enhance its translation-promoting
effect [143].
N4-acetylcytidine (ac4C) had been originally iden-
tified in tRNA and rRNA; its presence mRNA was
confirmed later. This PTM plays a critical role in reg-
ulating the transcript stability and gene expression.
Overexpression of NAT10, an enzyme catalyzing ac4C
modification, was found to promote proliferation of
malignant cell in many cancer types [144]. It has been
SELECTION OF UTRs 733
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
repeatedly demonstrated that ac4C, which predomi-
nates in the 3′  UTR and surrounds the stop codon, can
stabilize mRNA [145, 146]. The presence of ac4C in the
coding sequence is associated with the upregulated ex-
pression of the corresponding mRNA [147]. Also, the
presence of ac4C in the 5′ UTR influences translation
initiation by suppressing the usage of strong start co-
dons and facilitating alternative initiation at weaker
sites [148].
NSUN proteins are a family of RNA methyltrans-
ferases that catalyze the formation of 5-methylcytosine
(m
5
C), thus playing a critical role in RNA metabolism.
m
5
C is found throughout the transcripts, with a higher
concentration in the 5′ UTRs. Interestingly, m
5
C-con-
taining mRNAs demonstrate a reduced translational
efficiency, despite longer half-life [149]. The content of
N
7
-methylguanosine (m
7
G) in the coding sequence and
3′  UTR increases after heat shock and oxidative stress,
while the content in the 5′  UTR decreases, which con-
tributes to the increased translational efficiency of
such mRNAs [150]. m
7
G recruits initiation factors and
protects RNAs from 5′  exonucleases [151].
Therefore, incorporating modified bases in the
UTRs and coding sequencing, as well as taking into
account the functional properties of modified nucle-
otides, including their ability to trigger or attenuate
the immune response, regulate translation rate and
efficiency, and increase mRNA stability, can improve
the development of therapeutic mRNAs. However, it
is important to point out that the existing data are
limited, and further research is needed for a more
comprehensive understanding of the mechanisms un-
derlying the effect of modified bases.
UTRs IN RESEARCH
AND CLINICAL TRIALS
The development of gene therapeutics and mRNA
vaccines can be compared to the construction of a
building from different ‘building blocks’, for example,
3′ and 5′  UTRs, which can be tailored and combined
with the coding sequences to optimize the resulting
product.
One strategy to increase the expression efficiency
and stability of IVT mRNAs is the use of UTRs from
highly translated, constitutively expressed, and sta-
ble cellular endogenous genes  [67]. To achieve high
translational efficiency, many mRNA therapies uti-
lize the well-studied 3′ and 5′  UTRs of the human α-
and β-globin genes [68,  152-159] and rabbit β-globin
genes [160], as they improve the translation efficien-
cy and increase the stability of heterologous mRNAs
[16, 161-163].
It is believed that the 5′ UTR of β-globin mRNA
ensures efficient cap-dependent translation due to the
lack of stable secondary structures or translation-sup-
pressing elements [68]. The 3′  UTRs of the α- and β-glo-
bin mRNAs provide a high stability of mRNA due to
association with RBPs [164-168]. For this reason, a
combination of 5′ and 3′  UTRs from the globin mRNAs
is widely used in the studies of translation and stabil-
ity of IVT mRNA [67, 157], creation of vaccines for im-
munotherapy [152, 153, 161, 169], and cell reprogram-
ming into induced pluripotent stem cells (iPSCs) [154].
Two 3′  UTRs the from human β-globin mRNA cloned
head to tail were found to be superior to a single
3′  UTR in improving mRNA translation and stability,
inducing immune responses in mice. The efficiency of
such constructs is currently tested in ongoing clinical
trials (ClinicalTrials.gov: NCT01684241, NCT02035956,
NCT02316457, NCT02410733, and NCT03418480)  [170].
In addition to the widely used α- and β-globin
UTRs, UTRs from other genes can also be incorporated
into mRNAs for mRNA-based therapeutics. Some natu-
ral UTRs have been shown to improve the efficiency
of the corresponding products. For example, the UTRs
of the TSMB10, TMSB4X[171]; HBB, H4C2, Rabb, TPL
[163], mtRNR1, AES [172]; CYBA [173], Rps27a [44],
C3, CYP2E1, and APOA2 [63] genes, viral UTRs (e.g.,
from dengue virus) [174], TEV and TMV 5′ UTRs  [67,
175], and many others have been found to improve
the efficacy of therapeutics and can be potentially
used in developing the UTR design strategies aimed
to improve the mRNA translational efficiency. The
IRES (internal ribosome entry site)-containing 5′  UTR
from the encephalomyocarditis virus enhanced tran-
scription of uncapped mRNAs and increased transgene
expression in human and murine cultured dendritic
cells (DCs). Immunization of mice with DCs transfected
with the IRES-containing mRNA induced high levels
of antigen-specific T  cells, promoted elimination of
antigen-bearing cells, and protected immunized mice
from pulmonary metastasis of melanoma cells injected
intravenously, suggesting a potential use of IRES-con-
taining 5′  UTRs in the antitumor immunotherapy [176].
The IRES was also included in the IVT mRNA used in
the reprogramming of human fibroblasts into plurip-
otent stem cells [177]. Some regulatory motifs, such as
translation initiator of short 5′ UTRs (TISU)  [178], may
also enhance mRNA translation. However, all publi-
cations indicated that the observed effects were cell
type-specific [163]. The list of studied 3′ and 5′ UTRs
and their effects on the translational efficiency and
mRNA stability are summarized in Tables 1 and 2.
Due to the emergence of new computational meth-
ods and machine learning, artificially generated UTRs
with specified properties can be currently developed
to provide an increased mRNA stability and efficient
protein production compared to natural endogenous
UTRs. Thus, the creation of artificial 5′  UTRs ensuring
high translational efficiency was described in [185].
VOLKHIN et al.734
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Table 1. Studied 5′ UTRs and their impact on the translational efficiency and mRNA stability
Selected 5′ UTR Advantages
5′ UTR
used for
comparison
In vitro/
in vivo
model
Origin and length References
α-globin
α-globin UTR demonstrated
higher translational
efficiency in living cells,
whereas lower values
were observed
in in vitro lysates
β-globin Cos-7 cells
origin:
human α-globin mRNA
length:
34 nucleotides (nt) [179]
[68, 180]
α-globin UTR enhanced
mRNA stability and
protein translation
CYBA
ACE2
A549 and
HepG2 cells
[164]
Human Hsp70
UTR increased the
efficiency of mRNA
translation in cultured
cells and enhanced
antibody response in vivo
Grp 78
VEGF
HepG2 cells
Balb/c mice
origin:
human Hsp70 mRNA
length: 213 nt
[181]
HBB
HSPA1A
Rabb
H4C2
UTRs increased
the translational efficiency
and protein level
5′ UTR of
mRNA-1273
(Moderna)
5′ UTRs of
BNT162b2
(BioNTech)
C3
DC2.4 cells
HBB
origin:
human β-globin mRNA
length: 57 nt
HSPA1A
origin: human heat shock
70 kDa protein 1 mRNA
length: 227 nt
Rabb
origin:
rabbit α-globin mRNA
length: 66 nt
H4C2
origin: histone H4 mRNA
length: 42 nt
[163]
Adenoviral
tripartite leader
sequence (TPL)
UTR strengthened
the antigen-specific
T-cell response and
increased the number
of IFNγ-secreting cells
in immunized mice
5′ UTR of
mRNA-1273
(Moderna)
C57BL/6Cit
and
I/StSnEgYCit
mice
origin: tripartite
leader sequence
of adenovirus mRNA
length: 245 nt
[141]
CYBA
5′ and 3′ UTRs
increased protein levels
independently and
in combination without
affecting the half-life
of mRNA transcripts.
DECR1
GMFG
MAPBPIP
MYL6B
NIH3T3 and
A549 cells
origin: human cytochrome
b-245 alpha chain mRNA
length: 39 nt
[44, 173]
5′ UTR
of S27a-44
transcript
of Rps27a gene
UTR increased the
efficiency of transgene
expression
*S27a-44′ without the TOP
motif improved transgene
expression more efficiently
than the intact S27a-44.
CYBA
α-globin
S27a-45
transcript
of Rps27a
Hep3B and
293T cells
origin: modified UTR from
the ribosomal protein
S27a mRNA
length: different variants
[44]
SELECTION OF UTRs 735
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Table 1 (cont.)
Selected 5′ UTR Advantages
5′ UTR
used for
comparison
In vitro/
in vivo
model
Origin and length References
NASAR
incorporation of NCA-7d as
the 5′ UTR and S27a as the
3′ UTR with the functional
motif R3U enhanced
antigen translation
CYBA
α-globin
Hep3B and
293T cells
origin:
synthetic sequence
length: 70 nt
[44]
C3
CYP2E1
in vitro transcribed
mRNAs containing these
5′ UTRs demonstrated
improved translational
efficiency regardless
of  the 3′ UTRs present
ASL
ALB
FGA
ORM1
HPX
AGXT
APOA2
HepG2 cells
C3
origin: complement factor
3 mRNA
length: 92 nt
CYP2E1
origin: cytochrome C450
2E1 mRNA
length: 37 nt
[63]
Mouse COL1A2
Hoxa9 P4
Rpl18a TOP
RBCS1A
TMV
TEV
HCV
chimeric fusion
of hHBB 5′  UTR
with TEV or TOP
sequence
DEN2 (alone and
in combination
with DEN2
3′  UTR)
5′ UTRs of these genes
ensured higher ribosome
loading and translational
efficiency than the 5′  UTR
of human β globin gene
hHBB
HEK293T
cells
mouse COL1A2
origin: mouse Col1a2 gene
mRNA
length: 137 nt
Hoxa9 P4
origin: mouse Hoxa9
IRES-like element pairing
element 4
length: 323 nt
Rpl18a TOP
origin: ribosomal protein
18a with 5′ terminal
oligopyrimidine motif
length: the length of
Rpl18a is 89 nt; the TOP
motif is a cis-regulatory
RNA element that begins
directly after the m
7
G cap
structure and contains
the hallmark invariant
5′-cytidine followed
byan uninterrupted tract
of 4-15 pyrimidines
RBCS1A
origin:
Arabidopsis thaliana
ribulose bisphosphate
carboxylase small chain
1A mRNA
length: 418 nt
TEV
origin: Tobacco etch virus
mRNA
length: 47 nt
TMV DEN
origin: tobacco mosaic
virus UTR and dengue
virus UTR fusion
length: 68 nt for TMV
UTR; 96 nt for DEN UTR
[67]
VOLKHIN et al.736
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Table 1 (cont.)
Selected 5′ UTR Advantages
5′ UTR
used for
comparison
In vitro/
in vivo
model
Origin and length References
TMSB10
UTR provided higher
levels of gene expression
in cultured cells;
when administered
intramuscularly
to mice as a component
of mRNA vaccine,
the UTR promoted
the transgene
expression in whole
blood, spleen, and liver,
leading to enhanced
specific humoral and
cellular immune
responses
against the antigen
5′ and
3′ UTRs
of TMSB4X
B2M
ACTB
MTRNR2L2
HLA-DRA
HLA-DRB1
FTL
FTH1
DC2.4 cells
origin:
thymosin beta 10 mRNA
length: 78 nt
[171]
R27-UTRs
DC2.4 cells
BALB/c mice
[171]
TISU (translation
initiator of short
5′ UTR)
mRNA with TISU UTR
demonstrated better
expression than mRNAs
containing the Kozak
sequence only,
no 5′  UTR,
or eIF4G aptamer
as a 5′  UTR
invivo translation
efficacy was comparable
to that of mRNA
from the SpikeVax
vaccine with
5′ and 3′  UTRs
Kozak
sequence
eIF4G
aptamer
mRNA
containing
5′ and
3′ UTRs of
SpikeVax®
vaccine
HEK-293
cells
peripheral
blood
mononuclear
cells
human
T cells
C57BL/6 mice
length: minimum 4 nt [178]
Table 2. Studied 3′ UTRs and their effect on the translational efficiency and mRNA stability
3′ UTR Advantages
3′ UTR used
for comparison
In vitro/
in vivo model
Origin
and length
References
HBB-HBB
two 3′ UTRs of human
β-globin fused head to tail
improved the transcript
stability and protein yield,
increased the number of
antigen-specific peptide/MHC
complexes on the cell surface,
and improved expansion
of antigen-specific T cells
in vivo after injection of
transfected bone marrow-
derived DCs (BMDCs)
in mice
single β-globin
3′ UTR
poly-A tail
only, no UTR
human
immature and
mature DCs
EL4 cells
BMDCs
C57Bl/J6 mice
origin:
human
β-globin
mRNA
length:
240 nt [163]
[52, 182]
SELECTION OF UTRs 737
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Table 2 (cont.)
3′ UTR Advantages
3′ UTR used
for comparison
In vitro/
in vivo model
Origin
and length
References
HBVpA
BGHpA
HBVpA UTR increased the level
of in vitro antigen production
and antibody titer after
immunization of BALB/c mice
more efficiently than BGHpA
and betapA UTRs
HBVpA,
BGHpA, rabbit
β globin
betapA
CHO cells
BALB/c mice
HBVpA
origin:
hepatitis B
virus
length:
50-100 nt
[183]
CYBA
incorporation of 3′ and 5′ UTRs
enhanced protein translation
but did not increase the half-life
of mRNA transcripts.
DECR1
GMFG
MAPBPIP
MYL6B
NIH3T3
and A549 cells
origin:
human
cytochrome
b-245 alpha
chain mRNA
length:
200 nt
[52, 153]
Rps27a (S27a)
UTR increased translational
efficiency and protein expression
CYBA
TF
AAT
HCV
α-globin
Hep3B
and 293T cells
origin:
human 40S
ribosomal
protein S27a
mRNA
length:
170 nt
[44]
NASAR
incorporation of NCA-7d asthe
5′  UTR and S27a as 3′  UTR
with the R3U functional motif
promoted antigen
translation
CYBA
α-globin
Hep3B
and 293T cells
origin:
synthetic
sequence
length:
175-190 nt
[44]
mtRNR1-AES
AES-mtRNR1
UTRs increased protein
production and extended mRNA
half-life in cells, provided
higher splenic expression
of luciferase and stronger
CD8
+
T cell immune response
after intravenous vaccination
of BALB/c mice, and improved
the reprogramming of human
foreskin fibroblasts (HFFs)
into iPSCs
HBB
HBB-mtRNR1
mtRNR1
HBB-HBB
human DCs
C2C12 cells
HFFs
BALB/c mice
mtRNR1-AES
origin:
synthetic
sequence
length:
300 nt
AES-mtRNR1
origin:
synthetic
sequence
length:
300 nt
[172]
AES-AES
mtRNR-
mtRNR
UTRs provided higher
translational efficiency
AES-HBB
mtRNR-HBB
mRNA-1273
BNT162b2
DC2.4 cells
AES-AES
origin:
synthetic
sequence
length:
360 nt
mtRNR-
mtRNR
origin:
synthetic
sequence
length:
360 nt
[172]
VOLKHIN et al.738
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Table 2 (cont.)
3′ UTR Advantages
3′ UTR used
for comparison
In vitro/
in vivo model
Origin
and length
References
DEN2 (alone
and in
combination
with DEN
5′  UTR)
UTR promoted higher ribosome
loading and translational
efficiency independently;
combining it with the DEN
5′  UTR had an additive effect
on the translation efficiency
and mRNA stability
HBB
CYBA
HBA1
HEK293T cells
Origin:
dengue virus
Length:
451 nt [184].
[67]
rabbit α-globin Vero cells [174]
3′DENΔSL BHK cells [184]
TMSB10
UTR provided higher levels
of gene expression in cultured
cells, as well as in whole
blood, spleen, and liver after
intramuscular injection to mice,
leading to enhanced specific
humoral and cellular immune
responses against the antigen
5′ and 3′ UTRS
of TMSB4X
B2M
ACTB
MTRNR2L2
HLA-DRA
HLA-DRB1
FTL
FTH1
DC2.4 cells
origin:
human
TMSB10
mRNA
length:
240 nt
[171]
SV40
UTR enhanced
transgeneexpression
Hsp70
HepG2,
Hep3B, and
HEK293 cells
origin:
simian virus
40 large
T antigen
length:
different
variants
[171]
A recent study reported a series of minimal-length
(12-14 nt) synthetic 5′ UTRs that provided higher ex-
pression levels than the full-length human α-globin
5′  UTR in cell lines and in vivo [186]. Deep learning
and massively parallel reporter assay (MPRA) screen-
ing of a large set of different UTRs were used to pre-
dict the effect of different 5′  UTRs on the translational
efficiency to facilitate the design of 5′  UTRs for the
optimal protein production [130,  187]. Furthermore,
UTR optimization resulted in the optimal combination
of endogenous and denovo engineered 5′ and 3′  UTRs
(named NASARs) that were 5-10 times more efficient
than the tested endogenous CYBA and α-globin UTRs
[44]. Three synthetic 5′  UTRs were identified that sig-
nificantly outperformed natural 5′ UTRs in  [15]. Ar-
tificial UTRs were also used to engineer mRNAs for
cell reprogramming and vaccine development [154,
188-190]. Based on the above information, the use of
endogenous UTRs in combination with the de novo
design may be the most efficient approach to UTR
engineering capable of facilitating the development
of mRNA therapeutics [44]. Despite the proven effec-
tiveness of these UTRs, there is a lack of research data
on their comparative efficacy, as well as on selection
of particular UTRs best suited for specific applications.
The absence of standardization and comprehensive
control in the selection of UTRs for gene therapeutics
and mRNA vaccines is a significant challenge. While
some natural UTRs are known to improve the product
quality, their use is often based on precedent rather
than systematic evidence.
Even if a particular UTR outperforms another
UTR with the same coding region, there is no guar-
antee that this will be true for other coding sequence.
This context dependency emphasizes the importance
of interpreting the statements about the “best” UTRs
with caution. So far, no study has systematically com-
pared all possible UTR combinations, which illustrates
a serious gap in understanding the possible approach-
es for optimizing UTR selection for gene therapies
and vaccines. UTRs currently tested in clinical trials
are often derived from natural sequences, especially
those that have shown functional benefits in past re-
search or vaccine development. Commonly used UTRs
include those from the α- and β-globin genes, which
are known for their favorable impact on mRNA stabil-
ity and translational efficiency. These sequences have
been successfully used in approved RNA vaccines,
e.g., vaccines against COVID-19 [191].
However, the selection process for UTRs remains
largely empirical, with many decisions based on a
precedent rather than on rigorous comparative stud-
ies. The lack of a standardized framework or algo-
rithm for the UTR selection represents a significant
SELECTION OF UTRs 739
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
bottleneck in the development of RNA-based thera-
peutics. As UTRs strongly affect the mRNA stability,
translational efficiency, and interactions with regula-
tory molecules (e.g., proteins, miRNAs), their under-
explored potential may hinder innovation in gene
therapy and vaccine development.
Systematic strategies are needed to close this
gap, such as the high-throughput screening of UTR
libraries [15], computational modeling to predict the
UTR behavior in different contexts, and establishment
of standardized criteria for the UTR design. These ef-
forts could help to develop a more rational and in-
formed approach to the UTR selection, which will ul-
timately accelerate the progress in the field of mRNA
therapeutics.
UTR DESIGN
Although regulatory motifs and the secondary
structure of UTRs have no coding function, they sig-
nificantly influence the translational efficiency and
the half-life of transcripts. The design of UTRs for
the development of therapeutic molecules is based
on the inclusion of regulatory elements from natural
sequences flanking highly expressed genes. For exam-
ple, for the development of mRNA vaccines, Pfizer/
BioNTech utilized a 5′  UTR fragment from the human
α-globin mRNA in combination with the modified
Kozak sequence [191]. Although the understanding of
regulatory elements in the UTRs is still incomplete,
it has been shown that an efficient 5′  UTR design re-
quires formation of a less structured region upstream
of the start codon [68, 192, 193] to promote translation
initiation by improving the ribosome accessibility.
It should be mentioned that the results of UTR
efficiency tests performed in vivo cannot always be
extrapolated to the in vitro mRNA transcription sys-
tems. For example, it was shown that the 5′  UTR of
the CYBA mRNA provided a higher transcription of
the reporter gene in vivo compared to its transcrip-
tion in  vitro [179]. In other words, the same mRNA
can have different properties in cells and cell-free
solutions, which can be explained by the presence in
living systems of a large number of factors that can
interact with mRNA.
To summarize, in both basic and applied research,
ORFs are typically combined with UTRs whose ability
to increase the translation efficiency has been already
demonstrated. In some cases, these constructs are in-
deed more efficient compared to the wild-type mRNAs,
thus expanding the list of UTRs positively affecting the
mRNA productivity. However, this approach is not sys-
tematic; rather it is a naive enumeration of possible
options. New synthetic UTR sequences can be devel-
oped using machine learning approaches, which rely
on training datasets of sequences with different UTRs,
whose efficiency in protein synthesis has been mea-
sured. Based on this information, the machine learn-
ing methods can predict new UTR sequences outper-
forming natural UTRs. The quality of such predictions
strongly depends on the quality of training datasets.
Furthermore, the predicted variants should be validat-
ed experimentally. Recent computational approaches
to the UTR design are briefly described in Table 3.
EXPERIMENTAL VALIDATION
OF COMPUTATIONALLY PREDICTED UTRS
UTR optimization is a crucial step in the devel-
opment of mRNA-based therapeutics, as UTRs signifi-
cantly affect mRNA stability and protein production.
Several approaches have been developed to analyze
and compare the efficiency of UTRs, ranging from
traditional reporter gene systems to the state-of-the-
art high-throughput methods. Luciferase reporter as-
says are among the most commonly used techniques
for analyzing the UTR functionality. By coupling a
reporter gene (Luc2CP) to specific 3′  UTR sequences,
researchers can evaluate the kinetics of mRNA decay
and gain insight into the stability conferred by these
UTRs. This assay measures the functional half-life of
mRNAs by monitoring the activity of luciferase over
time, which ensures a straightforward and reliable
assessment of the UTR-mediated stability [172]. Ribo-
some profiling provides an overall view of translation-
al efficiency by mapping mRNA fragments protected
by ribosomes, thus allowing direct measurement of
UTR-mediated translational regulation. This technique
is especially useful for understanding ribosome stall-
ing, uORFs, and other regulatory elements that influ-
ence the translation rate [200]. Another approach is
polysome profiling [201], which is based on the sepa-
ration of translated mRNAs associated with polysomes
from untranslated mRNAs using a sucrose gradient.
MPRA enables a high-throughput evaluation of
UTR sequences by using synthetic mRNA libraries con-
taining a large number of UTR variants [202]. Each
variant is associated with a unique DNA barcode that
is incorporated into the reporter transcript. Cells are
transfected with the library containing multiple mRNA
variants. [203], the most efficient of which are select-
ed by polysome profiling [130, 204] or other methods
(e.g., translating ribosome affinity purification [205,
206]) to enrich actively translated mRNAs. Next, RNA
sequencing can be performed to identify the variants
with a high translational efficiency of the reporter
transcript. This approach enables simultaneous inter-
rogation of numerous UTRs to identify the sequence
elements that regulate the translational efficiency
and stability of mRNA. In addition, MPRA facilitate
VOLKHIN et al.740
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Table 3. Computational methods for UTR prediction
Name Purpose Approaches to model development Sequence length limit Availability References
FramePool
MRL (mean ribosome loads) prediction
for 5′  UTR, analysis of human 5′  UTR
variants, prediction of the effects
of translation initiation site located
upstream of the canonical start codon
one-hot encoding, convolution
neural network (CNN)
with framepooling*
any length
https://github.com/
Karollus/5UTR
[194]
Optimus
5-prime
MRL prediction for 5′  UTR,
development of synthetic 5′  UTR
CNN for MRL prediction;
genetic algorithm for evolving
new sequences
50 nt
https://github.com/
pjsample/
human_5utr_modeling
[16, 130]
Fast SeqProp
DNA, RNA, and protein design
(strong polyadenylation signals,
5′  UTRs, and enhancers.)
sequence optimization model with
VAE-regularization using APARENT,
Optimus 5, DragoNN, MPRA-
DragoNN, DeepSEA, and trRosetta
varying
https://github.com/
johli/seqprop
[195]
UTRGAN
development of synthetic 5′  UTR;
MRL and translation efficiency
optimization
Deep generative adversarial
network (GAN); gradient ascent
algorithm for optimization
with FramePool and MTtrans 3;
one-hot encoding
64-128 nt
https://github.com/
ciceklab/UTRGAN
[196]
DEN
development of synthetic 3′  UTRs,
splicing regulatory elements, enhancers
Deep exploration networks (DENs)
https://github.com/
johli/genesis
[197]
FunUV
prediction of functional 5′  UTR
and 3′  UTR variants
Gradient boosting
decision tree (GBDT)
50 nt for 5′  UTRs
and 100 nt for 3′  UTRs
https://github.com/
Wangxiaoyue-lab/FunUV
[198]
mRNA2vec
improvement of mRNA stability
and protein production
language model transformer
architecture-based embedding
for pre-training (using
5′  UTRs and ORFs
as input sequences):
average length, 459 nt;
for the downstream
task on 5′  UTR data:
average length, 91 nt
[199]
PARADE
design of 5′  UTRs and 3′  UTRs
for highly stable mRNAs with
a programmable cell type specificity
diffusion model; genetic algorithm;
random sampling;
motif-based design
https://github.com/
autosome-ru/parade
[120]
Note. * Frame pooling is a method used in CNNs that deal with multiple frames (such as shifts in nucleotide sequences). In this process, the network processes each
frame individually, typically by applying global pooling operations (such as max pooling and average pooling) to extract the most important features from each frame.
The results of these operations are then combined into a unified representation, preserving individual characteristics of each frame and the overall structure of the
input sequence. This enables the network to better understand patterns in genetic data.
SELECTION OF UTRs 741
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Table 4. Challenges and prospects of UTR engineering for gene therapy
Issue Challenges Prospects
Precise UTR
engineering
development of optimal UTR sequences
for specific therapeutic applications
is complicated due to the intricate
interactions between UTRs and cellular
factors which can affect mRNA stability,
localization, translational efficiency, and
other factors are critical for the control
of transgene expression
molecular mechanisms that control UTR
function need to be further explored; the
development of advanced computational
tools and invitro models is essential for
the prediction and optimization of UTR
sequences; machine learning algorithms
and high-throughput screening methods
can accelerate the design of UTRs tailored
to specific therapeutic outcomes
Long-term
safety and
efficacy
the long-term safety and efficacy of mRNA-
based gene therapies cannot be assured
based solely on the insilico data, including
predicted UTRs; unintended effects,
such as immune response or expression
of off-target genes, are difficult to predict
and pose a risk to patients
rigorous preclinical and clinical testing
conducted in compliance with the Good
Laboratory Practice and Good Manufacturing
Practice standards is essential to ensure
the safety of mRNA-based therapies; these
standards help to identify potential risks and
optimize the therapy protocols; in the context
of long-term monitoring in clinical trials,
novel technologies in the UTR design enable
the prediction of promising therapeutic
mRNAs at the beginning of selection process,
saving money and resources
Targeted gene
delivery
targeted gene delivery to specific tissues
or cell types is still a major issue; off-target
effects can reduce the therapeutic efficacy
and increase the risk of side effects of gene
therapies
engineering UTRs with sequences that
promote interaction with cellular receptors
or transmission vectors could improve tissue-
specific targeting; for example, inclusion
of tissue-specific miRBSs in the UTRs can
restrict gene expression to the desired
cell types; this approach would minimize
the off-target effects and improve
the therapeutic precision
Regulatory
and ethical
considerations
the regulatory environment for gene
therapies is complex, and UTR engineering
algorithms and approaches must meet
stringent safety and efficacy standards;
ethical considerations, such as equitable
access and potential misuse, must also
be taken into account
collaboration between researchers, regulatory
agencies, and industry stakeholders will
be crucial to manage the regulatory
process; transparent communication about
the benefits and risks of mRNA-based
therapies will help to gain public trust
and ensure ethical implementation of the
developed therapies
the investigation of ribosome-dependent and ribosome-
independent regulatory mechanisms, which makes it
an invaluable tool for the UTR optimization [187, 204].
Each of these experimental approaches has its own
strengths and limitations. Reporter assays are simple
and specific, but have a limited throughput. MPRA of-
fers a high-throughput screening of UTR variants, but
require advanced sequencing technologies. Ribosome
profiling and polysome profiling offer deep insights
into translation dynamics, however, these methods are
technically complex and resource intensive.
Together, these complementary approaches en-
able systematic assessment of UTRs and provide the
basis for rational mRNA design for both therapeutic
and research applications. Future advancements, par-
ticularly integration of machine learning and compu-
tational modeling, promise even greater precision in
the selection and optimization of UTRs.
CHALLENGES AND PROSPECTS
Although UTR engineering is a promising ap-
proach in gene therapy, a number of issues still re-
main to be resolved to fully realize its potential. These
include precise UTR engineering, the long-term safety
and efficacy of gene therapy, targeted gene delivery,
and ethical considerations (Table 4).
VOLKHIN et al.742
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
CONCLUSION
Despite a success in the identification of UTR vari-
ants affecting mRNA translation, the rational design
of UTRs continues to be a subject of active research.
For example, high-throughput screening of 5′  UTR
libraries by recombinase-mediated integration has
shown that certain UTR variants can significantly
increase expression of the transgenic GFP protein in
HEK293T cells [15]. In addition, optimization of UTR
sequences can be used for controlling the translation-
al capacity of mRNA to ensure the optimal protein
synthesis without exceeding the toxicity limits. For
example, altering the miRBSs in 3′  UTRs suppressed
the expression of toxic transgenes, which in turn re-
duced the cytotoxicity and increased the AAV vector
yield in HEK 293 cells  [207]. The modular nature of
mRNA allows to combine the selection of UTRs with
other approaches aimed to improve gene therapies.
For example, a combination of the UTR library screen-
ing and codon optimization in the ORF improved the
gene therapy for phenylketonuria  [208]. In another
study  [209], the inclusion of an expanded promoter
and modification of 3′  UTR elements led to a signif-
icant reduction in the hepatic toxicity in the treat-
ment of Rett syndrome in a mouse model. These re-
sults highlight the potential of using UTR selection
in combination with other gene therapy approaches,
such as promoter design  [210] and codon optimiza-
tion  [211]. The targeting of therapeutic transgenes to
specific tissues also remains a crucial aspect of gene
therapy.
Transcriptional (using tissue-specific promoters)
and transductional (using viral serotypes) targeting
strategies are established methods in the develop-
ment of tissue-specific therapeutics. There are also
significant opportunities to improve the tissue spec-
ificity of vector-mediated gene expression through
the post-transcriptional regulation, including the
miRNA-mediated suppression. Engineered expression
cassettes containing miRBSs in the 3′  UTRs allow tar-
geted degradation of transgene mRNAs in cells ex-
pressing the corresponding miRNAs [108]. The effica-
cy of miRNA-regulated transgene expression systems
depends on the tissue-specific expression of the se-
lected miRNAs. Recently, miRNAs with suitable expres-
sion profiles for a specific tissue have been discovered
and described in [106, 212, 213].
The principle of action of mRNA-based gene
therapeutics and vaccines includes protein synthesis
in a eukaryotic cell. The process of translation large-
ly depends on the UTRs selected for the design of a
particular construct. Therefore, the rational design
of gene therapeutics and vaccines should involve the
stage of UTR selection in order to develop high-effica-
cy therapeutic agents.
Abbreviations. AAVs, adeno-associated viruses;
CCR4-NOT, carbon catabolite repression – negative on
TATA-less; DEN2, dengue virus; m
6
A, N
6
-methylade-
nosine; mRNA, messenger RNA; miRNA, microRNA;
miRBSs, miRNA-binding sites; ORF, open reading
frame; PTMs, post-transcriptional modifications; RBPs,
RNA-binding proteins; uORF, upstream open reading
frame; UTRs, untranslated regions.
Contributions. O.N.M., P.Yu.V., and A.A.D. de-
veloped the concept and supervised the study; I.A.V.
wrote the section “The effect of UTRs on the protein
yield”. A.Iu.P. wrote the sections “UTR design” and
“The impact of modified nucleotides in the UTRs”;
M.A.D. wrote the sections “UTR design for tissue-spe-
cific expression”, “UTRs in research and clinical tri-
als”, “Experimental validation of computationally
predicted UTRs”; D.S.S. wrote the sections “Introduc-
tion”, “UTR design for tissue-specific expression”, and
“UTRs in research and clinical trials”, and prepared
the figures; R.E.P. wrote the section “Challenges and
prospects”. O.N.M., P.Yu.V., and A.A.D. edited the man-
uscript. All authors contributed substantially to the
preparation of the article, read and approved the final
version of the manuscript before publication.
Funding. This study was supported by the Rus-
sian Science Foundation (grant No.23-64-00002).
Ethics approval and consent to participate. This
work does not contain any studies involving human
and animal subjects performed by any of the authors
Conflict of interest. The authors of this work de-
clare that they have no conflicts of interest.
REFERENCES
1. Qin, S., Tang,X., Chen, Y., Chen,K., Fan,N., Xiao,W.,
Zheng,Q., Li,G., Teng,Y., Wu,M., and Song,X. (2022)
mRNA-based therapeutics: powerful and versatile
tools to combat diseases, Signal Transduct. Tar-
get. Ther., 7, 166, https://doi.org/10.1038/s41392-022-
01007-w.
2. Chandra,S., Wilson, J.C., Good,D., and Wei, M.Q. (2024)
mRNA vaccines: a new era in vaccine development,
Oncol. Res., 32, 1543-1564, https://doi.org/10.32604/
or.2024.043987.
3. Andries,O., Mc Cafferty,S., De Smedt, S.C., Weiss,R.,
Sanders, N. N., and Kitada, T. (2015) N1-meth-
ylpseudouridine-incorporated mRNA outperforms
pseudouridine-incorporated mRNA by providing
enhanced protein expression and reduced immuno-
genicity in mammalian cell lines and mice, J. Con-
trol. Release, 217, 337-344, https://doi.org/10.1016/
j.jconrel.2015.08.051.
4. Anderson, B. R., Muramatsu, H., Nallagatla, S. R.,
Bevilacqua, P. C., Sansing, L. H., Weissman, D., and
Karikó,K. (2010) Incorporation of pseudouridine into
SELECTION OF UTRs 743
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
mRNA enhances translation by diminishing PKR ac-
tivation, Nucleic Acids Res., 38, 5884-5892, https://
doi.org/10.1093/nar/gkq347.
5. Mulroney, T. E., Pöyry, T., Yam-Puc, J. C., Rust, M.,
Harvey, R. F., Kalmar, L., Horner, E., Booth, L., Fer-
reira, A.P., Stoneley, M., Sawarkar, R., Mentzer, A. J.,
Lilley, K.S., Smales, C.M., von der Haar,T., Turtle,L.,
Dunachie, S., Klenerman, P., Thaventhiran, J. E. D.,
and Willis, A. E. (2024) N 1-methylpseudouridyla-
tion of mRNA causes +1 ribosomal frameshifting,
Nature, 625, 189-194, https://doi.org/10.1038/s41586-
023-06800-3.
6. Keeler, A. M., and Flotte, T. R. (2019) Recombinant
adeno-associated virus gene therapy in Light of Lux-
turna (and Zolgensma and Glybera): where are we,
and how did we get here? Annu. Rev. Virol., 6, 601-
621, https://doi.org/10.1146/annurev-virology-092818-
015530.
7. Nieuwenhuis, B., Laperrousaz, E., Tribble, J. R., Ver-
haagen,J., Fawcett, J.W., Martin, K.R., Williams, P.A.,
and Osborne, A. (2023) Improving adeno-associated
viral (AAV) vector-mediated transgene expression in
retinal ganglion cells: comparison of five promot-
ers, Gene Ther., 30, 503-519, https://doi.org/10.1038/
s41434-022-00380-z.
8. Tabebordbar, M., Lagerborg, K. A., Stanton, A., King,
E. M., Ye, S., Tellez, L., Krunnfusz, A., Tavakoli, S.,
Widrick, J. J., Messemer, K. A., Troiano, E. C.,
Moghadaszadeh, B., Peacker, B. L., Leacock, K. A.,
Horwitz, N., Beggs, A. H., Wagers, A. J., and Sabeti,
P. C. (2021) Directed evolution of a family of AAV
capsid variants enabling potent muscle-directed
gene delivery across species, Cell, 184, 4919-4938.e22,
https://doi.org/10.1016/j.cell.2021.08.028.
9. Goertsen, D., Flytzanis, N. C., Goeden, N., Chuapoco,
M. R., Cummins, A., Chen, Y., Fan, Y., Zhang, Q.,
Sharma, J., Duan, Y., Wang, L., Feng, G., Chen, Y., Ip,
N. Y., Pickel, J., and Gradinaru, V. (2022) AAV capsid
variants with brain-wide transgene expression and
decreased liver targeting after intravenous delivery
in mouse and marmoset, Nat. Neurosci., 25, 106-115,
https://doi.org/10.1038/s41593-021-00969-4.
10. Luo,L., Jea, J.D.Y., Wang,Y., Chao, P.W., and Yen,L.
(2024) Control of mammalian gene expression by
modulation of polyA signal cleavage at 5′ UTR, Nat.
Biotechnol., 42, 1454-1466, https://doi.org/10.1038/
s41587-023-01989-0.
11. Zhong, G., Wang, H., He, W., Li, Y., Mou, H., Tickner,
Z.J., Tran, M.H., Ou,T., Yin,Y., Diao,H., and Farzan,
M. A. (2020) reversible RNA on-switch that controls
gene expression of AAV-delivered therapeutics in vivo,
Nat. Biotechnol., 38, 169-175, https://doi.org/10.1038/
s41587-019-0357-y.
12. Luoni, M., Giannelli, S., Indrigo, M. T., Niro, A.,
Massimino, L., Iannielli, A., Passeri, L., Russo, F.,
Morabito, G., Calamita, P., Gregori, S., Deverman, B.,
and Broccoli, V. (2020) Whole brain delivery of an
instability-prone Mecp2 transgene improves be-
havioral and molecular pathological defects in
mouse models of Rett syndrome, Elife, 9, e52629,
https://doi.org/10.7554/eLife.52629.
13. Chu, Y., Yu, D., Li, Y., Huang, K., Shen, Y., Cong, L.,
Zhang, J., and Wang, M. (2024) A 5’ UTR language
model for decoding untranslated regions of mRNA
and function predictions, Nat. Mach. Intell., 6,
449-460, https://doi.org/10.1038/S42256-024-00823-9.
14. Castillo-Hair, S.M., and Seelig,G. (2022) Machine learn-
ing for designing next-generation mRNA therapeutics,
Acc. Chem. Res., 55, 24-34, https://doi.org/10.1021/
acs.accounts.1c00621.
15. Cao, J., Novoa, E. M., Zhang, Z., Chen, W. C. W.,
Liu, D., Choi, G. C. G., Wong, A. S.L., Wehrspaun, C.,
Kellis,M., and Lu, T.K. (2021) High-throughput 5′ UTR
engineering for enhanced protein production in non-
viral gene therapies, Nat. Commun., 12, 4138, https://
doi.org/10.1038/s41467-021-24436-7.
16. Castillo-Hair, S., Fedak, S., Wang, B., Linder, J.,
Havens, K., Certo, M., and Seelig, G. (2024) Opti-
mizing 5’UTRs for mRNA-delivered gene editing us-
ing deep learning, Nat. Commun., 15, 5284, https://
doi.org/10.1038/S41467-024-49508-2.
17. Hajj, K. A., and Whitehead, K. A. (2017) Tools for
translation: non-viral materials for therapeutic
mRNA delivery, Nat. Rev. Mater., 2, 17056, https://
doi.org/10.1038/natrevmats.2017.56.
18. Kon, E., Elia, U., and Peer, D. (2022) Principles for
designing an optimal mRNA lipid nanoparticle vac-
cine, Curr. Opin. Biotechnol., 73, 329-336, https://
doi.org/10.1016/j.copbio.2021.09.016.
19. Park, K.S., Sun,X., Aikins, M.E., and Moon, J.J. (2021)
Non-viral COVID-19 vaccine delivery systems, Adv.
Drug Deliv. Rev., 169, 137-151, https://doi.org/10.1016/
j.addr.2020.12.008.
20. Lu, R. M., Hsu, H. E., Perez, S. J. L. P., Kumari, M.,
Chen, G.H., Hong, M.H., Lin, Y.S., Liu, C.H., Ko, S.H.,
Concio, C. A. P., Su, Y. J., Chang, Y. H., Li, W. S., and
Wu, H.C. (2024) Current landscape of mRNA technol-
ogies and delivery systems for new modality thera-
peutics, J.Biomed. Sci., 31, 89, https://doi.org/10.1186/
s12929-024-01080-z.
21. Wadhwa, A., Aljabbari, A., Lokras, A., Foged, C., and
Thakur,A. (2020) Opportunities and challenges in the
delivery of mRNA-based vaccines, Pharmaceutics, 12,
102, https://doi.org/10.3390/pharmaceutics12020102.
22. Jones, C. H., Androsavich, J.R., So, N., Jenkins, M. P.,
MacCormack,D., Prigodich, A., Welch,V., True, J. M.,
and Dolsten,M. (2024) Breaking the mold with RNA
a “RNAissance” of life science, NPJ Genomic Med.,
9, 2, https://doi.org/10.1038/s41525-023-00387-4.
23. Baek, R., Coughlan, K., Jiang, L., Liang, M., Ci, L.,
Singh,H., Zhang,H., Kaushal,N., Rajlic, I.L., Van,L.,
Dimen,R., Cavedon,A., Yin,L., Rice, L., Frassetto,A.,
VOLKHIN et al.744
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Guey, L., Finn, P., and Martini, P. G. V. (2024) Char-
acterizing the mechanism of action for mRNA ther-
apeutics for the treatment of propionic acidemia,
methylmalonic acidemia, and phenylketonuria, Nat.
Commun., 15, 3804, https://doi.org/10.1038/s41467-
024-47460-9.
24. Lundstrom, K. (2023) Viral vectors in gene therapy:
where do we stand in 2023? Viruses, 15, 698, https://
doi.org/10.3390/v15030698.
25. Kishimoto, T.K., and Samulski, R.J. (2022) Addressing
high dose AAV toxicity – “one and done” or “slower
and lower”? Expert Opin. Biol. Ther., 22, 1067-1071,
https://doi.org/10.1080/14712598.2022.2060737.
26. Zwi-Dantsis, L., Mohamed, S., Massaro, G., and
Moeendarbary, E. (2025) Adeno-associated virus
vectors: principles, practices, and prospects in gene
therapy, Viruses, 17, 239, https://doi.org/10.3390/
V17020239.
27. Wang, J. H., Gessler, D. J., Zhan, W., Gallagher, T. L.,
and Gao, G. (2024) Adeno-associated virus as a de-
livery vector for gene therapy of human diseases,
Signal Transduct. Target. Ther., 9, 78, https://doi.org/
10.1038/s41392-024-01780-w.
28. Li, C., and Samulski, R. J. (2020) Engineering ade-
no-associated virus vectors for gene therapy, Nat. Rev.
Genet., 21, 255-272, https://doi.org/10.1038/s41576-
019-0205-4.
29. Von der Haar, T., Mulroney, T. E., Hedayioglu, F.,
Kurusamy, S., Rust, M., Lilley, K. S., Thaventhiran,
J. E., Willis, A. E., and Smales, C. M. (2023) Transla-
tion of in vitro-transcribed RNA therapeutics, Front.
Mol. Biosci., 10, 1128067, https://doi.org/10.3389/
fmolb.2023.1128067.
30. Buchlis, G., Podsakoff, G. M., Radu, A., Hawk, S. M.,
Flake, A.W., Mingozzi,F., and High, K.A. (2012) Fac-
tor IX expression in skeletal muscle of a severe he-
mophilia B patient 10 years after AAV-mediated gene
transfer, Blood, 119, 3038-3041, https://doi.org/10.1182/
blood-2011-09-382317.
31. Fischer, M. D., Simonelli, F., Sahni, J., Holz, F. G.,
Maier,R., Fasser, C., Suhner, A., Stiehl, D.P., Chen,B.,
Audo,I., and Leroy, B.P. (2024) Real-world safety and
effectiveness of Voretigene Neparvovec: results up
to 2 years from the prospective, registry-based PER-
CEIVE study, Biomolecules, 14, 122, https://doi.org/
10.3390/biom14010122.
32. Strauss, K. A., Farrar, M. A., Muntoni, F., Saito, K.,
Mendell, J.R., Servais,L., McMillan, H.J., Finkel, R.S.,
Swoboda, K.J., Kwon, J.M., Zaidman, C.M., Chiriboga,
C. A., Iannaccone, S. T., Krueger, J. M., Parsons, J. A.,
Shieh, P. B., Kavanagh, S., Tauscher-Wisniewski, S.,
McGill, B.E., and Macek, T. A. (2022) Onasemnogene
abeparvovec for presymptomatic infants with two
copies of SMN2 at risk for spinal muscular atrophy
type 1: the phase III SPR1NT trial, Nat. Med., 28,
1381-1389, https://doi.org/10.1038/s41591-022-01866-4.
33. Ferreira, V., Twisk, J., Kwikkers, K., Aronica, E.,
Brisson, D., Methot, J., Petry, H., and Gaudet, D.
(2014) Immune responses to intramuscular admin-
istration of alipogene tiparvovec (AAV1-LPLS447X)
in a phase II Clinical trial of lipoprotein lipase de-
ficiency gene therapy, Hum. Gene Ther., 25, 180-188,
https://doi.org/10.1089/hum.2013.169.
34. Ozelo, M. C., Mahlangu, J., Pasi, K. J., Giermasz, A.,
Leavitt, A. D., Laffan, M., Symington, E., Quon, D. V.,
Wang, J.-D., Peerlinck, K., Pipe, S. W., Madan, B.,
Key, N.S., Pierce, G. F., O’Mahony,B., Kaczmarek, R.,
Henshaw, J., Lawal, A., Jayaram, K., Huang, M.,
Yang, X., Wong, W. Y., and Kim, B. (2022) Valoctoco-
gene roxaparvovec gene therapy for hemophilia A,
N.Engl. J. Med., 386, 1013-1025, https://doi.org/10.1056/
nejmoa2113708.
35. Sekayan, T., Simmons, D. H., and von Drygalski, A.
(2023) Etranacogene dezaparvovec-drlb gene therapy
for patients with hemophilia B (congenital factor IX
deficiency), Expert Opin. Biol. Ther., 23, 1173-1184,
https://doi.org/10.1080/14712598.2023.2282138.
36. Xu, S., Yang, K., Li, R., and Zhang, L. (2020) mRNA
vaccine era – mechanisms, drug platform and clin-
ical prospection, Int. J. Mol. Sci., 21, 6582, https://
doi.org/10.3390/ijms21186582.
37. Mu,X., and Hur,S. (2021) Immunogenicity of invitro-
transcribed RNA, Acc. Chem. Res., 54, 4012-4023,
https://doi.org/10.1021/acs.accounts.1c00521.
38. Gholamalipour, Y., Karunanayake Mudiyanselage, A.,
and Martin, C. T. (2018) 3′ end additions by T7 RNA
polymerase are RNA self-templated, distributive and
diverse in character– RNA-Seq analyses, Nucleic Ac-
ids Res., 46, 9253-9263, https://doi.org/10.1093/nar/
gky796.
39. Milligan, J. F., Groebe, D. R., Witherell, G. W., and
Uhlenbeck, O. C. (1987) Oligoribonucleotide synthe-
sis using T7 RNA polymerase and synthetic DNA
templates, Nucleic Acids Res., 15, 8783-8798, https://
doi.org/10.1093/nar/15.21.8783.
40. Pelletier,J., and Sonenberg, N. (2019) The organizing
principles of eukaryotic ribosome recruitment, Annu.
Rev. Biochem., 88, 307-335, https://doi.org/10.1146/
annurev-biochem-013118-111042.
41. To, K. K. W., and Cho, W. C. S. (2021) An overview
of rational design of mRNA-based therapeutics and
vaccines, Expert Opin. Drug Discov., 16, 1307-1317,
https://doi.org/10.1080/17460441.2021.1935859.
42. Shanmugasundaram, M., Charles, I., and Kore, A. R.
(2016) Design, synthesis and biological evaluation
of dinucleotide mRNA cap analog containing prop-
argyl moiety, Bioorganic Med. Chem., 24, 1204-1208,
https://doi.org/10.1016/j.bmc.2016.01.048.
43. Kore, A. R., Shanmugasundaram, M., Charles, I.,
Vlassov, A. V., and Barta, T. J. (2009) Locked nucleic
acid (LNA)-Modified dinucleotide mRNA cap an-
alogue: synthesis, enzymatic incorporation, and
SELECTION OF UTRs 745
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
utilization, J.Am. Chem. Soc., 131, 6364-6365, https://
doi.org/10.1021/ja901655p.
44. Zeng, C., Hou, X., Yan, J., Zhang, C., Li, W., Zhao, W.,
Du,S., and Dong,Y. (2020) Leveraging mRNA sequenc-
es and nanoparticles to deliver SARS-CoV-2 antigens
in vivo, Adv. Mater., 32, e2004452, https://doi.org/
10.1002/adma.202004452.
45. 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, 1062-1068, https://doi.org/10.1016/
j.bbrc.2005.12.105.
46. Strenkowska, M., Kowalska, J., Lukaszewicz, M.,
Zuberek, J., Su, W., Rhoads, R. E., Darzynkiewicz, E.,
and Jemielity, J. (2010) Towards mRNA with superi-
or translational activity: synthesis and properties of
ARCA tetraphosphates with single phosphorothioate
modifications, New J. Chem., 34, 993-1007, https://
doi.org/10.1039/b9nj00644c.
47. Grudzien, E., Stepinski, J., Jankowska-Anyszka, M.,
Stolarski, R., Darzynkiewicz, E., and Rhoads, R. E.
(2004) Novel cap analogs for in vitro synthesis of
mRNAs with high translational efficiency, RNA, 10,
1479-1487, https://doi.org/10.1261/rna.7380904.
48. Grudzien-Nogalska, E., Jemielity, J., Kowalska, J.,
Darzynkiewicz,E., and Rhoads, R.E. (2007) Phospho-
rothioate cap analogs stabilize mRNA and increase
translational efficiency in mammalian cells, RNA, 13,
1745-1755, https://doi.org/10.1261/rna.701307.
49. Kuhn, A. N., Diken, M., Kreiter, S., Selmi, A.,
Kowalska,J., Jemielity,J., Darzynkiewicz,E., Huber,C.,
Türeci, Ö., and Sahin, U. (2010) Phosphorothioate
cap analogs increase stability and translational effi-
ciency of RNA vaccines in immature dendritic cells
and induce superior immune responses in vivo,
Gene Ther., 17, 961-971, https://doi.org/10.1038/
gt.2010.52.
50. Peng, J., Murray, E. L., and Schoenberg, D. R. (2008)
In vivo and in vitro analysis of poly(a) length effects
on mRNA translation, Methods Mol. Biol., 419, 215-
230, https://doi.org/10.1007/978-1-59745-033-1_15.
51. Chang, H., Lim, J., Ha, M., and Kim, V. N. (2014)
TAIL-seq: Genome-wide determination of poly(A) tail
length and 3’ end modifications, Mol. Cell, 53, 1044-
1052, https://doi.org/10.1016/j.molcel.2014.02.007.
52. Holtkamp, S., Kreiter, S., Selmi, A., Simon, P.,
Koslowski, M., Huber, C., Türeci, Ö., and Sahin, U.
(2006) Modification of antigen-encoding RNA increas-
es stability, translational efficacy, and T-cell stimula-
tory capacity of dendritic cells, Blood, 108, 4009-4017,
https://doi.org/10.1182/blood-2006-04-015024.
53. Karikó, K., Buckstein, M., Ni, H., and Weissman, D.
(2005) Suppression of RNA recognition by toll-like
receptors: the impact of nucleoside modification
and the evolutionary origin of RNA, Immunity, 23,
165-175, https://doi.org/10.1016/j.immuni.2005.06.008.
54. Karikó, K., Muramatsu, H., Welsh, F. A., Ludwig, J.,
Kato,H., Akira, S., and Weissman, D. (2008) Incorpo-
ration of pseudouridine into mRNA yields superior
nonimmunogenic vector with increased translational
capacity and biological stability, Mol. Ther., 16, 1833-
1840, https://doi.org/10.1038/mt.2008.200.
55. Kozak, M. (1986) Point mutations define a sequence
flanking the AUG initiator codon that modulates
translation by eukaryotic ribosomes, Cell, 44, 283-292,
https://doi.org/10.1016/0092-8674(86)90762-2.
56. Kozak, M. (1997) Recognition of AUG and alterna-
tive initiator codons is augmented by G in position
+4 but is not generally affected by the nucleotides in
positions +5 and +6, EMBO J., 16, 2482-2492, https://
doi.org/10.1093/emboj/16.9.2482.
57. Hernández,G., Osnaya, V. G., and Pérez-Martínez, X.
(2019) Conservation and variability of the AUG ini-
tiation codon context in eukaryotes, Trends Bio-
chem. Sci., 44, 1009-1021, https://doi.org/10.1016/
j.tibs.2019.07.001.
58. Hernández, G., García, A., Weingarten-Gabbay, S.,
Mishra, R. K., Hussain, T., Amiri, M., Moreno-
Hagelsieb, G., Montiel-Dávalos, A., Lasko, P., and
Sonenberg, N. (2024) Functional analysis of the AUG
initiator codon context reveals novel conserved
sequences that disfavor mRNA translation in eu-
karyotes, Nucleic Acids Res., 52, 1064-1079, https://
doi.org/10.1093/nar/gkad1152.
59. Gallie, D.R. (1991) The cap and poly(A) tail function
synergistically to regulate mRNA translational efficien-
cy, Genes Dev., 5, 2108-2116, https://doi.org/10.1101/
gad.5.11.2108.
60. Tanguay, R. L., and Gallie, D. R. (1996) Translational
Efficiency Is Regulated by the Length of the 3′ Un-
translated Region, Mol. Cell. Biol., 16, 146-156, https://
doi.org/10.1128/mcb.16.1.146.
61. Aditham,A., Shi,H., Guo,J., Zeng,H., Zhou,Y., Wade,
S. D., Huang, J., Liu, J., and Wang, X. (2022) Chemi-
cally modified mocRNAs for highly efficient protein
expression in mammalian cells, ACS Chem. Biol.,
17, 3352-3366, https://doi.org/10.1021/acschembio.
1c00569.
62. Chen,H., Liu,D., Guo,J., Aditham,A., Zhou,Y., Tian,J.,
Luo,S., Ren, J., Hsu,A., Huang, J., Kostas,F., Wu, M.,
Liu, D. R., and Wang, X. (2024) Branched chemical-
ly modified poly(A) tails enhance the translation ca-
pacity of mRNA, Nat. Biotechnol., 43, 194-203, https://
doi.org/10.1038/s41587-024-02174-7.
63. Asrani, K. H., Farelli, J. D., Stahley, M. R., Miller,
R. L., Cheng, C. J., Subramanian, R. R., and Brown,
J. M. (2018) Optimization of mRNA untranslated
regions for improved expression of therapeutic
mRNA, RNA Biol., 15, 756-762, https://doi.org/10.1080/
15476286.2018.1450054.
VOLKHIN et al.746
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
64. Pardi, N., Hogan, M. J., and Weissman, D. (2020) Re-
cent advances in mRNA vaccine technology, Curr.
Opin. Immunol., 65, 14-20, https://doi.org/10.1016/
j.coi.2020.01.008.
65. Schwanhüusser, B., Busse, D., Li, N., Dittmar, G.,
Schuchhardt, J., Wolf, J., Chen, W., and Selbach, M.
(2011) Global quantification of mammalian gene
expression control, Nature, 473, 337-342, https://
doi.org/10.1038/nature10098.
66. Mayr,C. (2017) Regulation by 3′-untranslated regions,
Annu. Rev. Genet., 51, 171-194, https://doi.org/10.1146/
annurev-genet-120116-024704.
67. Leppek, K., Byeon, G. W., Kladwang, W., Wayment-
Steele, H.K., Kerr, C. H., Xu, A.F., Kim, D.S., Topkar,
V. V., Choe, C., Rothschild, D., Tiu, G. C., Wellington-
Oguri, R., Fujii,K., Sharma, E., Watkins, A. M., Nicol,
J. J., Romano, J., Tunguz, B., Diaz, F., Cai, H., Guo, P.,
Wu, J., Meng, F., Shi, S., Participants, E., Dormitzer,
P.R., Solórzano,A., Barna,M., and Das,R. (2022) Com-
binatorial optimization of mRNA structure, stability,
and translation for RNA-based therapeutics, Nat.
Commun., 13, 1536, https://doi.org/10.1038/s41467-
022-28776-w.
68. Babendure, J. R., Babendure, J. L., Ding, J. H., and
Tsien, R. Y. (2006) Control of mammalian transla-
tion by mRNA structure near caps, RNA, 12, 851-861,
https://doi.org/10.1261/rna.2309906.
69. Kozak, M. (1989) Circumstances and mechanisms of
inhibition of translation by secondary structure in eu-
caryotic mRNAs, Mol. Cell. Biol., 9, 5134-5142, https://
doi.org/10.1128/mcb.9.11.5134-5142.1989.
70. Edgil, D., Polacek, C., and Harris, E. (2006) Dengue
virus utilizes a novel strategy for translation initi-
ation when cap-dependent translation is inhibited,
J. Virol., 80, 2976-2986, https://doi.org/10.1128/jvi.
80.6.2976-2986.2006.
71. García-Montalvo, B. M., Medina, F., and Del Angel,
R. M. (2004) La protein binds to NS5 and NS3 and
to the 5′ and 3′ ends of Dengue 4 virus RNA, Virus
Res., 102, 141-150, https://doi.org/10.1016/j.virus-
res.2004.01.024.
72. Wayment-Steele, H. K., Kim, D. S., Choe, C. A., Nicol,
J. J., Wellington-Oguri, R., Watkins, A. M., Sperberg,
R. A. P., Huang, P.-S., Participants, E., and Das, R.
(2020) Theoretical basis for stabilizing messenger
RNA through secondary structure design, bioRxiv,
https://doi.org/10.1101/2020.08.22.262931.
73. Leppek, K., Das, R., and Barna, M. (2018) Function-
al 5′ UTR mRNA structures in eukaryotic translation
regulation and how to find them, Nat. Rev. Mol.
Cell Biol., 19, 158-174, https://doi.org/10.1038/nrm.
2017.103.
74. Huang,X., and Du, Z. (2024) Possible involvement of
three-stemmed pseudoknots in regulating translation-
al initiation in human mRNAs, PLoS One, 19, e0307541,
https://doi.org/10.1371/journal.pone.0307541.
75. Singha Roy, A., Majumder, S., and Saha, P. (2023)
Stable RNA G-quadruplex in the 5′-UTR of human
cIAP1 mRNA promotes translation in an IRES-inde-
pendent manner, Biochemistry, 63, 475-486, https://
doi.org/10.1021/acs.biochem.3c00521.
76. Iacono, M., Mignone, F., and Pesole, G. (2005) uAUG
and uORFs in human and rodent 5′-untranslated
mRNAs, Gene, 349, 97-105, https://doi.org/10.1016/
j.gene.2004.11.041.
77. Wethmar, K. (2014) The regulatory potential of up-
stream open reading frames in eukaryotic gene ex-
pression, Wiley Interdiscip. Rev. RNA, 5, 765-778,
https://doi.org/10.1002/wrna.1245.
78. Dever, T.E., Ivanov, I.P., and Hinnebusch, A.G. (2023)
Translational regulation by uORFs and start codon
selection stringency, Genes Dev., 37, 474-489, https://
doi.org/10.1101/gad.350752.123.
79. Manske, F., Ogoniak, L., Jürgens, L., Grundmann, N.,
Makałowski, W., and Wethmar, K. (2023) The new
uORFdb: integrating literature, sequence, and varia-
tion data in a central hub for uORF research, Nucleic
Acids Res., 51, 1029-1042, https://doi.org/10.1093/nar/
gkac899.
80. Orr, M. W., Mao, Y., Storz, G., and Qian, S. B. (2021)
Alternative ORFs and small ORFs: Shedding light on
the dark proteome, Nucleic Acids Res., 48, 1029-1042,
https://doi.org/10.1093/NAR/GKZ734.
81. Hinnebusch, A. G., Ivanov, I. P., and Sonenberg, N.
(2016) Translational control by 5′-untranslated re-
gions of eukaryotic mRNAs, Science, 352, 1413-1416,
https://doi.org/10.1126/science.aad9868.
82. Araujo, P. R., Yoon, K., Ko, D., Smith, A. D., Qiao, M.,
Suresh, U., Burns, S. C., and Penalva, L. O. F. (2012)
Before it gets started: Regulating translation at the
5′UTR, Comp. Funct. Genomics, 2012, 475731, https://
doi.org/10.1155/2012/475731.
83. Bottorff, T. A., Park, H., Geballe, A. P., and
Subramaniam, A. R. (2022) Translational buffering
by ribosome stalling in upstream open reading
frames, PLoS Genet., 18, e1010460, https://doi.org/
10.1371/journal.pgen.1010460.
84. Matsui, M., Yachie, N., Okada, Y., Saito, R., and
Tomita,M. (2007) Bioinformatic analysis of post-tran-
scriptional regulation by uORF in human and mouse,
FEBS Lett., 581, 4184-4188, https://doi.org/10.1016/
j.febslet.2007.07.057.
85. Russell, P. J., Slivka, J. A., Boyle, E. P., Burghes,
A.H.M., and Kearse, M.G. (2023) Translation reiniti-
ation after uORFs does not fully protect mRNAs from
nonsense-mediated decay, RNA, 29, 735-744, https://
doi.org/10.1261/rna.079525.122.
86. Brogna, S., and Wen, J. (2009) Nonsense-mediated
mRNA decay (NMD) mechanisms, Nat. Struct. Mol.
Biol., 16, 107-113, https://doi.org/10.1038/nsmb.1550.
87. Oliveira, C.C., and McCarthy, J.E.G. (1995) The rela-
tionship between eukaryotic translation and mRNA
SELECTION OF UTRs 747
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
stability. A short upstream open reading frame
strongly inhibits translational initiation and greatly
accelerates mRNA degradation in the yeast Saccha-
romyces cerevisiae, J.Biol. Chem., 270, 8936-8943,
https://doi.org/10.1074/jbc.270.15.8936.
88. Ruiz-Echevarría, M. J., and Peltz, S. W. (2000) The
RNA binding protein Pub1 modulates the stabili-
ty of transcripts containing upstream open reading
frames, Cell, 101, 741-751, https://doi.org/10.1016/
S0092-8674(00)80886-7.
89. Kim, S. C., Sekhon, S. S., Shin, W. R., Ahn, G., Cho,
B. K., Ahn, J. Y., and Kim, Y. H. (2022) Modifications
of mRNA vaccine structural elements for improving
mRNA stability and translation efficiency, Mol. Cell.
Toxicol., 18, 1-8, https://doi.org/10.1007/s13273-021-
00171-4.
90. Carrieri, C., Cimatti, L., Biagioli, M., Beugnet, A.,
Zucchelli,S., Fedele,S., Pesce,E., Ferrer,I., Collavin,L.,
Santoro, C., Forrest, A. R. R., Carninci, P., Biffo, S.,
Stupka, E., and Gustincich, S. (2012) Long non-cod-
ing antisense RNA controls Uchl1 translation through
an embedded SINEB2 repeat, Nature, 491, 454-457,
https://doi.org/10.1038/nature11508.
91. Muckenthaler, M. U., Galy, B., and Hentze, M. W.
(2008) Systemic iron homeostasis and the iron-re-
sponsive element/iron-regulatory protein (IRE/
IRP) regulatory network, Annu. Rev. Nutr., 28, 197-
213, https://doi.org/10.1146/annurev.nutr.28.061807.
155521.
92. Medenbach, J., Seiler, M., and Hentze, M. W. (2011)
Translational control via protein-regulated upstream
open reading frames, Cell, 145, 902-913, https://
doi.org/10.1016/j.cell.2011.05.005.
93. Evdokimova, V., Tognon, C., Ng, T., Ruzanov, P.,
Melnyk, N., Fink, D., Sorokin, A., Ovchinnikov, L. P.,
Davicioni, E., Triche, T. J., and Sorensen, P. H. B.
(2009) Translational activation of Snail1 and other
developmentally regulated transcription factors by
YB-1 promotes an epithelial-mesenchymal transi-
tion, Cancer Cell, 15, 402-415, https://doi.org/10.1016/
j.ccr.2009.03.017.
94. Mayya, V. K., and Duchaine, T. F. (2019) Ciphers and
executioners: how 3 0 -untranslated regions deter-
mine the fate of messenger RNAs, Front. Genet., 10,
6, https://doi.org/10.3389/fgene.2019.00006.
95. Zekri, L., Kuzuoǧlu-Öztürk, D., and Izaurralde, E.
(2013) GW182 proteins cause PABP dissociation from
silenced miRNA targets in the absence of deadenyla-
tion, EMBO J., 32, 1052-1065, https://doi.org/10.1038/
emboj.2013.44.
96. Kamenska,A., Simpson,C., Vindry,C., Broomhead,H.,
Bénard, M., Ernoult-Lange, M., Lee, B. P., Harries,
L.W., Weil,D., and Standart,N. (2016) The DDX6-4E-T
interaction mediates translational repression and
P-body assembly, Nucleic Acids Res., 44, 6318-6334,
https://doi.org/10.1093/nar/gkw565.
97. Lai, W.S., Carballo,E., Thorn, J.M., Kennington, E.A.,
and Blackshear, P.J. (2000) Interactions of CCCH Zinc
finger proteins with mRNA, J.Biol. Chem., 275, 17827-
17837, https://doi.org/10.1074/jbc.m001696200.
98. Brennan, C. M., and Steitz, J. A. (2001) HuR and
mRNA stability, Cell. Mol. Life Sci., 58, 266-277, https://
doi.org/10.1007/PL00000854.
99. Gong, C., and Maquat, L. E. (2011) LncRNAs trans-
activate STAU1-mediated mRNA decay by duplexing
with 39 UTRs via Alu eleme, Nature, 470, 329-333,
https://doi.org/10.1038/nature09701.
100. O’Brien, J., Hayder, H., Zayed,Y., and Peng, C. (2018)
Overview of microRNA biogenesis, mechanisms of
actions, and circulation, Front. Endocrinol., 9, 402,
https://doi.org/10.3389/fendo.2018.00402.
101. Liegro, C. M. D., Bellafiore, M., Izquierdo, J. M.,
Rantanen,A., and Cuezva, J.M. (2000) 3′-Untranslated
regions of oxidative phosphorylation mRNAs function
in vivo as enhancers of translation, Biochem. J., 352,
109-115, https://doi.org/10.1042/bj3520109.
102. Popa, I., Harris, M. E., Donello, J. E., and Hope, T. J.
(2002) CRM1-dependent function of a cis-acting RNA
export element, Mol. Cell. Biol., 22, 2057-2067, https://
doi.org/10.1128/mcb.22.7.2057-2067.2002.
103. Loeb, J. E., Cordier, W. S., Harris, M. E., Weitzman,
M. D., and Hope, T. J. (1999) Enhanced expression
of transgenes from adeno-associated virus vectors
with the woodchuck hepatitis virus posttranscrip-
tional regulatory element: implications for gene
therapy, Hum. Gene Ther., 10, 2295-2305, https://
doi.org/10.1089/10430349950016942.
104. Lianoglou, S., Garg, V., Yang, J. L., Leslie, C. S., and
Mayr, C. (2013) Ubiquitously transcribed genes use
alternative polyadenylation to achieve tissue-spe-
cific expression, Genes Dev., 27, 2380-2396, https://
doi.org/10.1101/gad.229328.113.
105. Xie, J., Xie, Q., Zhang, H., Ameres, S. L., Hung, J. H.,
Su, Q., He, R., Mu, X., Seher Ahmed, S., Park, S.,
Kato, H., Li, C., Mueller, C., Mello, C. C., Weng, Z.,
Flotte, T. R., Zamore, P. D., and Gao, G. (2011)
MicroRNA- regulated, systemically delivered rAAV9:
A step closer to CNS-restricted transgene expression,
Mol. Ther., 19, 526-535, https://doi.org/10.1038/mt.
2010.279.
106. Qiao, C., Yuan, Z., Li, J., He, B., Zheng, H., Mayer, C.,
Li, J., and Xiao, X. (2011) Liver-specific microR-
NA-122 target sequences incorporated in AAV vec-
tors efficiently inhibits transgene expression in the
liver, Gene Ther., 18, 403-410, https://doi.org/10.1038/
gt.2010.157.
107. Karali, M., Manfredi, A., Puppo, A., Marrocco, E.,
Gargiulo, A., Allocca, M., de Corte, M., Rossi, S.,
Giunti, M., Bacci, M. L., Simonelli, F., Surace, E. M.,
Banfi,S., and Auricchio,A. (2011) MicroRNA-Restrict-
ed transgene expression in the retina, PLoS One, 6,
e22166, https://doi.org/10.1371/journal.pone.0022166.
VOLKHIN et al.748
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
108. Geisler,A., and Fechner,H. (2016) MicroRNA-regulated
viral vectors for gene therapy, World J. Exp. Med., 6,
37-54, https://doi.org/10.5493/wjem.v6.i2.37.
109. Ludwig, N., Leidinger, P., Becker, K., Backes, C., Fe-
hlmann, T., Pallasch, C., Rheinheimer, S., Meder, B.,
Stähler, C., Meese, E., and Keller, A. (2016) Distribu-
tion of miRNA expression across human tissues, Nu-
cleic Acids Res., 44, 3865-3877, https://doi.org/10.1093/
nar/gkw116.
110. Kelly, E. J., Nace, R., Barber, G. N., and Russell, S. J.
(2010) Attenuation of vesicular stomatitis virus en-
cephalitis through microRNA targeting, J. Virol., 84,
1550-1562, https://doi.org/10.1128/jvi.01788-09.
111. Barnes,D., Kunitomi,M., Vignuzzi,M., Saksela,K., and
Andino,R. (2008) Harnessing endogenous miRNAs to
control virus tissue tropism as a strategy for devel-
oping attenuated virus vaccines, Cell Host Microbe,
4, 239-248, https://doi.org/10.1016/j.chom.2008.08.003.
112. Yen, L.C., Lin, Y.L., Sung, H.H., Liao, J.T., Tsao, C.H.,
Su, C. M., Lin, C. K., and Liao, C. L. (2013) Neurovir-
ulent flavivirus can be attenuated in mice by incor-
poration of neuron-specific microRNA recognition
elements into viral genome, Vaccine, 31, 5915-5922,
https://doi.org/10.1016/j.vaccine.2011.09.102.
113. Turner, J. D., and Muller, C.P. (2005) Structure of the
glucocorticoid receptor (NR3C1) gene 5′ untranslat-
ed region: identification, and tissue distribution of
multiple new human exon 1, J. Mol. Endocrinol., 35,
283-292, https://doi.org/10.1677/jme.1.01822.
114. Hirahara, Y., Tsuda, M., Wada, Y., and Honke, K.
(2000) cDNA cloning, genomic cloning, and tis-
sue-specific regulation of mouse cerebroside sulfo-
transferase, Eur. J. Biochem., 267, 1909-1917, https://
doi.org/10.1046/j.1432-1327.2000.01139.x.
115. Wortman, M. J., Hanson, L. K., Martínez-Sobrido, L.,
Campbell, A. E., Nance, J. A., García-Sastre, A., and
Johnson, E. M. (2010) Regulation of PURA gene tran-
scription by three promoters generating distinctly
spliced 5-prime leaders: a novel means of fine con-
trol over tissue specificity and viral signals, BMC Mol.
Biol., 11, 81, https://doi.org/10.1186/1471-2199-11-81.
116. Roberts, S. J., Chung, K.-N., Nachmanoff, K., and
Elwood, P. C. (1997) Tissue-specific promoters of the
α human folate receptor gene yield transcripts with
divergent 5′ leader sequences and different transla-
tional efficiencies, Biochem. J., 326, 439-447, https://
doi.org/10.1042/bj3260439.
117. Xiao, Y., Muhuri, M., Li, S., Qin, W., Xu, G., Luo, L.,
Li,J., Letizia, A.J., Wang, S. K., Chan, Y. K., Wang, C.,
Fuchs, S.P., Wang,D., Su,Q., Abu Nahid,M., Church,
G. M., Farzan, M., Yang, L., Wei, Y., Desrosiers, R. C.,
Mueller,C., Tai, P.W.L., and Gao,G. (2019) Circumvent-
ing cellular immunity by miR142-mediated regulation
sufficiently supports rAAV-delivered OVA expression
without activating humoral immunity, JCI Insight,
4, e99052, https://doi.org/10.1172/jci.insight.99052.
118. Majowicz, A., Maczuga, P., Kwikkers, K. L., van der
Marel,S., van Logtenstein,R., Petry,H., van Deventer,
S.J., Konstantinova,P., and Ferreira,V. (2013) Mir-142-
3p target sequences reduce transgene-directed immu-
nogenicity following intramuscular adeno-associated
virus 1 vector-mediated gene delivery, J. Gene Med.,
15, 219-232, https://doi.org/10.1002/jgm.2712.
119. Muhuri, M., Zhan, W., Maeda, Y., Li, J., Lotun, A.,
Chen, J., Sylvia, K., Dasgupta, I., Arjomandnejad, M.,
Nixon,T., Keeler, A.M., Manokaran,S., He,R., Su,Q.,
Tai, P. W. L., and Gao, G. (2021) Novel combinatori-
al microRNA-binding sites in AAV vectors synergis-
tically diminish antigen presentation and transgene
immunity for efficient and stable transduction,
Front. Immunol., 12, 674242, https://doi.org/10.3389/
fimmu.2021.674242.
120. Khoroshkin,M., Zinkevich,A., Aristova,E., Yousefi,H.,
and Sean, B. (2024) A generative framework for en-
hanced cell-type specificity in rationally designed
mRNAs, bioRxiv, https://doi.org/10.1101/2024.12.
31.630783.
121. Flamand, M.N., Tegowski,M., and Meyer, K.D. (2023)
The proteins of mRNA modification: writers, readers,
and erasers, Annu. Rev. Biochem., 92, 145-173, https://
doi.org/10.1146/annurev-biochem-052521-035330.
122. Khan, F. A., Nsengimana, B., Awan, U. A., Ji, X. Y.,
Ji,S., and Dong,J. (2024) Regulatory roles of N
6
-meth-
yladenosine (m
6
A) methylation in RNA process-
ing and non-communicable diseases, Cancer Gene
Ther., 31, 1439-1453, https://doi.org/10.1038/S41417-
024-00789-1.
123. Yu,X., Zhao,H., Wang,R., Chen,Y., Ouyang,X., Li,W.,
Sun,Y., and Peng,A. (2024) Cancer epigenetics: from
laboratory studies and clinical trials to precision med-
icine, Cell Death Discov., 10, 28, https://doi.org/10.1038/
s41420-024-01803-z.
124. Wilinski,D., and Dus,M. (2023) N 6-adenosine meth-
ylation controls the translation of insulin mRNA,
Nat. Struct. Mol. Biol., 30, 1260-1264, https://doi.org/
10.1038/s41594-023-01048-x.
125. Morais, P., Adachi, H., and Yu, Y.-T. (2021) The criti-
cal contribution of pseudouridine to mRNA COVID-19
vaccines, Front. Cell Dev. Biol., 9, 789427, https://
doi.org/10.3389/fcell.2021.789427.
126. Song,B., Tang,Y., Wei,Z., Liu,G., Su,J., Meng,J., and
Chen, K. (2020) PIANO: a web server for pseudou-
ridine-Site (Ψ) identification and functional anno-
tation, Front. Genet., 11, 88, https://doi.org/10.3389/
fgene.2020.00088.
127. Kierzek,E., Malgowska,M., Lisowiec,J., Turner, D.H.,
Gdaniec, Z., and Kierzek, R. (2014) The contribution
of pseudouridine to stabilities and structure of RNAs,
Nucleic Acids Res., 42, 3492-3501, https://doi.org/
10.1093/nar/gkt1330.
128. Karikó, K., Muramatsu, H., Keller, J. M., and Weiss-
man, D. (2012) Increased erythropoiesis in mice
SELECTION OF UTRs 749
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
injected with submicrogram quantities of pseudou-
ridine-containing mRNA encoding erythropoie-
tin, Mol. Ther., 20, 948-953, https://doi.org/10.1038/
mt.2012.7.
129. Chen, S., Liu, Z., Cai, J., Li, H., and Qiu, M. (2024)
N1-methylpseudouridine modification level cor-
relates with protein expression, immunogenicity,
and stability of mRNA, MedComm, 5, e691, https://
doi.org/10.1002/MCO2.691.
130. Sample, P. J., Wang, B., Reid, D. W., Presnyak, V.,
McFadyen, I. J., Morris, D. R., and Seelig, G. (2019)
Human 5′ UTR design and variant effect prediction
from a massively parallel translation assay, Nat. Bio-
technol., 37, 803-809, https://doi.org/10.1038/s41587-
019-0164-5.
131. Chang,Y., Jin,H., Cui,Y., Yang,F., Chen,K., Kuang,W.,
Huo,C., Xu,Z., Li,Y., Lin,A., Yang,B., Liu,W., Xie,S.,
and Zhou, T. (2024) PUS7-dependent pseudouridyla-
tion of ALKBH3 mRNA inhibits gastric cancer pro-
gression, Clin. Transl. Med., 14, e1811, https://doi.org/
10.1002/CTM2.1811.
132. Monroe,J., Eyler, D.E., Mitchell,L., Deb,I., Bojanows-
ki,A., Srinivas,P., Dunham, C.M., Roy,B., Frank, A.T.,
and Koutmou, K. S. (2024) N1-Methylpseudouridine
and pseudouridine modifications modulate mRNA
decoding during translation, Nat. Commun., 15, 8119,
https://doi.org/10.1038/s41467-024-51301-0.
133. Moradian, H., Roch, T., Anthofer, L., Lendlein, A.,
and Gossen, M. (2022) Chemical modification of uri-
dine modulates mRNA-mediated proinflammatory
and antiviral response in primary human macro-
phages, Mol. Ther. Nucleic Acids, 27, 854-869, https://
doi.org/10.1016/j.omtn.2022.01.004.
134. Li, X., Xiong, X., Zhang, M., Wang, K., Chen, Y.,
Zhou, J., Mao, Y., Lv, J., Yi, D., Chen, X.W., Wang, C.,
Qian, S. B., and Yi, C. (2017) Base-resolution map-
ping reveals distinct m1A methylome in nucle-
ar- and mitochondrial-encoded transcripts, Mol.
Cell, 68, 993-1005.e9, https://doi.org/10.1016/j.molcel.
2017.10.019.
135. Zhou, H., Kimsey, I. J., Nikolova, E. N., Sathyamoor-
thy, B., Grazioli, G., McSally, J., Bai, T., Wunderlich,
C.H., Kreutz,C., Andricioaei,I., and Al-Hashimi, H.M.
(2016) M1A and m1G disrupt A-RNA structure through
the intrinsic instability of Hoogsteen base pairs,
Nat. Struct. Mol. Biol., 23, 803-810, https://doi.org/
10.1038/nsmb.3270.
136. Qi, Z., Zhang, C., Jian, H., Hou, M., Lou, Y., Kang, Y.,
Wang, W., Lv, Y., Shang, S., Wang, C., Li, X., Feng,S.,
and Zhou,H. (2023) N1-Methyladenosine modification
of mRNA regulates neuronal gene expression and ox-
ygen glucose deprivation/reoxygenation induction,
Cell Death Discov., 9, 159, https://doi.org/10.1038/
s41420-023-01458-2.
137. Zhang, C., Yi, X., Hou, M., Li, Q., Li, X., Lu, L., Qi, E.,
Wu,M., Qi, L., Jian,H., Qi, Z., Lv, Y., Kong,X., Bi, M.,
Feng, S., and Zhou, H. (2023) The landscape of m1A
modification and its posttranscriptional regulato-
ry functions in primary neurons, Elife, 12, e85324,
https://doi.org/10.7554/elife.85324.
138. Boo, S.H., Ha,H., and Kim, Y.K. (2022) m1A and m
6
A
modifications function cooperatively to facilitate rap-
id mRNA degradation, Cell Rep., 40, 111317, https://
doi.org/10.1016/j.celrep.2022.111317.
139. Park, O.H., Ha,H., Lee,Y., Boo, S.H., Kwon, D.H., Song,
H.K., and Kim, Y.K. (2019) Endoribonucleolytic cleav-
age of m
6
A-containing RNAs by RNase P/MRP com-
plex, Mol. Cell, 74, 494-507.e8, https://doi.org/10.1016/
j.molcel.2019.02.034.
140. Lee,Y., Choe,J., Park, O.H., and Kim, Y.K. (2020) Mo-
lecular mechanisms driving mRNA degradation by
m
6
A modification, Trends Genet., 36, 177-188, https://
doi.org/10.1016/j.tig.2019.12.007.
141. Liu, N., Dai, Q., Zheng, G., He, C., Parisien, M., and
Pan, T. (2015) N6-methyladenosine-dependent RNA
structural switches regulate RNA-protein interac-
tions, Nature, 518, 560-564, https://doi.org/10.1038/
nature14234.
142. Guca, E., Alarcon, R., Palo, M. Z., Santos, L., Alonso-
Gil,S., Davyt,M., de Lima, L.H.F., Boissier,F., Das,S.,
Zagrovic,B., Puglisi, J.D., Hashem,Y., and Ignatova,Z.
(2024) N6-methyladenosine in 5′ UTR does not pro-
mote translation initiation, Mol. Cell, 84, 584-595.e6,
https://doi.org/10.1016/j.molcel.2023.12.028.
143. Meyer, K. D. (2019) m
6
A-mediated translation reg-
ulation, Biochim. Biophys. Acta Gene Regul. Mech.,
1862, 301-309, https://doi.org/10.1016/j.bbagrm.
2018.10.006.
144. Zhang, S., Liu, Y., Ma, X., Gao, X., Ru, Y., Hu, X., and
Gu, X. (2024) Recent advances in the potential role
of RNA N4-acetylcytidine in cancer progression,
Cell Commun. Signal., 22, 49, https://doi.org/10.1186/
s12964-023-01417-5.
145. Dominissini, D., and Rechavi, G. (2018) N4-acetyl-
ation of cytidine in mRNA by NAT10 regulates sta-
bility and translation, Cell, 175, 1725-1727, https://
doi.org/10.1016/j.cell.2018.11.037.
146. Liu, Y., Wang, X., Liu, Y., Yang, J., Mao, W., Feng, C.,
Wu,X., Chen,X., Chen,L., and Dong,P. (2023) N4-ace-
tylcytidine-dependent GLMP mRNA stabilization
by NAT10 promotes head and neck squamous cell
carcinoma metastasis and remodels tumor micro-
environment through MAPK/ERK signaling path-
way, Cell Death Dis., 14, 712, https://doi.org/10.1038/
s41419-023-06245-6.
147. Arango, D., Sturgill, D., Alhusaini, N., Dillman, A. A.,
Sweet, T.J., Hanson,G., Hosogane,M., Sinclair, W.R.,
Nanan, K. K., Mandler, M. D., Fox, S. D., Zengeya,
T.T., Andresson,T., Meier, J.L., Coller,J., and Oberdo-
erffer,S. (2018) Acetylation of cytidine in mRNA pro-
motes translation efficiency, Cell, 175, 1872-1886.e24,
https://doi.org/10.1016/j.cell.2018.10.030.
VOLKHIN et al.750
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
148. Arango, D., Sturgill, D., Yang, R., Kanai, T., Bauer, P.,
Roy, J., Wang, Z., Hosogane, M., Schiffers, S., and
Oberdoerffer, S. (2022) Erratum: direct epitranscrip-
tomic regulation of mammalian translation initiation
through N4-acetylcytidine, Mol. Cell, 82, 2797-2814.
e11, https://doi.org/10.1016/j.molcel.2022.06.022.
149. Schumann, U., Zhang, H. N., Sibbritt, T., Pan, A.,
Horvath, A., Gross, S., Clark, S. J., Yang, L., and Pre-
iss,T. (2020) Multiple links between 5-methylcytosine
content of mRNA and translation, BMC Biol., 18, 40,
https://doi.org/10.1186/s12915-020-00769-5.
150. Malbec,L., Zhang,T., Chen, Y.S., Zhang,Y., Sun, B.F.,
Shi, B.Y., Zhao, Y. L., Yang, Y., and Yang, Y. G. (2019)
Dynamic methylome of internal mRNA N
7
-methyl-
guanosine and its regulatory role in translation,
Cell Res., 29, 927-941, https://doi.org/10.1038/s41422-
019-0230-z.
151. Dong, R., Wang, C., Tang, B., Cheng, Y., Peng, X.,
Yang, X., Ni, B., and Li, J. (2024) WDR4 promotes
HCC pathogenesis through N7-methylguanosine by
regulating and interacting with METTL1, Cell. Sig-
nal., 118, 111145, https://doi.org/10.1016/j.cellsig.
2024.111145.
152. Schlake, T., Thess, A., Fotin-Mleczek, M., and Kallen,
K.-J. (2012) Developing mRNA-vaccine technolo-
gies, RNA Biol., 9, 1319-1330, https://doi.org/10.4161/
rna.22269.
153. Carralot, J. P., Weide, B., Schoor, O., Probst, J.,
Scheel,B., Teufel,R., Hoerr,I., Garbe,C., Rammensee,
H. G., and Pascolo, S. (2005) Production and charac-
terization of amplified tumor-derived cRNA libraries
to be used as vaccines against metastatic melanomas,
Genet. Vaccines Ther., 3, 6, https://doi.org/10.1186/
1479-0556-3-6.
154. Warren, L., Manos, P. D., Ahfeldt, T., Loh, Y. H.,
Li, H., Lau, F., Ebina, W., Mandal, P. K., Smith, Z. D.,
Meissner, A., Daley, G. Q., Brack, A. S., Collins, J. J.,
Cowan, C., Schlaeger, T. M., and Rossi, D. J. (2010)
Highly efficient reprogramming to pluripotency and
directed differentiation of human cells with synthet-
ic modified mRNA, Cell Stem Cell, 7, 618-630, https://
doi.org/10.1016/j.stem.2010.08.012.
155. Weng, Y., Li, C., Yang, T., Hu, B., Zhang, M., Guo, S.,
Xiao, H., Liang, X.-J., and Huang, Y. (2020) The chal-
lenge and prospect of mRNA therapeutics land-
scape, Biotechnol. Adv., 40, 107534, https://doi.org/
10.1016/j.biotechadv.2020.107534.
156. Malone, R. W., Felgner, P. L., and Verma, I. M. (1989)
Cationic liposome-mediated RNA transfection., Proc.
Natl. Acad. Sci. USA, 86, 6077-6081, https://doi.org/
10.1073/pnas.86.16.6077.
157. Karikó,K., Kuo,A., and Barnathan, E.S. (1999) Over-
expression of urokinase receptor in mammalian cells
following administration of the in vitro transcribed
encoding mRNA, Gene Ther., 6, 1092-1100, https://
doi.org/10.1038/sj.gt.3300930.
158. Kreiter, S., Selmi, A., Diken, M., Sebastian, M.,
Osterloh, P., Schild, H., Huber, C., Türeci, Ö., and
Sahin, U. (2008) Increased antigen presentation ef-
ficiency by coupling antigens to MHC class I traf-
ficking signals, J. Immunol., 180, 309-318, https://
doi.org/10.4049/jimmunol.180.1.309.
159. Kozak, M. (1994) Features in the 5′ non-coding se-
quences of rabbit α and β-globin mRNAs that affect
translational efficiency, J. Mol. Biol., 235, 95-110,
https://doi.org/10.1016/S0022-2836(05)80019-1.
160. Annweiler, A., Hipskind, R. A., and Wirth, T. (1991)
A strategy for efficient in vitro translation of cDNAs
using the rabbit β-globin leader sequence, Nucleic
Acids Res., 19, 3750, https://doi.org/10.1093/nar/
19.13.3750.
161. Conry, R.M., LoBuglio, A. F., Wright, M., Sumerel,L.,
Pike, M. J., Johanning, F., Benjamin, R., Lu, D., and
Curiel, D. T. (1995) Characterization of a messenger
RNA polynucleotide vaccine vector., Cancer Res., 55,
1397-1400.
162. Rohner,E., Yang,R., Foo, K.S., Goedel,A., and Chien,
K. R. (2022) Unlocking the promise of mRNA ther-
apeutics, Nat. Biotechnol., 40, 1586-1600, https://
doi.org/10.1038/s41587-022-01491-z.
163. Reshetnikov,V., Terenin,I., Shepelkova,G., Yeremeev,V.,
Kolmykov,S., Nagornykh,M., Kolosova,E., Sokolova,T.,
Zaborova, O., Kukushkin,I., Kazakova, A., Kunyk,D.,
Kirshina, A., Vasileva, O., Seregina, K., Pateev, I.,
Kolpakov, F., and Ivanov, R. (2024) Untranslated re-
gion sequences and the efficacy of mRNA vaccines
against tuberculosis, Int. J. Mol. Sci., 25, 888, https://
doi.org/10.3390/ijms25020888.
164. Yu, J., and Russell, J. E. (2001) Structural and func-
tional analysis of an mRNP complex that mediates
the high stability of human β-globin mRNA, Mol. Cell.
Biol., 21, 5879-5888, https://doi.org/10.1128/mcb.21.17.
5879-5888.2001.
165. Wang, Z., Day, N., Trifillis, P., and Kiledjian, M.
(1999) An mRNA stability complex functions with
poly(A)-binding protein to stabilize mRNA in vitro,
Mol. Cell. Biol., 19, 4552-4560, https://doi.org/10.1128/
mcb.19.7.4552.
166. Aviv,H., Voloch,Z., Bastos,R., and Levy,S. (1976) Bio-
synthesis and stability of globin mRNA in cultured
erythroleukemic friend cells, Cell, 8, 495-503, https://
doi.org/10.1016/0092-8674(76)90217-8.
167. Waggoner, S.A., and Liebhaber, S.A. (2003) Regulation
of α-globin mRNA stability, Exp. Biol. Med., 228, 387-
395, https://doi.org/10.1177/153537020322800409.
168. Holcik, M., and Liebhaber, S. A. (1997) Four highly
stable eukaryotic mRNAs assemble 3′ untranslated
region RNA-protein complexes sharing cis and trans
components, Proc. Natl. Acad. Sci. USA, 94, 2410-2414,
https://doi.org/10.1073/pnas.94.6.2410.
169. Wollner, C.J., Richner,M., Hassert, M.A., Pinto, A.K.,
Brien, J.D., and Richner, J.M. (2021) A Dengue Virus
SELECTION OF UTRs 751
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Serotype 1 mRNA-LNP Vaccine Elicits Protective
Immune Responses, J. Virol., 95, e02482-20, https://
doi.org/10.1128/jvi.02482-20.
170. Sahin, U., Derhovanessian, E., Miller, M., Kloke,
B. P., Simon, P., Löwer, M., Bukur, V., Tadmor,
A. D., Luxemburger, U., Schrörs, B., Omokoko, T.,
Vormehr, M., Albrecht, C., Paruzynski, A., Kuhn,
A. N., Buck, J., Heesch, S., Schreeb, K. H., Müller, F.,
Ortseifer,I., Vogler,I., Godehardt,E., Attig,S., Rae,R.,
Breitkreuz, A., Tolliver, C., Suchan, M., Martic, G.,
Hohberger, A., Sorn, P., Diekmann, J., Ciesla, J.,
Waksmann, O., Brück, A. K., Witt, M., Zillgen, M.,
Rothermel, A., Kasemann, B., Langer, D., Bolte, S.,
Diken, M., Kreiter, S., Nemecek, R., Gebhardt, C.,
Grabbe,S., Höller, C., Utikal, J., Huber, C., Loquai, C.,
and Türeci, Ö. (2017) Personalized RNA mutanome
vaccines mobilize poly-specific therapeutic immu-
nity against cancer, Nature, 547, 222-226, https://
doi.org/10.1038/nature23003.
171. Ding, X., Zhou, Y., He, J., Zhao, J., and Li, J. (2024)
Enhancement of SARS-CoV-2 mRNA vaccine efficacy
through the application of TMSB10 UTR for superior
antigen presentation and immune activation, Vaccines,
12, 432, https://doi.org/10.3390/vaccines12040432.
172. Orlandini von Niessen, A. G., Poleganov, M. A.,
Rechner, C., Plaschke, A., Kranz, L. M., Fesser, S.,
Diken, M., Löwer, M., Vallazza, B., Beissert, T.,
Bukur,V., Kuhn, A.N., Türeci,Ö., and Sahin,U. (2019)
Improving mRNA-based therapeutic gene delivery by
expression-augmenting 3′ UTRs identified by cellu-
lar library screening, Mol. Ther., 27, 824-836, https://
doi.org/10.1016/j.ymthe.2018.12.011.
173. Ferizi, M., Aneja, M. K., Balmayor, E. R., Badieyan,
Z.S., Mykhaylyk,O., Rudolph,C., and Plank,C. (2016)
Human cellular CYBA UTR sequences increase mRNA
translation without affecting the half-life of recom-
binant RNA transcripts, Sci. Rep., 6, 39149, https://
doi.org/10.1038/srep39149.
174. Chiu, W.-W., Kinney, R. M., and Dreher, T. W. (2005)
Control of translation by the 5′- and 3′-terminal re-
gions of the dengue virus genome, J.Virol., 79, 8303-
8315, https://doi.org/10.1128/jvi.79.13.8303-8315.2005.
175. Gallie, D. R., Tanguay, R. L., and Leathers, V. (1995)
The tobacco etch viral 5′ leader and poly(A) tail
are functionally synergistic regulators of trans-
lation, Gene, 165, 233-238, https://doi.org/10.1016/
0378-1119(95)00521-7.
176. Tan, X., and Wan, Y. (2008) Enhanced protein ex-
pression by internal ribosomal entry site-driven
mRNA translation as a novel approach for in vitro
loading of dendritic cells with antigens, Hum. Im-
munol., 69, 32-40, https://doi.org/10.1016/j.humimm.
2007.11.009.
177. Yakubov,E., Rechavi,G., Rozenblatt,S., and Givol, D.
(2010) Reprogramming of human fibroblasts to plu-
ripotent stem cells using mRNA of four transcription
factors, Biochem. Biophys. Res. Commun., 394, 189-
193, https://doi.org/10.1016/j.bbrc.2010.02.150.
178. Mamaghani, S., Penna, R. R., Frei, J., Wyss, C.,
Mellett, M., Look, T., Weiss, T., Guenova, E., Kündig,
T. M., Lauchli, S., and Pascolo, S. (2024) Synthetic
mRNAs containing minimalistic untranslated regions
are highly functional in vitro and in vivo, Cells, 13,
1242, https://doi.org/10.3390/cells13151242.
179. Ma,Q., Zhang,X., Yang,J., Li,H., Hao,Y., and Feng,X.
(2024) Optimization of the 5′ untranslated region
of mRNA vaccines, Sci. Rep., 14, 19845, https://
doi.org/10.1038/s41598-024-70792-x.
180. Schrom, E., Huber, M., Aneja, M., Dohmen, C.,
Emrich, D., Geiger, J., Hasenpusch, G., Herrmann-
Janson, A., Kretzschmann, V., Mykhailyk, O.,
Pasewald, T., Oak, P., Hilgendorff, A., Wohlleber, D.,
Hoymann, H.G., Schaudien,D., Plank,C., Rudolph, C.,
and Kubisch-Dohmen,R. (2017) Translation of angio-
tensin-converting enzyme 2 upon liver- and lung-tar-
geted delivery of optimized chemically modified
mRNA, Mol. Ther. Nucleic Acids, 7, 350-365, https://
doi.org/10.1016/j.omtn.2017.04.006.
181. Vivinus,S., Baulande,S., Van Zanten,M., Campbell,F.,
Topley, P., Ellis, J. H., Dessen, P., and Coste, H.
(2001) An element within the 5′ untranslated re-
gion of human Hsp70 mRNA which acts as a gener-
al enhancer of mRNA translation, Eur. J. Biochem.,
268, 1908-1917, https://doi.org/10.1046/j.1432-1327.
2001.02064.x.
182. Zhao,Y., Moon,E., Carpenito,C., Paulos, C.M., Liu,X.,
Brennan, A. L., Chew, A., Carroll, R. G., Scholler, J.,
Levine, B. L., Albelda, S. M., and June, C. H. (2010)
Multiple injections of electroporated autologous T
cells expressing a chimeric antigen receptor medi-
ate regression of human disseminated tumor, Cancer
Res., 70, 9053-9061, https://doi.org/10.1158/0008-5472.
CAN-10-2880.
183. Zinckgraf, J. W., and Silbart, L. K. (2003) Modulat-
ing gene expression using DNA vaccines with dif-
ferent 3′-UTRs influences antibody titer, seroconver-
sion and cytokine profiles, Vaccine, 21, 1640-1649,
https://doi.org/10.1016/S0264-410X(02)00740-5.
184. Holden, K. L., and Harris, E. (2004) Enhancement
of dengue virus translation: Role of the 3′ untrans-
lated region and the terminal 3′ stem-loop domain,
Virology, 329, 119-133, https://doi.org/10.1016/j.virol.
2004.08.004.
185. Linares-Fernández, S., Moreno, J., Lambert, E.,
Mercier- Gouy, P., Vachez, L., Verrier, B., and Exposito,
J. Y. (2021) Combining an optimized mRNA template
with a double purification process allows strong ex-
pression of in vitro transcribed mRNA, Mol. Ther.
Nucleic Acids, 26, 945-956, https://doi.org/10.1016/
j.omtn.2021.10.007.
186. Trepotec, Z., Aneja, M. K., Geiger, J., Hasenpusch, G.,
Plank, C., and Rudolph, C. (2019) Maximizing the
VOLKHIN et al.752
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
translational yield of mRNA therapeutics by mini-
mizing 5′-UTRs, Tissue Eng. Part A, 25, 69-79, https://
doi.org/10.1089/ten.tea.2017.0485.
187. Jia, L., Mao, Y., Ji, Q., Dersh, D., Yewdell, J. W., and
Qian, S.B. (2020) Decoding mRNA translatability and
stability from the 5′ UTR, Nat. Struct. Mol. Biol., 27,
814-821, https://doi.org/10.1038/s41594-020-0465-x.
188. Mandal, P. K., and Rossi, D.J. (2013) Reprogramming
human fibroblasts to pluripotency using modified
mRNA, Nat. Protoc., 8, 568-582, https://doi.org/10.1038/
nprot.2013.019.
189. Lee, J., Dykstra, B., Spencer, J. A., Kenney, L. L.,
Greiner, D. L., Shultz, L. D., Brehm, M. A., Lin, C. P.,
Sackstein, R., and Rossi, D. J. (2017) MRNA-mediated
glycoengineering ameliorates deficient homing of
human stem cell-derived hematopoietic progenitors,
J.Clin. Invest., 127, 2433-2437, https://doi.org/10.1172/
JCI92030.
190. Richner, J. M., Himansu,S., Dowd, K. A., Butler, S. L.,
Salazar, V., Fox, J. M., Julander, J. G., Tang, W. W.,
Shresta,S., Pierson, T.C., Ciaramella,G., and Diamond,
M. S. (2017) Modified mRNA vaccines protect against
Zika virus infection, Cell, 168, 1114-1125.e10,
https://doi.org/10.1016/j.cell.2017.02.017.
191. Xia, X. (2021) Detailed dissection and critical eval-
uation of the Pfizer/BioNTech and Moderna mRNA
vaccines, Vaccines, 9, 734, https://doi.org/10.3390/
vaccines9070734.
192. Ringnér, M., and Krogh, M. (2005) Folding free en-
ergies of 5′-UTRs impact post-transcriptional regu-
lation on a genomic scale in yeast, PLoS Comput.
Biol., 1, e72, https://doi.org/10.1371/journal.pcbi.
0010072.
193. Pelletier, J., and Sonenberg, N. (1985) Insertion mu-
tagenesis to increase secondary structure within the
5′ noncoding region of a eukaryotic mRNA reduc-
es translational efficiency, Cell, 40, 515-526, https://
doi.org/10.1016/0092-8674(85)90200-4.
194. Karollus,A., Avsec,Ž., and Gagneur,J. (2021) Predict-
ing mean ribosome load for 5’UTR of any length us-
ing deep learning, PLoS Comput. Biol., 17, e1008982,
https://doi.org/10.1371/journal.pcbi.1008982.
195. Linder, J., and Seelig, G. (2021) Fast activation max-
imization for molecular sequence design, BMC Bio-
informatics, 22, 510, https://doi.org/10.1186/s12859-
021-04437-5.
196. Barazandeh, S., Ozden, F., Hincer, A., Seker, U. O. S.,
and Cicek, A. E. (2023) UTRGAN: learning to gen-
erate 5’ UTR sequences for optimized translation
efficiency and gene expression, bioRxiv, https://
doi.org/10.1101/2023.01.30.526198.
197. Linder, J., Bogard,N., Rosenberg, A.B., and Seelig,G.
(2020) A generative neural network for maximiz-
ing fitness and diversity of synthetic DNA and pro-
tein sequences, Cell Syst., 11, 49-62.e16, https://
doi.org/10.1016/j.cels.2020.05.007.
198. Li, G., Wu, J., and Wang, X. (2024) Predicting func-
tional UTR variants by integrating region-specific fea-
tures, Brief. Bioinform., 25, bbae248, https://doi.org/
10.1093/bib/bbae248.
199. Zhang,H., Gao,X., Zhang,J., and Lai,L. (2025) mRNA-
2vec: mRNA embedding with language model in the
5’UTR-CDS for mRNA design, Proc. Thirty-Ninth AAAI
Conf. Artif. Intell., 1, 1057-1065, https://doi.org/10.1609/
aaai.v39i1.32092.
200. Brar, G.A., and Weissman, J.S. (2015) Ribosome pro-
filing reveals the what, when, where and how of pro-
tein synthesis, Nat. Rev. Mol. Cell Biol., 16, 651-664,
https://doi.org/10.1038/nrm4069.
201. Chassé, H., Boulben,S., Costache, V., Cormier, P., and
Morales, J. (2017) Analysis of translation using poly-
some profiling, Nucleic Acids Res., 45, e15, https://
doi.org/10.1093/nar/gkw907.
202. Zheng, Y., and VanDusen, N. J. (2023) Massively
parallel reporter assays for high-throughput
in vivo analysis of cis-regulatory elements, J. Car-
diovasc. Dev. Dis., 10, 144, https://doi.org/10.3390/
jcdd10040144.
203. Gallego Romero, I., and Lea, A. J. (2023) Leveraging
massively parallel reporter assays for evolutionary
questions, Genome Biol., 24, 26, https://doi.org/10.1186/
s13059-023-02856-6.
204. Griesemer, D., Xue, J. R., Reilly, S. K., Ulirsch, J. C.,
Kukreja,K., Davis, J.R., Kanai, M., Yang, D.K., Butts,
J. C., Guney, M. H., Luban, J., Montgomery, S. B.,
Finucane, H.K., Novina, C.D., Tewhey,R., and Sabeti,
P. C. (2021) Genome-wide functional screen of 3′UTR
variants uncovers causal variants for human dis-
ease and evolution, Cell, 184, 5247-5260.e19, https://
doi.org/10.1016/j.cell.2021.08.025.
205. Lagunas,T., Plassmeyer, S.P., Fischer, A.D., Friedman,
R. Z., Rieger, M. A., Selmanovic, D., Sarafinovska, S.,
Sol, Y.K., Kasper, M.J., Fass, S.B., Aguilar Lucero, A.F.,
An, J. Y., Sanders, S. J., Cohen, B. A., and Dougherty,
J. D. (2023) A Cre-dependent massively parallel re-
porter assay allows for cell-type specific assessment
of the functional effects of non-coding elements in
vivo, Commun. Biol., 6, 1151, https://doi.org/10.1038/
s42003-023-05483-w.
206. Reynoso, M. A., Juntawong, P., Lancia, M., Blanco,
F.A., Bailey-Serres,J., and Zanetti, M.E. (2015) Trans-
lating ribosome affinity purification (TRAP) followed
by RNA sequencing technology (TRAP-SEQ) for quan-
titative assessment of plant translatomes, Methods
Mol. Biol., 1284, 185-207, https://doi.org/10.1007/978-
1-4939-2444-8_9.
207. Blahetek, G., Mayer, C., Zuber, J., Lenter, M., and
Strobel, B. (2024) Suppression of toxic transgene ex-
pression by optimized artificial miRNAs increases
AAV vector yields in HEK-293 cells, Mol. Ther. Meth-
ods Clin. Dev., 32, 101208, https://doi.org/10.1016/
j.omtm.2024.101280.
SELECTION OF UTRs 753
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
208. Perez-Garcia, C.G., Diaz-Trelles,R., Vega, J.B., Bao,Y.,
Sablad, M., Limphong, P., Chikamatsu, S., Yu, H.,
Taylor,W., Karmali, P.P., Tachikawa,K., and Chivuku-
la, P. (2022) Development of an mRNA replacement
therapy for phenylketonuria, Mol. Ther. Nucleic Acids,
28, 87-98, https://doi.org/10.1016/j.omtn.2022.02.020.
209. Gadalla, K. K. E., Vudhironarit, T., Hector, R. D.,
Sinnett, S., Bahey, N. G., Bailey, M. E. S., Gray, S. J.,
and Cobb, S. R. (2017) Development of a novel AAV
gene therapy cassette with improved safety features
and efficacy in a mouse model of Rett syndrome,
Mol. Ther. Methods Clin. Dev., 5, 180-190, https://
doi.org/10.1016/j.omtm.2017.04.007.
210. Artemyev,V., Gubaeva,A., Paremskaia, A.I., Dzhioeva,
A. A., Deviatkin, A., Feoktistova, S. G., Mityaeva, O.,
and Volchkov, P.Y. (2024) Synthetic promoters in gene
therapy: design approaches, features and applications,
Cells, 13, 1963, https://doi.org/10.3390/cells13231963.
211. Paremskaia, A. I., Kogan, A. A., Murashkina, A.,
Naumova, D.A., Satish, A., Abramov, I. S., Feoktistova,
S.G., Mityaeva, O.N., Deviatkin, A.A., and Volchkov,
P. Y. (2024) Codon-optimization in gene therapy:
promises, prospects and challenges, Front. Bioeng.
Biotechnol., 12, 1371596, https://doi.org/10.3389/fbioe.
2024.1371596.
212. Gentner, B., and Naldini, L. (2012) Exploiting
microRNA regulation for genetic engineering, Tis-
sue Antigens, 80, 393-403, https://doi.org/10.1111/
tan.12002.
213. Geisler,A., Jungmann,A., Kurreck,J., Poller,W., Katus,
H. A., Vetter, R., Fechner, H., and Müller, O. J. (2011)
MicroRNA122-regulated transgene expression in-
creases specificity of cardiac gene transfer upon in-
travenous delivery of AAV9 vectors, Gene Ther., 18,
99-209, https://doi.org/10.1038/gt.2010.141.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in published
maps and institutional affiliations. AI tools may have
been used in the translation or editing of this article.