ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 11, pp. 1741-1756 © Pleiades Publishing, Ltd., 2025.
Russian Text © The Author(s), 2025, published in Biokhimiya, 2025, Vol. 90, No. 11, pp. 1862-1878.
1741
Activity of METTL4 Methyltransferase
Is Crucial for Maintaining Optimal Splicing
Efficiency in HeLa S3 Cells
Anastasiia K. Bolikhova
1,2,3,a
*
#
, Andrey I. Buyan
3,4#
, Maria A. Khokhlova
1,3
,
Sofia S. Mariasina
2,3,5
, Anton R. Izzi
1,6
, Alexander Y. Rudenko
2
,
Marina V. Serebryakova
2
, Alexander M. Mazur
7
,
Olga A. Dontsova
1,2,3,8
, and Petr V. Sergiev
1,2,3
1
Skolkovo Institute of Science and Technology, Center for Life Sciences, 121205 Skolkovo, Russia
2
Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University,
119991 Moscow, Russia
3
Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
4
Institute of Protein Research, Russian Academy of Sciences,
142290 Pushchino, Moscow Region, Russia
5
Research and Educational Resource Center “Pharmacy”, RUDN University, 117198 Moscow, Russia
6
Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University,
119991 Moscow, Russia
7
Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences,
119071 Moscow, Russia
8
Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry,
Russian Academy of Sciences, 117997 Moscow, Russia
a
e-mail: anastasia_b7@mail.ru
Received August 8, 2025
Revised November 11, 2025
Accepted November 13, 2025
AbstractMethyltransferases that modify spliceosomal small nuclear RNAs (snRNAs) play a crucial role in
the cell by ensuring proper maturation of snRNAs, which is essential for optimal function of spliceosome.
In this study, we investigated the enzyme METTL4, which catalyzes N6-methylation of 2′-O-methyladenosine
at position 30 of U2 snRNA. Function of both the protein and the modification in splicing remains unclear.
We demonstrated that inactivation of the METTL4 gene in HeLa S3 cells leads to significant changes in al-
ternative splicing, general slowdown in spliceosome activity, and intron accumulation. In the cells lacking
METTL4, expression of the set of genes associated with ribosomal RNA maturation is reduced, and the number
of coilin-positive structures, most likely Cajal bodies, is decreased in the nuclei of these cells.
DOI: 10.1134/S0006297925602382
Keywords: splicing, small nuclear RNAs (snRNAs), METTL4, U2 snRNA, methylation
* To whom correspondence should be addressed.
# These authors contributed equally to this study.
INTRODUCTION
N6-methyladenosine (m
6
A) is one of the most
common modified nucleotides in eukaryotic RNA [1].
m
6
A is found in various types of RNA, including mes-
senger RNA (mRNA), transfer RNA (tRNA), ribosomal
RNA (rRNA), long non-coding RNAs, and small nuclear
RNAs (snRNAs) [2-6].
Addition of a methyl group to the N6 atom of
adenosine could perform diverse regulatory functions
depending on position of the nucleotide and type of
the modified RNA. In the nucleus, the pattern of m
6
A
distribution in pre-mRNA correlates with different
BOLIKHOVA et al.1742
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
splicing rates of introns [7]; in the cytoplasm, recog-
nition of m
6
A could either enhance translation effi-
ciency or lead to mRNA degradation [8, 9].
In the case of non-coding RNAs, modifications,
including m
6
A, could play regulatory and function-
al roles. For example, cessation of N6-methylation of
A1832 in 18S rRNA or A4220 in 28S rRNA disrupts
ribosome assembly, which, in turn, affects translation
efficiency [10, 11]. The modified m
6
A43 residue in U6
snRNA directly participates in splicing by stabilizing
interaction between the U6 snRNA and the 5′ splice
site [12, 13].
The range of enzymes that perform N6-methyla-
tion of adenosine is extremely diverse [14]. The meth-
yltransferase complex METTL3–METTL14, which has
several RNA targets and is involved in regulation of
cell differentiation, is well studied [15]. Another such
methyltransferase is METTL4, whose function has not
yet been fully deciphered. The role of this enzyme,
according to the current understanding, may vary
depending on the type and state of the cell. Under
hypoxia, METTL4 causes DNA methylation, regulating
the cluster of genes associated with hypoxia response
[16, 17]. Formation of m
6
A in the mitochondrial DNA
by METTL4 plays an important role in the develop-
ment of inflammation [18]. METTL4 can methylate
mRNA and microRNA under certain conditions [19,
20]. However, the constitutive and evolutionarily
conserved function of METTL4 is formation of m
6
Am
from 2′-O-methyladenosine (Am) at position 30 of U2
snRNA [21,  22]. In some organisms, such as worms
and fruit flies, the enzyme that 2′-O-methylates the
30th adenine in U2 snRNA is absent, and METTL4
orthologs modify A30 instead of Am30 [23,  24]. The
30th adenine of U2 snRNA is located immediately up-
stream the branch point recognition site [25], making
modification of this residue a potential mechanism
for regulating efficiency and accuracy of splicing.
At the later stages of splicing, m
6
Am30 is located
next to the duplex formed by the complementary
parts of U2 and U6 [26], potentially affecting stability
and conformation of the U2–U6 complex.
In this work, we aimed to study how inactiva-
tion of the gene encoding the METTL4 protein af-
fects HeLa S3 cells. We confirmed earlier observa-
tions that disappearance of METTL4 in human cells
leads to cessation of N6-methylation of Am30 in U2
snRNA [27]. We demonstrated significant changes in
alternative splicing in the HeLa S3 ΔMETTL4 cell line,
showed that inactivation of METTL4 is accompanied
by the decrease in the number of Cajal bodies (CBs),
and general decrease in the accuracy and efficiency
of splicing. Thus, METTL4 appears to play an im-
portant role in maturation of the U2 snRNA, and its
activity is important for optimal functioning of the
spliceosome.
MATERIALS AND METHODS
Oligonucleotide synthesis. All oligonucleotides
(primers, probes) were synthesized by Lumiprobe
LLC (Russia and EU); all sequences are provided in
Table S1 in the Online Resource 1.
Cell culturing. HeLa S3 cells were cultured in
a DMEM/F12 medium (Gibco, USA). Before use, the
medium was supplemented with fetal bovine serum
(50  mL per 500  mL of medium; Gibco), a mixture
of penicillin and streptomycin antibiotics (5  mL per
500  mL of medium; Gibco), and GlutaMAX (5  mL per
500  mL of medium; Gibco). Cells were cultured at
37°C, 5%  CO
2
. Passaging was performed by scraping
upon reaching 70-80% confluency.
Inactivation of METTL4 gene. The METTL4 gene
was inactivated in the HeLa S3 cell line using the
CRISPR-Cas9 method. A guide RNA (gRNA) sequence
was generated using CRISPR guide RNA design (https://
benchling.com). This gRNA (5′-GGAGCCGTTCGTAATGC-
GAG-3′) targets exon  4. Sense and antisense oligonu-
cleotides (MET4_hum_CrF and MET4_hum_CrR) were
annealed and cloned into a pX458 vector (Addgene:
48138) [28].
HeLa S3 cells were transfected with the plasmid
using a Lipofectamine™ 3000 reagent (Invitrogen,
USA). After 48  h, GFP-positive cells were sorted using
a BD FACSAria™ III (BD Biosciences, USA) and plat-
ed as single clones. Individual clones were analyzed
by PCR (MET4_hum_ChF and MET4_hum_ChR) and
Sanger sequencing of the target region.
Western blotting. The following antibodies were
used to detect proteins: anti-METTL4 (Invitrogen),
anti- GAPDH (Abcam, UK).
Isolation and analysis of U2 snRNA. Total RNA
was isolated from cells using an ExtractRNA reagent
(Evrogen, Russia). U2 snRNA was isolated using a
5′-biotinylated oligonucleotide (U2_BIO), as described
in Laptev et al. [29] and in the Supplementary Meth-
ods section in the Online Resource 1.
Fluorescence in situ hybridization (FISH). DNA
probes complementary to U2 snRNA targets and la-
beled with Cy5 fluorescent dye were designed. HeLa
S3 cells were seeded onto poly-lysine slides, fixed
with a fresh 4% paraformaldehyde (PFA) solution.
Cells were permeabilized in PBS (Sigma-Aldrich, USA)
containing 0.01% Triton X-100. Background blocking
was performed with a 1% glycine solution in PBS.
Pre-hybridization was carried out in a ULTRAhyb™-
Oligo buffer (Thermo Fisher Scientific, USA); hybrid-
ization was performed in the same buffer with ad-
dition of 100  ng of probe. After washing, cells were
stained with a mixture of DAPI (1  mg/mL) and Mowiol
(1  :  1000; Sigma-Aldrich). Samples were analyzed us-
ing a Celena X fluorescence microscope (Logos Biosys-
tems, South Korea). The staining protocol is detailed
IMPORTANCE OF METTL4 FOR MAINTAINING EFFICIENT SPLICING 1743
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
in the Supplementary Methods section in the Online
Resource 1.
Immunofluorescence of Cajal Bodies. HeLa S3
cells were seeded onto poly-lysine slides. Cells were
fixed with a 4% PFA solution. Cells were permeabi-
lized in PBST (PBS containing 0.05% Tween-20) and
blocked with a 1% bovine serum albumin solution
in PBST. Cells were stained with primary antibod-
ies against coilin (Abcam) and secondary antibodies
against rabbit antibodies labeled with Cy5 fluorescent
dye (Thermo Fisher Scientific). After staining with a
mixture of Mowiol and DAPI, samples were analyzed
using a Nikon C2 confocal microscope (Nikon, Japan)
in 9 planes, after which the stack was projected onto
a single plane. DAPI-stained nuclei were used to de-
tect regions of interest (ROI), and number of CBs
within each ROI was automatically counted. Count-
ing was performed using ImageJ software; the script
is available at: https://github.com/bio-mother/Cajal-
bodies-counting-jython-. The staining protocol is de-
tailed in the Supplementary Methods section in the
Online Resource 1.
RT-PCR and qPCR. For total RNA extraction,
HeLa S3 cells were grown to a concentration of 1 mil-
lion/mL (~40% confluency); cells were collected and
resuspended in 300  µL of QIAzol reagent (QIAGEN,
Germany) per 1 million cells. RNA samples were ex-
tracted according to the manufacturer’s instructions.
After removal of genomic DNA using DNase  I
(Thermo Fisher Scientific), cDNA synthesis was per-
formed using a Maxima First Strand cDNA Synthe-
sis Kit for RT-qPCR (Thermo Fisher Scientific). PCR
was performed using a DreamTaq PCR Master Mix
(2X) (Thermo Fisher Scientific) according to the rec-
ommended protocol; annealing temperature for all
primers was 60°C. PCR products were separated on
a 1% agarose gel at 200  V. For real-time PCR, a Maxi-
ma SYBR Green/ROX qPCR Master Mix (Thermo Fisher
Scientific) was used.
Isolation of a newly synthesized RNA. Cells
were seeded before labeling at a concentration of
1  million/mL. The culture medium was supplement-
ed with 5-ethynyluridine (5-EU) at concentration of
0.5  mM (200  mM 5-EU stock solution in DMSO was
stored at −20°C). Cells were incubated with 5-EU for
10 and 20 min. After incubation, cells were collect-
ed, separated from the medium, and resuspended in
300  µL of a QIAzol lysis reagent (QIAGEN, Germany)
per 1 million cells. RNA samples were extracted ac-
cording to the manufacturers instructions. Newly
synthesized RNA labeled with 5-EU was purified us-
ing a Click-iT™ Nascent RNA Capture Kit (Invitrogen,
USA) according to the protocol described previously
[30]; a brief description of the protocol is available
in the Supplementary Methods section in the Online
Resource 1.
RNA sequencing library preparation. A NEBNext
Ultra II RNA Library Prep Kit for Illumina (NEB, USA)
was used for library preparation. Beads were mixed
with a first-strand buffer, random primers, and wa-
ter according to the manufacturers protocol and
incubated at 94°C for 12  min to obtain longer RNA
fragments. Subsequent steps were performed accord-
ing to the manufacturer protocol; the resulting cDNA
library was sequenced using an Illumina Hiseq1500
Sequencing System (Illumina, USA).
High-throughput sequencing data analysis. De-
multiplexed fastq files from Illumina were processed
using TrimGalore (version 0.6.6) to remove adapters
and low-quality bases from the ends of reads and
filter them by a minimum length of 20 nucleotides
[31]. After quality control using FastQC (version 0.11.9),
human genome GRCh38 indices were generated using
STAR (version 2.7.10) -runMode genomeGenerate and
gencode annotation (version 42) with parameters two-
passMode Basic and others specified in the ENCODE
standard long RNA-seq pipeline (see STAR manual) [32,
33]. To identify chimeric reads, default STAR-Fusion
parameters were added, including
--chimSegmentMin 12,
--chimJunctionOverhangMin 8,
--chimOutJunctionFormat 1,
--alignSJstitchMismatchNmax 5-1 5 5,
--chimMultimapScoreRange 3,
--chimMultimapNmax 20,
--chimScoreJunctionNonGTAG-4,
--chimNonchimScoreDropMin 10,
--alignInsertionFlush Right.
To estimate the proportion of reads mapping
to exons, they were counted on genes using featu-
reCounts from the Rsubread package (v.2.12.0) [34].
Only uniquely aligned reads were retained using sam-
tools (version 1.3.1) -q255, and those corresponding to
small and repetitive RNA genes (including ribosomal,
transfer, small nuclear and nucleolar, and microRNAs)
were filtered out according to gencode comprehen-
sive (v.42), RNAatlas, UCSC RepeatMasker, sno/miRNA,
and tRNA annotations [35-37]. For this, bedtools (ver-
sion 2.30.0) intersect was used with parameters -split,
-v, and -f 0.9 (minimum fraction of the read over-
lapping with small RNA) [38]. Next, gene and exon
coverage were calculated using featureCounts with
allowMultiOverlap  =  T and GTF.featureType  =  “gene”
or GTF.featureType  =  “exon,” respectively, using the
same gencode comprehensive annotation as in the
alignment step. Finally, genes with a non-zero total
read count in each sample and at least 3cpm (counts
per million) in at least one sample were retained.
All plots presented in the article were generated
using the R package ggplot2 (version 3.4.0). Principal
component analysis was performed using fviz_pca_ind
from the R package factoextra (version 1.0.7).
BOLIKHOVA et al.1744
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Splicing rate assessment. To assess kinetics of
splicing at the level of individual donor and acceptor
sites, proportion of unspliced reads among all over-
lapping regions was calculated using the R package
FRASER (v.1.10.0) [39]. For this, bam files contain-
ing uniquely aligned reads were imported into R
using the FraserDataSet function, after which read
counting was performed using countRNAData with
parameters keepNonStandardChromosomes  =  FALSE,
filter  =  FALSE, and the genome from the BSgenome.
Hsapiens.UCSC.hg38 package (version 1.4.4). Next, for
each splicing site, Ψ values were calculated using the
calculatePSIValues function by default. These values
reflect proportion of unspliced reads overlapping the
RNA region of interest among all reads crossing it.
Finally, the splicing rate at 10 and 20 min was cal-
culated as 1  −  (Ψ_{10  min/20  min}  −  Ψ_{total RNA})
and adjusted by multiplying by (1  −  Ψ_{total RNA})
to account for intron retention events.
Splicing accuracy assessment. Splicing accuracy
was assessed by calculating the Shannon entropy of
Ψ values for 5′ and 3′ splice sites using FRASER (ver-
sion 1.10.0) [39]; a detailed description of the meth-
od is available in the Supplementary Methods section
in the Online Resource 1.
Differential expression analysis. Differential ex-
pression analysis was performed using edgeR (version
3.42.4). Read counts on gene exons were normalized
using calcNormFactors, followed by model parameter
estimation and testing using estimateDisp, glmQLFit,
and glmQLFTest.
Gene set enrichment analysis (GSEA) was per-
formed using the fgsea package (version 1.26.0) for
the genes ranked by log
2
FC (for differential expres-
sion) or negative log
10
Pv with the sign of deltaPSI
(for alternative splicing), using gene sets from MSigDB
(2022.1) [40].
We also calculated number of the reads on in-
trons and exons using gencode v.42 annotation with
addition of intronic regions, using the featureCounts
function with parameters allowMultiOverlap  =  T and
GTF.featureType  =  “exon” or “intron.” Previously, we
filtered out reads mapped to repetitive genes and
small RNA genes. Calculation of the number of reads
on introns and exons was used to assess changes in
the intron and exon expression levels in the ΔMETTL4
cells compared to the wild-type (WT) cells relative
to the overall change in gene expression (i.e., to as-
sess changes in intron usage considering differential
gene expression). This was done using edgeR (version
3.42.4) in the same way as for differential expression
analysis, but using the diffSpliceDGE function.
Alternative splicing. Alternative splicing
events were identified using rMATS (version 4.1.2)
with parameters: -t single, -variable-read-length,
-ReadLength 120, and gencode v.42 annotation [41].
Complementarity of the splice sites to snRNA was
determined as proposed in Bolikhova et al. [30]; a
brief description is available in the Supplementary
Methods section in the Online Resource 1.
RESULTS
METTL4 methyltransferase is responsible for
the formation of m
6
Am30 in U2 snRNA. Previous
studies have shown that METTL4 methyltransferase
in human cells is responsible for formation of m
6
Am
from Am at position 30 of the spliceosomal U2 snRNA
[21, 27]. To confirm this function, we created the HeLa
S3 cell line with inactivated METTL4 gene (HeLa S3
ΔMETTL4). CRISPR/Cas9 technology and guide RNA
targeting exon 4 of the gene were used to create the
cell line (the cleavage site was chosen so that a frame-
shift mutation would eliminate the main structural
part of the protein, including the SAM binding site).
A monoclonal cell line was selected in which a single
nucleotide insertion occurred in both alleles encod-
ing METTL4 (Fig. 1a, “i1”). The high-throughput RNA
sequencing data from the HeLa S3 WT and HeLa S3
ΔMETTL4 cells confirmed presence of the insertion in
the mRNA encoding METTL4. Western blot analysis
confirmed absence of the METTL4 protein in the HeLa
S3 ΔMETTL4 cells compared to the wild-type cells
(HeLa S3 WT; Fig. 1b). Disappearance of METTL4 did
not affect growth rate of the HeLa S3 cells (Fig.S1 in
the Online Resource 1). In agreement with the previ-
ous studies, inactivation of METTL4 led to disappear-
ance of one methyl group in the corresponding region
of U2 snRNA, as shown by mass spectrometry (Fig.1c).
Absence of METTL4 methyltransferase does
not affect quantity or localization of the spliceo-
somal snRNAs. Modifications could significantly af-
fect processing, localization, and stability of certain
RNA groups [42,  43]. Using RT-qPCR, we showed that
expression of the U2 snRNA in the HeLa S3 ΔMETTL4
cells does not change compared to the wild type
(Fig. 2a). Similarly, expression of other spliceoso-
mal snRNAs does not change with disappearance of
METTL4 (Fig.2a). Localization of various snRNAs also
does not depend on the presence of METTL4 in the
cell, as shown by RT-qPCR; total RNA for the reaction
was isolated separately from the cytoplasm and nu-
clei of the HeLa S3 WT and HeLa S3 ΔMETTL4 cells
(Fig. 2b). Same distribution of the U2 snRNA in both
cell lines was additionally confirmed by staining with
FISH probes specifically binding to U2 (Fig. 2c).
Part of the U2 snRNA maturation process oc-
curs within Cajal bodies (CBs) – nuclear structures
where snRNA modification and final stages of the
small nuclear ribonucleoprotein (snRNP) maturation
take place [44-46]. It is also known that disruption
IMPORTANCE OF METTL4 FOR MAINTAINING EFFICIENT SPLICING 1745
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Fig.  1. Confirmation of the HeLa S3 ΔMETTL4 phenotype. a) Sanger sequencing of the target region in the METTL4 gene
(“i1” in the cell line with gene inactivation indicates insertion of one nucleotide). gRNA indicates the sequence corre-
sponding to the guide RNA; Protospacer adjacent motif (PAM) is the site recognized by the CRISPR/Cas9 system. b)Amount
of METTL4 protein in total lysates of the HeLa S3 WT and HeLa S3 ΔMETTL4 cell lines, shown by Western blotting.
GAPDH was used as a loading control. c)Mass spectra of U2 snRNA after RNase T1 treatment. The fragment CUAAGmAUCA
m
6
AmGp (calculated average mass-to-charge ratio (m/z) is 3619) corresponds to the modified form of U2 snRNA; the frag-
ment CUAAGmAUCAAmGp (calculated average m/z is 3619) corresponds to the form without 2′-O-methylation at position 30.
Peaks corresponding to these fragments are indicated by arrows on the graph. The spectra reflect disappearance of the
modification in the cell line with METTL4 inactivation.
of snRNP maturation could affect the number of
CBs in the cell [47]. The HeLa S3 WT and HeLa S3
ΔMETTL4 cells were stained with antibodies against
coilin (a protein that forms the structure of CBs [48])
and DAPI, which stains DNA. By analyzing the num-
ber of coilin-positive signals in the 1070 WT and
780 ΔMETTL4 nuclei, we found a significant reduc-
tion in the number of such signals in the cells lack-
ing methyltransferase (Fig. 2d and e; images of cells
stained for coilin, with nuclei indicated, are available
in Fig. S1 in the Online Resource 1).
Disappearance of METTL4 leads to noticeable
changes in gene expression. Although METTL4 does
not directly affect the quantity of snRNAs, changes
in the expression levels of various genes are one
of the main mechanisms allowing the cell to re-
spond to changing conditions [49]. By performing
high-throughput RNA sequencing of the HeLa S3 WT
BOLIKHOVA et al.1746
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Fig. 2. Overall effect of METTL4 inactivation on quantity and maturation of spliceosomal snRNAs. a) Analysis of snRNA
expression in the HeLa S3 WT and HeLa S3 ΔMETTL4 cell lines performed using RT-qPCR. Six biological replicates were
used for each cell line. Expression level of the target was normalized to the expression level of the housekeeping gene
GAPDH. The graph shows mean expression value and spread of values. Gray bars denote WT, purple bars denote ΔMETTL4.
Statistical significance was determined using the two-sided Mann–Whitney U test; ns, p  > 0.05. b) Analysis of distribution
of spliceosomal snRNAs in the HeLa S3 WT and HeLa S3 ΔMETTL4 cells performed using RT-qPCR. Three biological rep-
licates were used for each cell line. Y-axis shows the ratio of the amount of specific RNA in the nuclear and cytoplasmic
fractions. Bars reflect the mean and spread of values. Gray bars denote WT, purple bars denote ΔMETTL4; ns, p  > 0.05.
c) Micrographs of the HeLa S3 WT and HeLa S3 ΔMETTL4 cells: nuclei stained with DAPI (left panel, blue), U2 snRNA
stained with Cy5-labeled FISH probes (middle panel, purple); right panel is a merged image. d)Distribution of the number
of coilin-positive signals in the nuclei of the HeLa S3 WT (gray) and HeLa S3 ΔMETTL4 (purple) cells. Sample sizes: 1070
and 780 nuclei for WT and ΔMETTL4, respectively. Statistical significance was determined using the unpaired Student’s
t-test; ***p  0.001. e) Micrographs of the HeLa S3 WT and HeLa S3 ΔMETTL4 cells taken in 9 planes and projected onto a
single plane: nuclei stained with DAPI (left panel, blue), cells stained with antibodies against coilin (middle panel, purple);
the right panel is a merged image.
IMPORTANCE OF METTL4 FOR MAINTAINING EFFICIENT SPLICING 1747
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Fig. 3. Analysis of expression changes occurring upon METTL4 inactivation. a) Results of differential expression analysis
of 13,813 genes (with cpm ≥ 3 in at least three samples) in the HeLa S3 ΔMETTL4 cells compared to the wild-type cells.
Vertical dashed lines correspond to |log
2
FC| = 0.5; horizontal line denotes FDR = 0.05. In total, 1,217 genes are more ac-
tively expressed in the ΔMETTL4 cells compared to the wild type (highlighted in blue), and 946 are less actively expressed
(highlighted in red). Genes with qPCR-confirmed differential expression are marked with a dark border and additionally
labeled. b) Confirmation of high-throughput sequencing results. RT-qPCR was performed in six replicates for each cell
line. Expression level of the target gene was normalized to the expression level of the housekeeping gene GAPDH. Gray
bars denote WT, purple bars denote ΔMETTL4. Statistical significance was determined using the one-sided Mann-Whitney
U test; * p 0.05, ** p 0.01. c) Results of gene set enrichment analysis (GSEA) for 15 pathways with increased (blue) and
decreased (red) regulation and FDR < 0.05. NES – normalized enrichment score.
and HeLa S3 ΔMETTL4 cells (Fig.S2a, TableS2 in the
Online Resource1) and comparing the obtained tran-
scriptomes, we found a number of genes that are more
or less actively expressed in the cells without METTL4
compared to the wild type cells (Fig. 3a); some find-
ings were confirmed using RT-qPCR (Fig. 3b). When
conducting gene set enrichment analysis (GSEA) of
the differentially expressed genes with gene ontol-
ogy terms, we found activation of the pathways as-
sociated with the development of immune response
(“Positive regulation of Natural Killer cell activation”
and “Interferon alpha response”; Fig.  3c) in the HeLa
S3 ΔMETTL4. Among the pathways inhibited in the
HeLa S3 ΔMETTL4 cells are those associated with RNA
processing (e.g., “rRNA metabolic process” and “RNA
processing”; Fig. 3c).
BOLIKHOVA et al.1748
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
a
b
Fig. 4. Analysis of alternative splicing in the HeLa S3 ΔMETTL4 cells compared to the wild-type cells. a)  Overall changes
identified by examining 68,005 alternative splicing events (6,020 alternative 3′ splice sites, 4,046 alternative 5′ splice sites,
45,727 exon skipping events, 4,732 intron retention events, 7,480 mutually exclusive exons), where at least one isoform
had coverage of ≥10 reads in all replicates. Red dots indicate events with FDR <  0.05 and |ΔPSI|  >  5%. Number of alter-
native splicing events exceeding the threshold values is indicated in red on the graph. Events successfully confirmed by
RT-qPCR are additionally highlighted with a dark border and labeled. b) Confirmation of the high-throughput sequenc-
ing results using RT-PCR followed by gel electrophoresis. Each experiment was performed for three biological replicates.
For each event, the electrophoresis of RT-PCR products in agarose gel is shown below, and quantitative assessment of
the product ratio on the gel is shown above. Gray bars denote WT, purple bars denote ΔMETTL4. Statistical significance
was determined using the one-sided Mann–Whitney U test; *  p ≤  0.05.
IMPORTANCE OF METTL4 FOR MAINTAINING EFFICIENT SPLICING 1749
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Alternative splicing shifts in the HeLa S3
ΔMETTL4 cells. Presence of modified bases in spliceo-
somal snRNAs plays an important role in both regula-
tion of the splice site and branch point recognition, as
well as in maintaining interactions that ensure struc-
ture of the spliceosome [6]. Earlier studies suggested
that modification of A30 in the U2 snRNA specifically
affects splicing, as this residue is located immediate-
ly before the branch point recognition site [21, 27].
We compared the ratio of alternative splicing prod-
ucts in the HeLa S3 WT and the HeLa S3 ΔMETTL4
cells. The analysis revealed a significant number of
differentially spliced genes, some of which were con-
firmed using RT-PCR (Fig.  4, a and b; Table  S3 in the
Online Resource 1).
It can be noted that the largest number of events
changing upon the METTL4 inactivation belongs to
“Exon Skipping,” with both increased inclusion and
exclusion of the exon in the HeLa S3 ΔMETTL4 com-
pared to the wild type depending on the specific splic-
ing event.
Splicing changes in the HeLa S3 ΔMETTL4 cells
are general in nature. Modifications of snRNAs can
have both general and specific effects on splicing [6,
13]. We examined which groups of alternative exons
are most susceptible to change upon the METTL4 in-
activation. For the alternative exons that are includ-
ed to a greater, equal, or lesser extent in the HeLa
S3 ΔMETTL4 cells in comparison with the wild-type
cells, we compared lengths of the exons themselves,
as well as of the surrounding introns (Fig. 5a). As a
result, it was found that the long alternative exons
are on average more represented in the ΔMETTL4
(ΔPSI  <  −5%), while the short ones are more likely to
be excluded in the cells without the methyltransferase
(ΔPSI  >  5%). At the same time, length of the following
intron is slightly greater for the events characterized
by exon skipping in the ΔMETTL4. In the next step,
for similar groups of the alternatively spliced exons,
we compared complementarity of the 5′ splice site of
the upstream and downstream introns to the sequenc-
es of U1, U5, and U6 snRNAs, as well as the region
including the branch point – U2 snRNA (Fig.S3a and
S3b in the Online Resource 1). No significant correla-
tion between complementarity and splicing changes
in the ΔMETTL4 was found. A similar analysis was
performed for the second most changing group of
splicing events – intron retention (Fig. 5b; Fig. S3c in
the Online Resource 1). The main and only pattern
is that the longer introns are more often retained in
the cell line without the METTL4 methyltransferase.
To demonstrate potential functional role of splic-
ing changes, we identified biological processes en-
riched in the alternatively spliced genes (GSEA); the
analysis results are shown in Fig. 5c. As in the case
of differential expression, pathways related to RNA
maturation and immune response were found. A pos-
sible explanation may be that some of the splicing
changes are direct consequences of the changes in
gene expression upon METTL4 inactivation, and vice
versa, that the alternative splicing of certain genes
could directly affect expression profile of the cell.
We also determined whether the METTL4 meth-
yltransferase has a general effect on splicing by com-
paring efficiency and accuracy of splicing of all in-
trons in the cell. As a measure of splicing efficiency,
we used the average proportion of exon representa-
tion relative to the total gene coverage (Fig.5d). As a
measure of splicing accuracy, the Shannon entropy of
Ψ values of 5′ and 3′ splice sites was calculated (see
“Materials and Methods” section; Fig.5e). We showed
that the average splicing efficiency decreases upon
METTL4 inactivation (the median proportion of exon
reads in total RNA of HeLa S3 WT is 0.9097649, in
total RNA of the HeLa S3 ΔMETTL4 it is 0.9001537;
p < 10
−15
), but the average splicing accuracy remains
unchanged.
Splicing rate in the HeLa S3 ΔMETTL4 cells is
significantly lower than in the wild-type cells. Splic-
ing is an extremely dynamic process, and changes in
it may not be noticeable when considering only total
RNA. To determine how METTL4 affects the splicing
rate, we used the analysis method described in Bolik-
hova et al. [30]. Briefly, the newly synthesized RNA
after 10 and 20 min of labeling cells with 5-EU was
isolated and subjected to high-throughput sequenc-
ing (Fig.  S2a and S2b in the Online Resource  1). By
comparing the degree of splicing of each intron in
the newly synthesized RNA separately after 10 and
20 min of labeling and in the poly-A fraction of total
RNA (used as an indicator of isoform ratio in mature
RNA), we calculated the splicing rate for each donor
and acceptor site. The rate values obtained for the
RNA after 10 and 20 minutes of labeling were similar
(Fig.S2c and S2d in the Online Resource  1), confirming
applicability of this method for assessing splicing rate
of each site. Analysis of the values for RNA isolated
after 10min of labeling is presented in the main text,
and after 20 min in Fig. S4 in the Online Resource 1.
The average splicing rate in the HeLa S3 ΔMETTL4
cell line was lower than in the wild type (Fig.  6a;
Fig.  S4a in the Online Resource 1). By analogy with
the analysis of the total poly-A fraction, we calculated
the Shannon entropy of Ψ values of 5′ and 3′ splice
sites in the newly synthesized RNA (Fig. 6b; Fig. S4b
in the Online Resource  1). As seen in the graphs, in
the newly synthesized RNA isolated after 10  min, the
splicing accuracy in the ΔMETTL4 is significantly
lower than in the wild type (Fig. 6b), but this effect
disappears after 20 min of labeling. Thus, only in the
“youngest” RNAs the presence of METTL4 does affect
splicing accuracy, and this effect is quickly neutralized
BOLIKHOVA et al.1750
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
a
b
c
d
Fig. 5. Splicing changes upon METTL4 inactivation. a)  Distribution of lengths of alternatively included exons (central graph),
as well as the upstream and downstream introns (left and right graphs, respectively) depending on the degree of inclusion/
exclusion of the alternative exon in the HeLa S3 ΔMETTL4 cells compared to the wild type. b)  Distribution of lengths of
alternative introns (central graph), as well as the upstream and downstream exons (left and right graphs, respectively)
depending on the degree of retention of the alternative intron in the HeLa S3 ΔMETTL4 cells compared to the wild type.
c)  Results of gene set enrichment analysis (GSEA) of alternatively spliced genes with FDR < 0.05. NES, normalized enrich-
ment score. d) Analysis of global splicing efficiency by assessing average proportion of exon representation for 15,035
genes with cpm ≥ 3 in at least three samples. e) Analysis of splicing accuracy through assessment of Shannon entropy
of 5′ and 3′ splice site Ψ values. In total, 56,062 splice sites were considered, for each of which the cpm of the splicing
product was ≥1 in at least one sample. Statistical significance was determined using the one-sided Wilcoxon rank test;
ns, p > 0.05; * p ≤ 0.05; *** p ≤ 0.001; **** p≤10
−15
. Gray color denotes WT, purple denotes ΔMETTL4.
IMPORTANCE OF METTL4 FOR MAINTAINING EFFICIENT SPLICING 1751
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Fig. 6. Analysis of splicing rate and accuracy (RNA isolated after 10min of 5-EU labeling) in the HeLa S3 WT and the HeLa
S3 ΔMETTL4 cells. a)  Comparison of average splicing rates. Values for 68,288 splice sites are shown, for each of which
the cpm of the splicing product is ≥10 in at least three samples. b)  Comparison of splicing accuracy through assessment
of Shannon entropy of 5′ and 3′ splice sites Ψ values. In total, 10,481 splice sites were considered, for each of which the
cpm of the product is ≥1 in at least one sample. Statistical significance was determined using the one-sided Wilcoxon
rank test; **** p≤10
−15
. Gray color denotes WT, purple denotes ΔMETTL4. c)  Relationship between the difference in splic-
ing rates (ΔMETTL4 minus WT) and the average splicing rate (divided into deciles). ART ANOVA p < 10
−15
. d)  Relation-
ship between the difference in splicing rates (ΔMETTL4 minus WT) and the expression level of the corresponding genes
(log
10
(TPM), values divided into deciles). ART ANOVA p < 10
–15
. Dots correspond to the median values; the lower and upper
error bars show 25% and 75% quantiles, respectively. Dashed line reflects the overall trend of the median. Significances
of the Tukey test for the difference between the first and last groups are shown on the graphs (***  p ≤  0.001).
over time, for example, due to degradation of the in-
correctly spliced RNA.
To better understand splicing features in the
HeLa S3 ΔMETTL4 line, we performed correlation
analysis of the degree of change in the splicing rate
with various characteristics of the corresponding in-
trons and exons (only significant results are present-
ed). It was shown that the slow-down of splicing in
the absence of METTL4 is more pronounced for the
initially “slower” introns (Fig.  6c; Fig.  S4c in the On-
line Resource 1). On the other hand, splicing of the
introns that are part of the highly expressed genes,
which is initially quite fast [30], slows down slight-
ly more in the absence of METTL4 (Fig. 6d; Fig. S4d
in the Online Resource 1).
DISCUSSION
Modification of snRNAs plays an important role
in the regulation of splicing, ensuring correct recog-
nition of introns and maintaining optimal structure
of the spliceosome [6,  13, 50]. In this work, we con-
firmed that the METTL4 methyltransferase catalyzes
the N6-methylation of Am at position 30 of the U2
snRNA in human cells [21,  22]. To understand the
role of METTL4 methyltransferase in the life of a eu-
karyotic cell, we analyzed the changes occurring in
the HeLa S3 cells after inactivation of the METTL4
gene. Growth rate of the cells without METTL4 did
not differ from growth rate of the wild-type cells,
which is consistent with the results previously shown
in the HEK293T cell line [27].
Modification of certain RNAs is often a mech-
anism of post-transcriptional regulation of the ex-
pression of the corresponding genes, while changes
in the expression level of snRNAs themselves could
also be an important factor [51,  52]. We did not find
fundamental differences in the expression level of
spliceosomal snRNAs between the wild-type cells
and the cells without METTL4 (Fig.  2a). Maturation
of snRNAs following synthesis is, in turn, a complex,
BOLIKHOVA et al.1752
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
multi-stage process, part of which occurs in the nu-
cleus and part in the cytoplasm [53-55]. The ratio of
the amount of snRNAs in the nucleus and cytoplasm
turned out to be independent of METTL4 (Fig.  2,
b and c). At the same time, we managed to detect
a statistically significant decrease in the number of
coilin-positive signals in the HeLa S3 ΔMETTL4 cells
compared to the HeLa S3 WT (Fig.  2d). Coilin is a
known marker of CBs, but it could also be part of oth-
er intranuclear structures – histone locus bodies [56].
Histone locus bodies are non-membrane intranuclear
structures that play a role in maturation of the his-
tone mRNAs; CBs are similar structures that play an
important role in maturation of the snRNPs [45, 57].
It has been shown that disruption of the final stag-
es of snRNP maturation by reducing the number of
proteins involved in this process could lead to the in-
crease in the number of CBs, thereby helping to main-
tain optimal concentration of the spliceosome compo-
nents [47]. Decrease in the number of coilin-positive
signals hints at the presence of a mechanism by
which inactivation of METTL4 affects maturation of
the U2 snRNA, associated with the presence of U2 in
CBs. However, one cannot rule out a more complex,
possibly not snRNA-mediated, connection between
METTL4 and functioning of the histone locus bodies.
Thus, this issue requires further study.
The most obvious process that could be affected
by snRNA modification is splicing [6]. m
6
Am30 in the
U2 snRNA is located immediately upstream the branch
point recognition site [30], while at the later stages
of splicing, this residue is located next to the duplex
formed by the complementary parts of U2 and U6 [26].
We found that in the cells without METTL4, alternative
splicing is significantly altered, with the largest num-
ber of events changing upon disappearance of the MET-
TL4 belonging to the exon skipping, and distribution
between the more included and excluded exons in the
ΔMETTL4 is equally probable (Fig.4, a and b). It was
noted that splicing in the absence of METTL4 proceeds
more slowly (Fig. 6a), leading, apparently, to a small
but general accumulation of introns for most genes
(Fig.5d). It is important to note the implicit pattern of
the splicing slowdown. Initially, the “slower” introns
are most susceptible to the disappearance of METTL4
(Fig.  6b; Fig.  S3b in the Online Resource  1), which
could be explained by less optimized splicing in these
regions. However, the RNA molecules of more highly
expressed genes, which are normally characterized by
increased splicing rates [30], are more susceptible to
the slow-down upon METTL4 inactivation. A possible
mechanism partially explaining the observed phenom-
ena may be decrease in the concentration of the ma-
ture snRNPs associated with reduction in the number
of CBs. Such change in the availability snRNPs would
more strongly affect both less efficient splicing sites,
which initially attract snRNPs weaker, and the sites of
highly represented RNAs, which, due to their greater
quantity, require higher concentration of snRNPs to
maintain optimal splicing rates.
Modified nucleotides of snRNAs under certain
conditions could directly or indirectly regulate tran-
scription [58, 59]. In the HeLa S3 ΔMETTL4 cells, we
observe a decrease in the expression of a number of
genes associated with rRNA maturation compared to
the wild type. It should be noted that small nucleolar
RNAs (snoRNAs), such as U3, also pass through CBs
during their maturation [48,  60]. At the same time,
snoRNAs play an important role in the maturation of
ribosome subunits [61-63], which could explain con-
nection between the disruption of snRNA maturation
and inhibition of the expression of genes associated
with rRNA maturation. At the same time, in the cells
with METTL4 inactivation, more active expression of
a group of genes involved in the development of in-
flammatory response is observed. A similar effect in
response to the inhibition of splicing, leading to in-
crease in the amount of intracellular double-stranded
RNA due to hybridization of the repetitive elements
within introns, has been described in the literature
before [64].
In summary, inactivation of the METTL4 gene
in the HeLa S3 cells leads to disruption of the U2
snRNA maturation and, through a previously unstud-
ied mechanism, causes decrease in the number of
coilin-positive signals in the cells, which may affect
expression of the genes associated with rRNA matu-
ration. At the same time, the ΔMETTL4 cells exhibit
decrease in the splicing rate, theoretically explained
both by the decrease in concentration of the mature
snRNPs, including due to the reduction in the CB num-
ber, and by the “imbalance” of the spliceosome due
to the disappearance of the N6-methyl group of Am
at position 30 of the U2 snRNA.
CONCLUSION
N6-methylation of adenosine, leading to forma-
tion of m
6
A, is an extremely common modification
found in various types of eukaryotic RNA. m
6
A can
both perform a regulatory function, affecting stability
and transport of mRNA, and play a structural role,
for example, for nucleotides that are part of ribo-
somes. In this work, we studied the role of the MET-
TL4 methyltransferase, one of whose most conserved
functions is N6-methylation of Am at position 30 of
the U2 snRNA.
We showed that disappearance of METTL4 in
the HeLa S3 cells leads to a slow-down in splicing,
thereby causing intron accumulation. A possible
reason for the slow-down in splicing is disruption
IMPORTANCE OF METTL4 FOR MAINTAINING EFFICIENT SPLICING 1753
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
of snRNP maturation associated with the decrease in
the number of CBs. Another possible consequence
of the decrease in the number of CBs is reduc-
tion in concentration of the RNA genes associated
with rRNA maturation, also observed in the cells
without METTL4.
Although importance of METTL4 for splicing effi-
ciency in the HeLa S3 cells is clear, the exact mecha-
nism linking methylation, splicing, and maturation of
various RNAs remains to be investigated.
Abbreviations
5-EU 5-ethynyluridine
Am 2′-O-methyladenosine
CBs Cajal bodies
gRNA Guide RNA
m
6
A N6-methyladenosine
m
6
Am N6,2′-O-dimethyladenosine
qPCR quantitative polymerase chain reaction
RT-qPCR reverse transcription quantitative PCR
snRNA small nuclear RNA
snRNP small nuclear ribonucleoprotein
Supplementary information
The online version contains supplementary material
available at https://doi.org/10.1134/S0006297925602382.
Acknowledgments
We are grateful to the Lomonosov Moscow State Uni-
versity Development Program for providing access
tothe Celena X multiparametric cell analysis system.
Contributions
A. K. Bolikhova and A. I. Buyan designed and conduct-
ed experiments, analyzed data, and wrote the man-
uscript. M. A. Khokhlova conducted experiments and
analyzed data. S. S. Mariasina designed and conduct-
ed experiments, analyzed data, and wrote the manu-
script. A. R. Izzi, A. Y. Rudenko, M. V. Serebryakova, and
A. M. Mazur conducted experiments. O. A. Dontsova su-
pervised the work. P. V. Sergiev supervised the work,
conceptualized the study, and wrote the manuscript.
Funding
This research was financially supported by the state
assignment of Lomonosov Moscow State University.
The experimental work was supported by the Russian
Science Foundation (grant no.21-64-00006-P).
Ethics approval and consent to participate
This work does not contain any studies involving hu-
man and animal subjects.
Conflict of interest
The authors of this work declare that they have
noconflicts of interest.
REFERENCES
1. Jiang, X., Liu, B., Nie, Z., Duan, L., Xiong, Q., Jin, Z.,
Yang,C., and Chen,Y. (2021) Therole of m6A modifi-
cation in the biological functions and diseases, Signal
Transduct. Target. Ther., 6, 74, https://doi.org/10.1038/
s41392-020-00450-x.
2. Boo, S.H., and Kim, Y.K. (2020) Theemerging role of
RNA modifications in the regulation of mRNA stabili-
ty, Exp. Mol. Med., 52, 400-408, https://doi.org/10.1038/
s12276-020-0407-z.
3. Suzuki, T. (2021) The expanding world of tRNA
modifications and their disease relevance, Nat. Rev.
Mol. Cell Biol., 22, 375-392, https://doi.org/10.1038/
s41580-021-00342-0.
4. Sergiev, P. V., Aleksashin, N. A., Chugunova, A. A.,
Polikanov, Y.S., and Dontsova, O.A. (2018) Structural
and evolutionary insights into ribosomal RNA
methylation, Nat. Chem. Biol., 14, 226-235, https://
doi.org/10.1038/nchembio.2569.
5. Kazimierczyk, M., and Wrzesinski, J. (2021) Long
non-coding RNA epigenetics, Int.J. Mol. Sci., 22, 6166,
https://doi.org/10.3390/IJMS22116166.
6. Morais,P., Adachi,H., and Yu, Y.T. (2021) Spliceosomal
snRNA epitranscriptomics, Front. Genet., 12, 652129,
https://doi.org/10.3389/FGENE.2021.652129/XML.
7. Louloupi, A., Ntini,E., Conrad,T., and Ørom, U.A. V.
(2018) Transient N-6-methyladenosine transcrip-
tome sequencing reveals a regulatory role of m6A in
splicing efficiency, Cell. Rep., 23, 3429-3437, https://
doi.org/10.1016/j.celrep.2018.05.077.
8. Wang,X., Zhao, B.S., Roundtree, I.A., Lu,Z., Han,D.,
Ma, H., Weng,X., Chen, K., Shi,H., and He,C. (2015)
N6-methyladenosine modulates messenger RNA
translation efficiency, Cell, 161, 1388-1399, https://
doi.org/10.1016/j.cell.2015.05.014.
9. Wang,X., Lu,Z., Gomez,A., Hon, G.C., Yue,Y., Han,D.,
Fu, Y., Parisien, M., Dai, Q., Jia, G., Ren, B., Pan, T.,
and He, C. (2013) N6-methyladenosine-dependent
regulation of messenger RNA stability, Nature, 505,
117-120, https://doi.org/10.1038/nature12730.
10. Peng,H., Chen,B., Wei,W., Guo,S., Han,H., Yang,C.,
Ma,J., Wang,L., Peng,S., Kuang,M., and Lin,S. (2022)
N6-methyladenosine (m6A) in 18S rRNA promotes
fatty acid metabolism and oncogenic transformation,
Nat. Metab., 4, 1041-1054, https://doi.org/10.1038/
s42255-022-00622-9.
11. Ma,H., Wang,X., Cai,J., Dai,Q., Natchiar, S.K., Lv,R.,
Chen, K., Lu, Z., Chen, H., Shi, Y. G., Lan, F., Fan, J.,
Klaholz, B. P., Pan, T., Shi, Y., and He, C. (2019) N
6-Methyladenosine methyltransferase ZCCHC4 medi-
ates ribosomal RNA methylation, Nat. Chem. Biol., 15,
88-94, https://doi.org/10.1038/S41589-018-0184-3.
12. Warda, A. S., Kretschmer, J., Hackert, P., Lenz, C.,
Urlaub, H., Höbartner, C., Sloan, K. E., and Bohnsack,
M.T. (2017) Human METTL16 is a N6-methyladenos-
BOLIKHOVA et al.1754
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
ine (m6A) methyltransferase that targets pre-mR-
NAs and various non-coding RNAs, EMBO Rep., 18,
2004-2014, https://doi.org/10.15252/EMBR.201744940.
13. Ishigami, Y., Ohira, T., Isokawa, Y., Suzuki, Y., and
Suzuki, T. (2021) A single m6A modification in U6
snRNA diversifies exon sequence at the 5′ splice
site, Nat. Commun., 12, 3244, https://doi.org/10.1038/
s41467-021-23457-6.
14. Yang, Y., Hsu, P. J., Chen, Y.S., and Yang, Y. G. (2018)
Dynamic transcriptomic m6A decoration: Writers,
erasers, readers and functions in RNA metabolism,
Cell. Res., 28, 616-624, https://doi.org/10.1038/S41422-
018-0040-8,.
15. Liu, J., Yue, Y., Han, D., Wang, X., Fu, Y., Zhang, L.,
Jia,G., Yu,M., Lu, Z., Deng,X., Dai,Q., Chen,W., and
He, C. (2014) A METTL3-METTL14 complex mediates
mammalian nuclear RNA N6-adenosine methylation,
Nat. Chem. Biol., 10, 93-95, https://doi.org/10.1038/
nchembio.1432.
16. Lai, J. C. Y., Hsu, K. W., and Wu, K. J. (2024) Inter-
rogation of the interplay between DNA N6-meth-
yladenosine (6mA) and hypoxia-induced chroma-
tin accessibility by a randomized empirical model
(EnrichShuf), Nucleic Acids Res., 52, 13605-13624,
https://doi.org/10.1093/NAR/GKAE1152.
17. Hsu, K. W., Lai, J.C., Chang, J. S., Peng, P. H., Huang,
C. H., Lee, D. Y., Tsai, Y. C., Chung, C. J., Chang, H.,
Chang, C.H., Chen, J.L., Pang, S.T., Hao,Z., Cui, X.L.,
He,C., and Wu, K.J. (2022) METTL4-mediated nuclear
N6-deoxyadenosine methylation promotes metastasis
through activating multiple metastasis-inducing tar-
gets, Genome. Biol., 23, 249, https://doi.org/10.1186/
s13059-022-02819-3.
18. Zheng, L., Chen, X., He, X., Wei, H., Li, X., Tan, Y.,
Min,J., Chen,M., Zhang,Y., Dong,M., Yin,Q., Xue,M.,
Zhang, L., Huo, D., Jiang, H., Li, T., Li, F., Wang, X.,
Li, X., and Chen, H. (2025) METTL4-Mediated mito-
chondrial DNA N6-methyldeoxyadenosine promot-
ing macrophage inflammation and atherosclerosis,
Circulation, 151, 946-965, https://doi.org/10.1161/
CIRCULATIONAHA.124.069574.
19. Sang, A., Zhang, J., Zhang, M., Xu, D., Xuan, R.,
Wang,S., Song,X., and Li,X. (2024) METTL4 mediat-
ed-N6-methyladenosine promotes acute lung injury by
activating ferroptosis in alveolar epithelial cells, Free
Radic. Biol. Med., 213, 90-101, https://doi.org/10.1016/
j.freeradbiomed.2024.01.013.
20. Van den Homberg, D.A.L., van der Kwast, R.V.C.T.,
Quax, P.H.A., and Nossent, A.Y. (2022) N-6-methylad-
enosine in vasoactive microRNAs during hypoxia;
a novel role for METTL4, Int.J. Mol. Sci., 23, 1057,
https://doi.org/10.3390/IJMS23031057.
21. Chen, H., Gu, L., Orellana, E. A., Wang, Y., Guo, J.,
Liu, Q., Wang, L., Shen, Z., Wu, H., Gregory, R. I.,
Xing, Y., and Shi, Y. (2020) METTL4 is an snRNA
m6Am methyltransferase that regulates RNA splicing,
Cell. Res., 30, 544-547, https://doi.org/10.1038/S41422-
019-0270-4,.
22. Luo, Q., Mo, J., Chen, H., Hu, Z., Wang, B., Wu, J.,
Liang,Z., Xie,W., Du,K., Peng,M., Li,Y., Li,T., Zhang,Y.,
Shi, X., Shen, W. H., Shi, Y., Dong, A., Wang, H., and
Ma,J. (2022) Structural insights into molecular mech-
anism for N6-adenosine methylation by MT-A70 fam-
ily methyltransferase METTL4, Nat. Commun., 13,
5636, https://doi.org/10.1038/s41467-022-33277-x.
23. Gu, L., Wang, L., Chen, H., Hong, J., Shen, Z.,
Dhall, A., Lao, T., Liu, C., Wang, Z., Xu, Y., Tang,
H. W., Chakraborty, D., Chen, J., Liu, Z., Rogulja, D.,
Perrimon, N., Wu, H., and Shi, Y. (2020) CG14906
(mettl4) mediates m6A methylation of U2 snRNA in
Drosophila, Cell Discov., 6, 44, https://doi.org/10.1038/
s41421-020-0178-7.
24. Sendinc, E., and Shi, Y. (2023) RNA m6A methyla-
tion across the transcriptome, Mol. Cell., 83, 428-441,
https://doi.org/10.1016/j.molcel.2023.01.006.
25. Van der Feltz, C., and Hoskins, A. A. (2019) Struc-
tural and functional modularity of the U2 snRNP in
pre-mRNA splicing, Crit. Rev. Biochem. Mol. Biol.,
54, 443-465, https://doi.org/10.1080/10409238.2019.
1691497.
26. Karunatilaka, K. S., and Rueda, D. (2014) Post-tran-
scriptional modifications modulate conformational
dynamics in human U2-U6 snRNA complex, RNA, 20,
16-23, https://doi.org/10.1261/RNA.041806.113.
27. Goh, Y.T., Koh, C.W.Q., Sim, D.Y., Roca,X., and Goh,
W. S. S. (2020) METTL4 catalyzes m6Am methylation
in U2 snRNA to regulate pre-mRNA splicing, Nucleic
Acids Res., 48, 9250-9261, https://doi.org/10.1093/NAR/
GKAA684.
28. Ran, F. A., Hsu, P. D., Wright, J., Agarwala, V., Scott,
D.A., and Zhang,F. (2013) Genome engineering using
the CRISPR-Cas9 system, Nat. Protoc., 8, 2281-2308,
https://doi.org/10.1038/nprot.2013.143.
29. Laptev,I., Shvetsova,E., Levitskii,S., Serebryakova,M.,
Rubtsova, M., Zgoda, V., Bogdanov, A., Kamenski, P.,
Sergiev, P., and Dontsova, O. (2020) METTL15 inter-
acts with the assembly intermediate of murine mito-
chondrial small ribosomal subunit to form m4C840
12S rRNA residue, Nucleic Acids Res., 48, 8022-8034,
https://doi.org/10.1093/NAR/GKAA522.
30. Bolikhova, A.K., Buyan, A.I., Mariasina, S.S., Rudenko,
A.Y., Chekh, D.S., Mazur, A.M., Prokhortchouk, E.B.,
Dontsova, O.A., and Sergiev, P.V. (2024) Study of the
RNA splicing kinetics via invivo 5-EU labeling, RNA,
30, 1356-1373, https://doi.org/10.1261/rna.079937.123.
31. Krueger, F., James, F., Ewels, P., Afyounian, E.,
Weinstein, M., Schuster-Boeckler, B., Hulselmans,
G., sclamons (2023) FelixKrueger/TrimGalore: ver-
sion0.6.8, https://doi.org/10.5281/ZENODO.7579519.
32. Dobin, A., Davis, C. A., Schlesinger, F., Drenkow, J.,
Zaleski,C., Jha,S., Batut,P., Chaisson,M., and Gingeras,
T.R. (2013) STAR: Ultrafast universal RNA-seq aligner,
IMPORTANCE OF METTL4 FOR MAINTAINING EFFICIENT SPLICING 1755
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Bioinformatics, 29, 15-21, https://doi.org/10.1093/
BIOINFORMATICS/BTS635.
33. Loveland, J. E., Mudge, J. M., Sisu, C., Wright, J. C.,
Armstrong,J., Barnes,I., Berry,A., Bignell,A., Boix,C.,
Carbonell Sala, S., Cunningham, F., Di Domenico, T.,
Donaldson,S., Fiddes, I.T., García Girón,C., Gonzalez,
J. M., Grego, T., Hardy, M., Hourlier, T., Howe, K.,
Hunt, T., et al. (2021) GENCODE, Nucleic Acids Res.,
49, D916-D923, https://doi.org/10.1093/NAR/GKAA1087.
34. Liao,Y., Smyth, G.K., and Shi,W. (2019) TheRpackage
Rsubread is easier, faster, cheaper and better for align-
ment and quantification of RNA sequencing reads,
Nucleic Acids Res., 47, e47, https://doi.org/10.1093/
NAR/GKZ114.
35. Danecek, P., Bonfield, J. K., Liddle, J., Marshall, J.,
Ohan, V., Pollard, M. O., Whitwham, A., Keane, T.,
McCarthy, S. A., Davies, R. M., and Li, H. (2021)
Twelve years of SAMtools and BCFtools, Gigascience,
10, giab008, https://doi.org/10.1093/GIGASCIENCE/
GIAB008.
36. Lorenzi,L., Chiu, H.S., Avila Cobos,F., Gross,S., Volders,
P. J., Cannoodt, R., Nuytens, J., Vanderheyden, K.,
Anckaert, J., Lefever, S., Tay, A. P., de Bony, E. J.,
Trypsteen,W., Gysens,F., Vromman,M., Goovaerts,T.,
Hansen, T. B., Kuersten, S., Nijs, N., Taghon, T.,
Vermaelen, K., Bracke, K., Saeys, Y., Meyer, T.,
Deshpandeet,N., etal. (2021) TheRNA Atlas expands
the catalog of human non-coding RNAs, Nat. Biotech-
nol., 39, 1453-1465, https://doi.org/10.1038/S41587-
021-00936-1.
37. Karolchik, D., Hinrichs, A. S., Furey, T. S., Roskin,
K.M., Sugnet, C.W., Haussler,D., and Kent, W.J. (2004)
The UCSC table browser data retrieval tool, Nucleic
Acids Res., 32, D493-D496, https://doi.org/10.1093/
NAR/GKH103.
38. Quinlan, A.R., and Hall, I.M. (2010) BEDTools: Aflex-
ible suite of utilities for comparing genomic features,
Bioinformatics, 26, 841-842, https://doi.org/10.1093/
BIOINFORMATICS/BTQ033.
39. Mertes, C., Scheller, I. F., Yépez, V. A., Çelik, M. H.,
Liang, Y., Kremer, L. S., Gusic, M., Prokisch, H., and
Gagneur, J. (2022) Author Correction: Detection of
aberrant splicing events in RNA-seq data using
FRASER, Nat. Commun., 13, 3474, https://doi.org/
10.1038/S41467-022-31242-2.
40. Subramanian, A., Kuehn, H., Gould, J., Tamayo, P.,
and Mesirov, J. P. (2007) GSEA-P: A desktop applica-
tion for gene set enrichment analysis, Bioinformatics,
23, 3251-3253, https://doi.org/10.1093/bioinformatics/
btm369.
41. Wang, Y., Xie, Z., Kutschera, E., Adams, J. I., Kadash-
Edmondson, K. E., and Xing, Y. (2024) rMATS-turbo:
an efficient and flexible computational tool for alter-
native splicing analysis of large-scale RNA-seq data,
Nat. Protoc., 19, 1083-1104, https://doi.org/10.1038/
S41596-023-00944-2.
42. Lirussi,L., Demir,Ö., You, P., Sarno, A., Amaro, R. E.,
and Nilsen,H. (2021) RNA metabolism guided by RNA
modifications: the role of SMUG1 in rRNA quality
control, Biomolecules, 11, 76, https://doi.org/10.3390/
BIOM11010076.
43. Qiu, L., Jing, Q., Li, Y., and Han, J. (2023) RNA mod-
ification: mechanisms and therapeutic targets,
Mol. Biomed., 4, 25, https://doi.org/10.1186/S43556-
023-00139-X.
44. Darzacq, X., Jády, B. E., Verheggen, C., Kiss, A. M.,
Bertrand, E., and Kiss, T. (2002) Cajal body-specific
small nuclear RNAs: a novel class of 2′-O-methyla-
tion and pseudouridylation guide RNAs, EMBO J.,
21, 2746-2756, https://doi.org/10.1093/EMBOJ/
21.11.2746.
45. Nesic,D., Tanackovic,G., and Krämer,A. (2004) Arole
for Cajal bodies in the final steps of U2 snRNP bio-
genesis, J. Cell Sci., 117, 4423-4433, https://doi.org/
10.1242/JCS.01308.
46. Staněk, D. (2017) Cajal bodies and snRNPs – friends
with benefits, RNA Biol., 14, 671-679, https://doi.org/
10.1080/15476286.2016.1231359.
47. Novotný, I., Malinová, A., Stejskalová, E., Matějů, D.,
Klimešová, K., Roithová, A., Švéda, M., Knejzlík, Z.,
and Staněk,D. (2014) SART3-dependent accumulation
of incomplete spliceosomal snRNPs in Cajal bodies,
Cell Rep., 10, 429-440, https://doi.org/10.1016/j.celrep.
2014.12.030.
48. Staněk, D. (2023) Coilin and Cajal bodies, Nucleus,
14, 2256036, https://doi.org/10.1080/19491034.2023.
2256036.
49. Casamassimi,A., and Ciccodicola,A. (2019) Transcrip-
tional regulation: molecules, involved mechanisms,
and misregulation, Int.J. Mol. Sci., 20, 1281, https://
doi.org/10.3390/IJMS20061281.
50. Duchemin, A., O’Grady, T., Hanache, S., Mereau, A.,
Thiry, M., Wacheul, L., Michaux, C., Perpète, E.,
Hervouet, E., Peixoto, P., Ernst, F. G. M., Audic, Y.,
Dequiedt,F., Lafontaine, D.L.J., and Mottet,D. (2021)
DHX15-independent roles for TFIP11 in U6 snRNA
modification, U4/U6.U5 tri-snRNP assembly and
pre-mRNA splicing fidelity, Nat. Commun., 12, 6648,
https://doi.org/10.1038/s41467-021-26932-2.
51. Vazquez-Arango, P., and O’Reilly, D. (2018) Variant
snRNPs: new players within the spliceosome system,
RNA Biol., 15, 17-25, https://doi.org/10.1080/15476286.
2017.1373238.
52. Devany, E., Park, J. Y., Murphy, M. R., Zakusilo, G.,
Baquero, J., Zhang, X., Hoque, M., Tian, B., and
Kleiman, F. E. (2016) Intronic cleavage and polya-
denylation regulates gene expression during DNA
damage response through U1 snRNA, Cell Discov., 2,
16013, https://doi.org/10.1038/celldisc.2016.13.
53. Köhler, A., and Hurt, E. (2007) Exporting RNA from
the nucleus to the cytoplasm, Nat. Rev. Mol. Cell Biol.,
8, 761-773, https://doi.org/10.1038/nrm2255.
BOLIKHOVA et al.1756
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
54. Pánek, J., Roithová, A., Radivojević, N., Sýkora, M.,
Prusty, A. B., Huston, N., Wan, H., Pyle, A. M.,
Fischer, U., and Staněk, D. (2023) The SMN complex
drives structural changes in human snRNAs to en-
able snRNP assembly, Nat. Commun., 14, 6580, https://
doi.org/10.1038/s41467-023-42324-0.
55. Zieve, G. W. (1987) Cytoplasmic maturation of
the snRNAs, J. Cell Physiol., 131, 247-254, https://
doi.org/10.1002/JCP.1041310215.
56. Nizami, Z., Deryusheva, S., and Gall, J. G. (2010)
The Cajal body and histone locus body, Cold Spring
Harb. Perspect. Biol., 2, a000653, https://doi.org/
10.1101/CSHPERSPECT.A000653.
57. Geisler, M. S., Kemp, J. P., and Duronio, R. J. (2023)
Histone locus bodies: a paradigm for how nuclear
biomolecular condensates control cell cycle regu-
lated gene expression, Nucleus, 14, 2293604, https://
doi.org/10.1080/19491034.2023.2293604.
58. Mauer, J., Sindelar, M., Despic, V., Guez, T., Hawley,
B. R., Vasseur, J. J., Rentmeister, A., Gross, S. S.,
Pellizzoni, L., Debart, F., Goodarzi, H., and Jaffrey,
S.R. (2019) FTO controls reversible m6Am RNA meth-
ylation during snRNA biogenesis, Nat. Chem. Biol.,
15, 340-347, https://doi.org/10.1038/S41589-019-0231-8.
59. Karijolich, J., Kantartzis, A., and Yu, Y.T. (2010) RNA
modifications: a mechanism that modulates gene
expression, Methods Mol. Biol., 629, 1-19, https://
doi.org/10.1007/978-1-60761-657-3_1.
60. Verheggen, C., Lafontaine, D. L., Samarsky, D.,
Mouaikel, J., Blanchard, J. M., Bordonné, R., and
Bertrand,E. (2002) Mammalian and yeast U3 snoRNPs
are matured in specific and related nuclear com-
partments, EMBO J., 21, 2736-2745, https://doi.org/
10.1093/EMBOJ/21.11.2736.
61. Dragon, F., Gallagher, J. E., Compagnone-Post,
P. A., Mitchell, B. M., Porwancher, K. A., Wehner,
K. A., Wormsley, S., Settlage, R. E., Shabanowitz, J.,
Osheim, Y., Beyer, A. L., Hunt, D. F., and Baserga,
S. J. (2002) A large nucleolar U3 ribonucleoprotein
required for 18S ribosomal RNA biogenesis, Nature,
417, 967-970, https://doi.org/10.1038/nature00769.
62. Kass, S., Tyc, K., Steitz, J. A., and Sollner-Webb, B.
(1990) The U3 small nucleolar ribonucleoprotein
functions in the first step of preribosomal RNA pro-
cessing, Cell, 60, 897-908, https://doi.org/10.1016/
0092-8674(90)90338-F.
63. Lackmann,F., Belikov,S., Burlacu,E., Granneman,S.,
and Wieslander, L. (2018) Maturation of the 90S
pre-ribosome requires Mrd1 dependent U3 snoRNA
and 35S pre-rRNA structural rearrangements, Nucle-
ic Acids Res., 46, 3692-3706, https://doi.org/10.1093/
nar/GKY036.
64. Bowling, E. A., Wang, J. H., Gong, F., Wu, W., Neill,
N. J., Kim, I. S., Tyagi, S., Orellana, M., Kurley, S. J.,
Dominguez-Vidaña, R., Chung, H. C., Hsu, T. Y.,
Dubrulle, J., Saltzman, A. B., Li, H., Meena, J. K.,
Canlas, G. M., Chamakuri, S., Singh, S., Simon, L. M.,
Olson, C., Dobrolecki, L., Lewis, M., Zhang, B., Gold-
ing, I., Rosen, J. M., Young, D. W., Malovannaya, A.,
Stossi, F. Miles, G. et al. (2021) Spliceosome-targeted
therapies trigger an antiviral immune response in tri-
ple-negative breast cancer, Cell, 184, 384-403, https://
doi.org/10.1016/j.cell.2020.12.031.
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.