ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 6, pp. 650-670 © Pleiades Publishing, Ltd., 2025.
Published in Russian in Biokhimiya, 2025, Vol. 90, No. 6, pp. 695-719.
650
REVIEW
The Role of m
6
A-RNA Methylation
in the Development, Progression, and Treatment
Response of Bladder Cancer
Tatiana Sinyagovskaya
1,a
*, Yuliya Li
2
, Natalya Vinchevskaya-Khmelnitskaya
2
,
Aisha Agabalaeva
2
, Natalia Ponomareva
1
, Sergey Brezgin
1
, Irina Goptar
1
,
Vladimir Chulanov
3,4
, Alim Dymov
2
, Andrey Vinarov
2
, Dmitry Kostyushev
1,4,5
,
and Anastasiya Kostyusheva
1
1
Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical
and Vector-Borne Diseases, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
2
Institute for Urology and Reproductive Health,
Sechenov First Moscow State Medical University, 119991 Moscow, Russia
3
Department of Infectious Diseases, Diseases,
Sechenov First Moscow State Medical University, 119991 Moscow, Russia
4
Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
5
Faculty of Bioengineering and Bioinformatics,
Sechenov First Moscow State Medical University, 119991 Moscow, Russia
a
e-mail: tsv.relentless@gmail.com
Received December 11, 2024
Revised April 20, 2025
Accepted April 23, 2025
AbstractBladder cancer (BCa) remains a significant clinical challenge because of high recurrence rates
and variable response to immunotherapy and chemotherapy. Recent studies have highlighted the role of
N6-methyladenosine (m
6
A) modification in RNA in the regulation of various cellular processes, including tumor
progression and drug resistance. The review examines the impact of m
6
A methylation on BCa pathogenesis,
with a particular special focus on the role of m
6
A pathway factors and m
6
A-modified RNAs in tumorigene-
sis, proliferation, invasion, and migration of cancer cells. The mechanisms of m
6
A-mediated chemotherapy
resistance in BCa cells are discussed, including single nucleotide polymorphisms in m
6
A-associated patterns.
Significant advances in the high-throughput analysis of m
6
A methylation have enabled development of novel
m
6
A-based biomarkers for the risk assessment, early diagnostics, and prediction of relapse and treatment
response in BCa. The review outlines the prospects of the m
6
A-based molecular diagnostics in BCa.
DOI: 10.1134/S0006297924604441
Keywords: m
6
A, N6-methyladenosine, bladder cancer (BCa), biomarker, prognosis
* To whom correspondence should be addressed.
INTRODUCTION
Bladder cancer (BCa) remains the most common
malignancy of the urinary tract. According to the
World Health Organization (WHO) data for 2022, BCa
ranks 6th in prevalence among cancers in men, 9th –
among both sexes, and 13th – in global mortality rates
[1]. In the Russian Federation, it holds the 6th position
in prevalence among cancers in men, 9th – among
both sexes, and 16th – in mortality rates among all
oncological diseases [1].
BCa is categorized according to the depth of in-
vasion into the muscular layer and is subdivided into
non-muscle-invasive BCa (NMIBC) and muscle-invasive
BCa (MIBC). Approximately 75% of newly diagnosed
cases are NMIBC. The overall 5-year survival rate
for NMIBC exceeds 70%, but remains below 6% for
MIBC [2]. The ‘gold standard’ treatment for NMIBC
m
6
A-RNA METHYLATION IN BLADDER CANCER DEVELOPMENT 651
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
is transurethral resection of bladder tumor (TURBT),
followed by adjuvant intravesical chemotherapy or im-
munotherapy. MIBC requires more aggressive therapy,
such as trimodal treatment, which combines TURBT,
radiotherapy, and systemic chemotherapy. The recur-
rence rate for NMIBC varies widely, with the reported
rates between 40 and 90% [3-5]. The rate of NMIBC
progression to MIBC ranges from 5 to 55% [6].
One of the cancer biomarkers that has attracted
significant attention is N6-methyladenosine (m
6
A), a
dynamic and reversible epitranscriptomic RNA mod-
ification involved in various cellular processes. m
6
A
methylation occurs exclusively within the context-
specific sequences known as DRA*CH motifs (where
D  =  A, G, or U; R  =  A or G; H  =  A, C, or U). A unique
distribution and density of m
6
A in RNA molecules is
typically referred to as an m
6
A pattern. The distribu-
tion of m
6
A patterns across RNA transcripts is non-
random; they are highly conserved but can vary de-
pending on the RNA type and cellular context, tissue,
developmental stage, and disease. m
6
A patterns can
be assessed within individual RNA transcripts or in
the entire transcriptome. DRA*CH and alternative m
6
A
consensus motifs are typically evolutionary conserved
and appear mostly near 3′- and 5′-untranslated regions
(UTRs), whereas their presence in coding regions is
less frequent [7]. m
6
A methylation can occur in spe-
cific mRNAs or non-coding RNAs (ncRNAs), affecting
their stability, splicing, translation, and decay. The
presence, location, and density of m
6
A modifications
in individual transcripts can determine their fate and
functions. Together, these factors influence a complex
network of m
6
A-mediated regulation, which impacts
all aspects of RNA metabolism.
Abnormal m
6
A methylation patterns have been
observed in various pathological conditions, such as
neurodevelopmental and neurological disorders [8, 9],
cardiovascular diseases [10,  11], viral infections [12],
metabolic disorders [13], and immune response [14].
Altered m
6
A methylation patterns have been observed
and extensively studied in various cancers, including
breast, lung, liver, and colorectal cancers, as it was
found that specific m
6
A modifications correlate with
tumor progression, metastasis, and patient progno-
sis [15].
m
6
A modifications have been identified as a sig-
nificant factor in cancer biology, affecting various as-
pects of tumorigenesis and cancer progression. m
6
A
methylation of mRNA serves as a gene regulatory
mechanism and influences proliferation and inva-
sion of tumor cell, as well as tumor immune evasion
and metastasis, by coordinating and modulating gene
expression in a reversible and highly dynamic man-
ner [16].
The review discusses the role of m
6
A methylation
in the development of BCa, the pro- and anti-oncogenic
functions of m
6
A pathway factors, m
6
A methylation of
mRNAs and ncRNAs related to oncogenes and tumor
suppressors, and their potential diagnostic and prog-
nostic significance. We also explored how m
6
A meth-
ylation contributes to the development of resistance
to chemotherapeutic agents.
CURRENT CHALLENGES IN THE DIAGNOSTICS
AND TREATMENT OF NMIBC
At present, BCa diagnosis is confirmed exclusive-
ly by histological examination of biopsies obtained
during the TURBT. Urine cytology is used to detect ex-
foliated tumor cells. This method is highly sensitive in
the detection of carcinoma in situ and high-grade (G3)
tumors, but is much less efficient in the identification
of low-grade (G1) tumors [17]. Thus, the sensitivity of
urine cytology test in detecting carcinoma in situ var-
ies widely, ranging from 28 to 100% [18]. The positive
result indicates the presence of transitional cell carci-
noma, which can originate in any part of the urinary
tract, but the negative result does not rule out the
possibility of BCa [19].
Due to the low sensitivity of urine cytology in
the diagnostics of low-grade BCa, numerous methods
for analyzing BCa molecular markers have been de-
veloped. However, none of these markers have been
widely adopted as a standard with a high prognostic
value [20].
Currently, biomarkers can be categorized into two
distinct groups: (i)  diagnostic markers, which help con-
firm a BCa diagnosis; (ii)  prognostic markers, which
assess the risks of disease progression and recurrence
in patients already diagnosed with BCa [21].
Five diagnostic test systems have been approved
by the FDA: NMP22 test kit, NMP22 BladderChek Test,
BTA TRAK, BTA stat, and UroVysion [22,  23]. Despite
the established efficacy of these test systems in the
identification of BCa markers, their application in clin-
ical practice remains limited [24].
At present, there are no urinary BCa biomarkers
that can substitute for cystoscopy or reduce the fre-
quency of cystoscopic examinations [20], indicating an
ancillary role of non-invasive urine analysis methods
conducted prior to definitive diagnosis.
A standard of treatment for NMIBC is transure-
thral bladder resection followed by adjuvant chemo-
or immunotherapy. The two techniques for bladder
resection are TURBT (resection of the bladder wall
with the tumor as a single block) or piecemeal tumor
resection [19].
Current clinical guidelines recommend a single
instillation of a chemotherapeutic agent (most com-
monly, mitomycin  C, epirubicin, or pirarubicin) after
TURBT, which has been proven to significantly reduce
SINYAGOVSKAYA et al.652
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
the recurrence rates. A comprehensive review by Syl-
vester et al. [25] reported that this treatment lowers
the five-year recurrence rate from 59 to 45%. Such
improvement in the outcome (14%) means that ap-
proximately one in seven patients will benefit from
the therapy, while the remaining six will be unaffect-
ed but still exposed to its potential side effects and
risks. Hence, a single instillation of a chemotherapeu-
tic agent cannot be considered a definitive solution,
thus emphasizing the need for continued research
aimed to further reduce the risk of tumor recurrence
and metastasis.
After obtaining histological results and determin-
ing the degree of tumor differentiation, patients are
stratified into four or three risk groups [19]. Adjuvant
therapy is not recommended for patients in the low-
risk group. Patients with the intermediate-, high-, or
very high-risk NMIBC typically undergo induction
and maintenance courses of intravesical BCG (Bacil-
lus Calmette–Guérin) therapy lasting from one to
three years [26]. Intravesical chemotherapy may be
recommended for intermediate-risk patients with in-
tolerance or contraindications to the BCG therapy [19].
Despite a high efficacy of current treatment op-
tions secured in clinical guidelines, the recurrence
rate of NMIBC remains significant (from 40 to 90%)
[3-5], and the rate of NMIBC progression to MIBC var-
ies between 5 to 55% [6], which emphasizes the ongo-
ing need for new approaches to BCa therapy.
Such high recurrence and progression rates re-
quire a deeper understanding of molecular mech-
anisms driving BCa development. Early detection is
critical for improving the clinical outcomes, which is
why the development of reliable biomarkers capable
of improving the diagnostic and prognostic accuracy
and guiding the treatment decisions, has become a
major research focus.
In summary, the challenges associated with BCa
are multifaceted and include (1)  early BCa diagnosis
and risk assessment; (2)  evaluation of risk of NMIBC
progression to MIBC; (3)  prediction of NMIBC recur-
rence; and (4)  classification of patients into respond-
ers and non-responders to current anti-cancer regi-
mens. While some of these challenges can be partially
addressed through existing methods, such as imaging,
cytological analysis, and genomic mutation profiling,
there is an urgent need to identify novel molecular
biomarkers to enhance the diagnostic accuracy and
treatment efficacy in BCa. One of the promising re-
search areas is the use of m
6
A modification as a po-
tential biomarker.
Molecular mechanism of m
6
A-methylation. m
6
A
is the most prevalent epitranscriptomic modification
of both mRNAs and ncRNAs, including lncRNAs (long
non-coding RNAs), circRNAs (circular RNAs), miRNAs
(microRNAs), and others. m
6
A methylation is catalyzed
by the methyltransferase complex (or ‘writer’), con-
sisting of the METTL3/METTL14 (methyltransferase
like proteins  3 and 14) heterodimer  [27] and aux-
iliary components, such as WTAP (Wilms tumor  1
associated protein), VIRMA (KIAA1429; vir like m
6
A
methyltransferase associated), RBM15/15B (RNA bind-
ing motif protein 15/15B), ZC3H13 (zinc finger CCCH-
type containing  13), and others, which lack the meth-
yltransferase activity and play a regulatory role in
the complex functioning  [28]. Methyl group can be re-
moved by demethylases (or ‘erasers’), such as ALKBH5
(AlkB homolog  5)  [29] and FTO (fat mass and obesi-
ty-associated protein)  [30], indicating the reversible
and highly dynamic nature of the m
6
A modification.
m
6
A-binding proteins, known as ‘readers’, recog-
nize the m
6
A mark and determine the fate (stabiliza-
tion/translation or degradation) of the RNA molecule.
Reader proteins belong to various protein fami-
lies and include YTH-domain proteins (YTHDF1/2/3,
YTHDC1/2)  [31], heterogeneous nuclear ribonucleo-
proteins (HNRNPC, HNRNPG, HNRNPA2B1)  [32], in-
sulin-like growth factor  2 mRNA-binding proteins
(IGF2BP1/2/3)  [33], and eukaryotic initiation factors
(elF3)  [34]. The specificity of reader proteins is de-
termined by a combination of biochemical, spatial,
and contextual factor that ultimately make the deter-
mination of RNA fate based on m
6
A marks a highly
context-dependent process. Understanding this mech-
anism is important for the accurate prediction of the
outcome of downstream processes, which could shed
light on aberrant m
6
A functions leading to various
diseases.
m
6
A modification can influence expression of
both oncogenes and tumor suppressor genes, playing
a dual role in cancer progression, which emphasizes
its importance in a complex network of molecular in-
teractions that sustain tumor growth and determine its
resistance to therapy. m
6
A modifications can promote
accumulation of oncogenic proteins by stabilizing on-
cogenic mRNAs and inhibiting decay of tumor-promot-
ing mRNAs, which can result in the dysregulation of
cell proliferation, promotion of cell cycle progression,
inhibition of apoptosis, and other cancer hallmarks
[35]. Some studies indicate that m
6
A modifications can
significantly impact the metastatic potential of cancer
cells by regulating key genes involved in cell adhesion,
migration, and invasion, thus contributing to tumor
spreading. Conversely, m
6
A modifications can ensure
the stability of tumor suppressor mRNAs by prevent-
ing their degradation, maintaining their inhibitory
effect on cancer cells [36, 37].
m
6
A pathway in BCa. m
6
A modifications modu-
late expression of genes associated with cell prolifer-
ation, apoptosis, and metastasis [15]. m
6
A methylation
also plays a critical role in the progression of BCa [38].
Alterations in the m
6
A methylation patterns in BCa
m
6
A-RNA METHYLATION IN BLADDER CANCER DEVELOPMENT 653
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
can lead to the upregulation of oncogenes or down-
regulation of tumor suppressor genes, thus contribut-
ing to tumorigenesis [38]. Expression of m
6
A-pathway
factors in BCa and healthy tissues have been investi-
gated in several transcriptomic studies. For instance,
Chen et al. [39] studied the levels of key m
6
A reg-
ulators and analyzed correlation of their expression
with clinicopathological variables. Clinicopathological
information for 408  BCa patients and healthy donors
was obtained from the TCGA (The Cancer Genome At-
las) database, allowing for the comparison between
the non-tumor tissue group and BCa samples of vary-
ing grades and stages. It was shown that KIAA1429
(one of the major m
6
A methyltransferases) and YTHDF
(m
6
A reader protein) were highly expressed in high-
grade BCa. Expression of ALKBH5 (m
6
A demethylase)
positively correlated with the tumor grade and M1
stage. Although METTL3 (another major m
6
A methyl-
transferase) was highly expressed in cancer tissues,
its expression decreased as the grade increased and
was low in high-grade tumors. FTO (an important
m
6
A demethylase) was expressed poorly in BCa  [39].
Bioinformatic analysis revealed differential patterns
in the expression of m
6
A-related genes across tu-
mor tissues. Specifically, expression of FTO, ZC3H13,
YTHDF3, YTHDC1, WTAP, METTL16, and METTL14
was downregulated in cancerous tissues compared
to normal tissues. Conversely, HNRNPA2B1, IGF2BP1,
IGF2BP3, METTL3, YTHDF2, and YTHDF1 demon-
strated high expression levels. Moreover, expression
of HNRNPA2B1, HNRNPC, IGF2BP2, RBM15, YTHDF1,
and YTHDF2 was associated with the advanced clin-
ical stages of BCa [40]. Alterations in the expression
of m
6
A factors indicate a critical role of m
6
A methyl-
ation in BCa development. Therefore, both expression
of m
6
A-related factors and m
6
A-methylation patterns
could potentially serve as prognostic markers to help
clinicians define the clinical stage of cancer, differ-
entiate cancer subtypes, predict disease progression
and outcome, evaluate the risk of recurrence, and
design the treatment plans accordingly. For instance,
abnormal m
6
A levels in transcripts associated with
tumor suppression may correlate with the advanced
clinical stages, while changes in the methylation levels
of oncogenic transcripts can indicate aggressive can-
cer subtypes or higher risk of disease progression.
By analyzing the m
6
A methylation status of key tran-
scripts, clinicians can more accurately define the
clinical stage of BCa, differentiate between the lumi-
nal and basal subtypes, predict the likelihood of dis-
ease progression, and estimate the outcomes. Specific
m
6
A methylation signatures could help evaluate the
risk of recurrence and guide personalized treatment
plans, e.g., help in selecting patients who may benefit
from immunotherapy or targeted therapy. While this
approach is still in the realm of clinical theory, ad-
vancements in RNA epigenomics and single-molecule
sequencing technologies are paving the way for its
practical application in clinical practice in the fore-
seeable future.
m
6
A factors with the oncogenic role in BCa.
Inrecent years, the m
6
A demethylase FTO has gained
significant attention due to its potential role in vari-
ous cancers, including BCa. Several targets of FTO in
BCa have been identified, including PTPN6 (tyrosine
protein phosphatase non-receptor type  6), a non-re-
ceptor tyrosine phosphatase that dephosphorylates
and regulates the activity of numerous proteins in-
volved in signal transduction, cell growth, differenti-
ation, cell cycle, and oncogenic transformation [41].
PTPN6 is aberrantly expressed in various cancers,
such as hepatocellular carcinoma, renal cell carcino-
ma, gastric cancer, and BCa [41]. Wu et  al.  [42] dis-
covered that FTO induces expression of PTPN6 and
stabilizes it, thus promoting proliferation and meta-
static capabilities of BCa cells. The authors analyzed
human BCa and adjacent normal tissues collected
from 20 patients diagnosed with BCa (sex and age of
the patients were not taken into account), as well as
investigated human BCa cell lines (T24, 5637, RT4, J82,
and HTT1378) and normal bladder epithelial cell line
SV-HUC-1 in in vitro experiments The samples were
not stratified into groups based on the disease stage or
tumor grade, limiting the generalizability of the study.
While m
6
A-modified PTPN6 mRNA was detected, no
specific m
6
A sites were mapped, which hinders the
understanding of how FTO regulates this transcript.
Despite the limitations, the findings of this study sug-
gest that the FTO-PTPN6 axis may serve as a potential
prognostic marker in BCa.
Metastasis-associated lung adenocarcinoma tran-
script  1 (MALAT1), an lncRNA involved in the regu-
lation of gene expression through epigenetic mecha-
nisms, has been associated with the tumorigenesis in
most cancer types, including BCa [43]. It was found
that in BCa, FTO acts as an oncogene by reducing the
m
6
A methylation of the 5′-UTR of MALAT1, leading
to the increased stability and upregulated expression
of this ncRNA. Elevated MALAT1 then functions as a
sponge for miR-384, reducing its availability, which
increases MAL2 expression and promotes cancer cell
viability and tumor growth. This study was conduct-
ed in human BCa paired tissue samples (tumor and
adjacent noncancerous) collected from 25 patients;
144 cancer tissue microarrays were used for the val-
idation. Human BCa cell lines (5637, J82, 253J, T24,
SCABER) and SV-HUC-1 cells were investigated in the
in vitro experiments. The authors did not stratify pa-
tients by a specific BCa type or distinct m
6
A modifica-
tion profile; tissue samples were analyzed primarily
for the overall pathologic stage (pTa-T1 vs. pT2-T4). Up-
regulated FTO expression was found to be associated
SINYAGOVSKAYA et al.654
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
with more advanced pathologic stages (pT2-T4), while
no statistically significant association between the FTO
or MAL2 levels and patients’ sex or age was observed.
FTO has been shown to modify the maturation
of pri-miR-576 in an m
6
A-dependent manner and to
promote proliferation, migration, and invasion of
BCa cells by regulating the miR-576-CDK6 pathway
[44]. The authors examined BCa and adjacent nor-
mal tissues from 20 patients who underwent radical
cystectomy. BCa cell lines (T24, 5637, UM-UC-3) and
SV-HUC cells were used in in vitro experiments. Tis-
sue microarrays from 67 BCa patients were analyzed.
It was found that FTO expression did not differ sig-
nificantly between males and females or different age
groups (p =  0.755 for sex, p =  0.995 for age), but was
associated with a higher TNM stage (p =  0.035). The
authors provided detailed insights into the m
6
A mod-
ification-based mechanisms by which FTO regulates
the miR-576-CDK6 axis and contributes to BCa pro-
gression. By identifying FTO as a potential prognos-
tic biomarker, the study facilitates future diagnostic
or therapeutic advancements in BCa. The correlation
between the FTO expression and higher TNM stages
suggests a potential use of FTO for predicting cancer
progression.
Another key m
6
A-related regulator is the reader
protein IGF2BP3 (insulin like growth factor 2 mRNA
binding protein  3) that may play an oncogenic role
in BCa progression. Expression of IGF2BP3 was high-
er in BCa cells compared to adjacent healthy tissues
[45] and was associated with several factors indicat-
ing more advanced stages of BCa: (1)  advanced tu-
mor characteristics and (2)  more aggressive disease
progression [45]. BCa samples were collected from
95 patients who underwent resection; SV-HUC-1 cells
and BCa cell lines (5637, J82, UMUC3, and T24) were
used for in vitro analysis. The study included the data
for 414 BCa patients from the TCGA, categorized by
the IGF2BP3 expression levels (high and low). The pa-
tients were classified as ≤70 years vs. >70 years old,
but no significant correlation between the IGF2BP3
expression and age was found (p =  0.372). The ele-
vated IGF2BP3 expression levels correlated with poor
overall survival (p =  0.015) and were associated with
more advanced tumor stages (T3/T4) and high tumor
histologic grade, reinforcing the role of IGF2BP3 as
a marker of BCa progression. However, no correla-
tion between the IGF2BP3 expression and nodal (N)
or distant metastasis (M) status was observed, which
was unexpected given the role of IGF2BP3 in cell
migration and invasion. This discrepancy suggests
that IGF2BP3 may be more involved in the local
tumor progression rather than in metastasis. The
study highlighted a strong correlation between the
IGF2BP3 expression and immune cell infiltration in
BCa. High IGF2BP3 levels were associated with an
increased infiltration of macrophages, neutrophils,
and CD8
+
T  cells, suggesting a potential immuno-
modulatory role of IGF2BP3. Moreover, the elevated
IGF2BP3 levels have been linked to poorer outcomes
for BCa patients, suggesting that IGF2BP3 may partic-
ipate in determining the severity of BCa and influ-
ence patients’ survival. In a recent study  [33], it was
suggested that IGF2BP family proteins can recognize
m
6
A in the transcripts and stabilize such transcripts,
thus increasing their half-life time and duration of
protein expression. For example, stabilization of the
neuropilin1 (NRP1) mRNA led to the M2 macrophage
polarization associated with BCa progression
[46].
High-mobility group protein  B1 (HMGB1) was
identified as a target of IGF2BP3. HMGB1 is a nucle-
ar DNA-binding protein integral to numerous cellular
processes, including inflammation, cell differentia-
tion, and apoptosis. Interestingly, the levels of IGF2BP3
were notably elevated in the individuals who showed
positive response to immunotherapy in contrast to
those who did not respond. Moreover, increased ex-
pression of IGF2BP3 and its m
6
A-mediated interaction
with HMGB1 mRNA correlated with the improved
overall survival, suggesting that IGF2BP3 may im-
pact the immune microenvironment of BCa, thus in-
fluencing the efficacy of immunotherapy [47]. Using
bioinformatic methods, the authors identified multi-
ple m
6
A motifs in HMGB1 mRNA that could serve as
IGF2BP3-binding sites. IGF2BP3 expression and func-
tion was investigated in BCa cell lines RT112/84 and
BFTC905.
Another gene of significant interest is ITGA6,
which encodes integrin alpha-6 subunit. ITGA6 is a
cell surface receptor involved in the cell–cell and cell–
substrate interaction, cell migration, differentiation,
tissue repair, and regulation of cell growth [48,  49].
The association between the ITGA6 expression and
cancer progression is rather complex. The upregula-
tion of ITGA6 has been linked to the enhanced inva-
sion, metastasis, and poor prognosis in various can-
cers [50-52]. Thus, ITGA6 was found to maintain cell
adhesion-mediated drug resistance in ovarian cancer
and leukemia [53, 54].
Recently, a mechanism of ITGA6 regulation by the
m
6
A modification has been proposed [55]. Tissue sam-
ples from of 186 BCa patients who underwent radical
cystectomy and bladder biopsies were investigated for
m
6
A methylation. It was confirmed that the 3′-UTR
of ITGA6 mRNA contains four m
6
A motifs methylat-
ed by METTL3. Methylation promoted translation of
the ITGA6 transcript through binding of the YTHDF1
and YTHDF3 reader proteins in vitro and in vivo.
Interestingly, mutation of a single m
6
A site did not
decrease the translation rate of the transcript, whereas
introduction of several mutations into multiple m
6
A
sites noticeably slowed it down [55]. Inanother study,
m
6
A-RNA METHYLATION IN BLADDER CANCER DEVELOPMENT 655
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
removal of m
6
A from several positions in the ITGA6
transcript by the designed multisite editor dCasRx-m
6
A
led to the reduction in the transcript translation and
inhibition of BCa progression in vitro and in vivo,
suggesting a significance of cooperative transcript reg-
ulation by multiple m
6
A sites. The off-target activity of
the dCasRx–m
6
A editor has been assessed and proven
to be limited [56]. These findings highlight a potential
importance of m
6
A methylation of the ITGA6 mRNA
in tumorigenesis and suggest that further research
in this area could lead to the identification of novel
therapeutic targets for BCa.
The CUB domain-containing protein 1 (CDCP1) is a
transmembrane protein that acts as a protein-protein
interaction hub for proteins regulating cell–cell and
cell–substrate adhesion through proteolytic process-
ing and tyrosine phosphorylation  [57]. Dysregulation
of CDCP1 expression in cancer cells has been observed
in colon [58,  59], lung  [60], and kidney  [61] cancers.
CDCP1 was found to induce cell detachment and
promote metastasis in breast cancer  [57]. In general,
an elevated expression of CDCP1 is associated with
the enhanced migratory capabilities of cancer cells,
which facilitates tumor dissemination and metastasis
and leads to poor disease outcome  [62]. The oncogenic
role of CDCP1 in BCa, including promotion of cancer
development and progression, has been demonstrated
in several studies.
The METTL3-m
6
A-CDCP1 axis was also found to
be implicated in BCa oncogenesis, suggesting a regula-
tory role of m
6
A modification in the CDCP1 expression
and translation in aberrant uroepithelial cells [63].
The authors utilized 114 BCa and 30 cystitis samples
from patients who underwent radical cystectomy and
bladder biopsies and identified specific m
6
A modifi-
cation sites in the 3′-UTR and coding region of the
CDCP1 mRNA, which were observed in both control
and malignant BCa cells, whereas CDCP1 expression
itself was barely detectable in cystitis tissue. Methyl-
transferase METTL3 facilitated methylation at these
sites, which served as a signal for the YTHDF1 reader
protein to bind to the CDCP1 transcript, thus stabiliz-
ing it and promoting its translation. Conversely, de-
pletion of METTL3 moderately inhibited proliferation,
invasion, and migration of BCa malignant cells [63].
Another research confirmed these data by demon-
strating that the targeted RNA methylation system
RCas9-METTL3 introduced m
6
A modification to the
3′-UTR of the CDCP1 transcript, leading to its enhanced
translation and promotion of tumor growth in vitro
and in vivo. The system used exhibited satisfactory
on-target activity [64].
As key factors in cell adhesion, ITGA6 and CDCP1
are linked to cancer progression, drug resistance, and
poor prognosis in BCa [55, 63]. Both are regulated by
the METTL3-mediated m
6
A modification, which sug-
gests a common regulatory axis for potential thera-
peutic targeting. While these findings are promising,
several challenges related to the specificity, resistance,
and clinical implications must be addressed. Although
the analyzed samples were from post-radical cystecto-
my BCa patients, the data on the comorbidities were
lacking and no stratification based on the tumor stage,
grade, or molecular subtype was performed, raising
concerns about generalizability and interpretation of
the study results. The possibility of using ITGA6-CDCP1
and related m
6
A factors (e.g., METTL3) as therapeu-
tic targets is questionable, since their inhibition may
disrupt the downstream pathways, potentially, even in
healthy cells. Utilizing m
6
A sites as direct prognostic
markers is a novel approach [65-67]. The use of ad-
vanced molecular tools has demonstrated a potential
for precise RNA editing and its therapeutic applica-
tions (for instance, removal of aberrant modifications
to nullify the negative effects of said modifications).
However, challenges remain, such as predicting long-
term effects of RNA editing and ensuring safe and ef-
ficient delivery of editing systems via nanoparticles,
exosomes, or viral vectors, [68-70]. The cooperative
regulation of ITGA6 by multiple m
6
A sites compli-
cates the development of the targeted therapy. Ab-
errant m
6
A profiles correlating with the tumor stage
and grade, could serve as prognostic markers for dis-
ease progression, help stratify patients into high- and
low-risk groups, and guide the treatment decisions.
The studies have provided mechanistic insights into
the regulation of ITGA6 and CDCP1 by m
6
A modifica-
tions, linking RNA methylation to cancer progression.
Development of targeted tools, e.g., molecular editors,
offers exciting possibilities for precision medicine, but
further research is needed to validate these findings
and translate them into effective therapies for BCa
patients.
m
6
A factors acting as tumor suppressors inBCa.
The effects of m
6
A modifications are diverse and may
include inhibition of tumorigenesis. NOTCH1 (Notch
receptor  1), a component of the Notch pathway, is
responsible for the self-renewal of tumor-inducing
cells (TICs) and oncogenesis [71]. Primary BCa sam-
ples (ranging from early non-invasive to advanced
invasive BCa) from 6 patients have been studied in
order to confirm the role of the NOTCH1-m
6
A axis in
tumorigenesis. It was found that the m
6
A modification
of the NOTCH1 transcript by METTL14 attenuated its
expression [71], but the exact mechanism of this pro-
cess remains largely unknown and requires further
research. These findings revealed a novel METTL14-
m
6
A-Notch1 regulatory axis in BCa, highlighting a
possible implementation of the m
6
A modification as
a therapeutic target and biomarker of BCa progres-
sion. However, the study used only six BCa samples
for the initial analysis of the m
6
A content. Such small
SINYAGOVSKAYA et al.656
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
sample size limited the generalizability of the findings;
moreover, the samples varied significantly in terms
of age, sex, tumor size, stage, and metastasis status.
Also, no tissues from healthy controls were analyzed,
which could potentially affect the results of m
6
A map-
ping.
m
6
A may also act as an mRNA decay signal, as
it was shown for the tumor-suppressor factors SETD7
(SET domain containing 7), KLF4 (Krüppel-like fac-
tor 4), and SYTL1 (synaptotagmin-like protein 1). The
roles of SETD7 and KLF4 in cancer still remain con-
tradictory. Downregulation of SETD7 (lysine methyl-
transferase involved in histone and non-histone pro-
tein methylation) has been linked to the enhanced
migration and invasion of lung cancer cells [72] and
to correlate with a poor outcome in patients with
gastric cancer [73]. A large body of evidence suggests
that SETD7 acts as an oncogene, based on the positive
correlation between its expression and cancer stage
in hepatocellular adenocarcinoma [74]. Conversely,
SETD7 has been associated with the prevention of
the epithelial-to-mesenchymal transition in various
cancer types, indicating its potential involvement in
the inhibition of metastasis [75]. KLF4 was found to
act as a tumor suppressor by preventing metastasis
in colorectal cancer [76]), but also as an oncogene in
osteosarcoma by promoting tumorigenesis invivo [77].
KLF4 is significantly downregulated in urothelial BCa
cells, indicating poor overall survival and risk for re-
currence [78, 79]. KLF4 overexpression induced by the
CRISPR-ON transcriptional activation system inhibited
cell proliferation and promoted cell cycle arrest in G1
phase via regulation of the AKT-p21 signaling path-
way [79]. The data obtained suggest that SETD7 and
KLF4 mRNAs are the downstream targets of METTL3,
which installs m
6
A in their transcripts. The modified
transcripts are recognized by the YTHDF2 reader pro-
tein, resulting in mRNA degradation. Repression of
METTL3 correlated with the cell cycle arrest in G1
phase and reduction in cell proliferation and migra-
tion invitro [80].
KLF4-m
6
A and SETD7-m
6
A axes represent prom-
ising but complex targets for cancer therapy. While
the contradictory roles of SETD7 and KLF4 as both
tumor suppressors and oncogenes in different cancers
pose serious challenges, advancements in understand-
ing molecular mechanisms underlying their activity
could lead to novel therapeutic and prognostic strate-
gies. Future research should focus on resolving these
contradictions, identifying context-specific factors, and
developing targeted interventions with minimal tox-
ic effects. However, the implication of the aforemen-
tioned axes yields a prognostic value, since aberrant
m
6
A profiles, as well as SETD7 and KLF4 expression
levels, may correspond to specific tumor stages and
grades.
SYTL1 plays a critical role in enhancing the an-
ti-tumor immune response. Thus, the overexpression
of SYTL1 was found to activate natural killer (NK)
cells, promoting the effect of the anti-tumor immunity
both in vitro and in vivo. [81] The m
6
A methyltrans-
ferase WTAP downregulates SYTL1 expression via m
6
A
methylation of the SYTL1 mRNA, which marks it for
degradation after recognition by the YTHDF2 reader
protein. The authors analyzed 35 BCa and adjacent
tissue samples collected from patients who underwent
radical cystectomy. BCa cell lines (5637, T24, SW780,
J82), SV-HUC-1 cells, and NK-92 cells were used in the
in vitro experiments, and the results of these experi-
ments were validated in vivo. No sex, age, or disease
stage were taken into account in this work, as the
study was primarily focused on BCa samples and
cell lines. The authors did not investigate the SYTL1
expression in healthy individuals and patients with
other diseases or comorbidities. However, analysis
of bioinformatics databases allowed them to identify
broader expression patterns, in particular, reduction
in the SYTL1 expression in BCa cells compared to ad-
jacent normal tissues. No changes in the SYTL1 ex-
pression over time or in response to treatment were
studied. Although the authors suggested that modu-
lation of SYTL1 expression could enhance the tumor
suppression mediated by NK cells, they did not pro-
vide therapeutic strategies for clinical use.
Ferroptosis and m
6
A modifications in BCa. Fer-
roptosis, a form of regulated cell death characterized
by iron-dependent lipid peroxidation, has been in-
creasingly recognized for its role in various patholog-
ical conditions. Ferroptosis is driven by peroxidation
of phospholipids in cellular membranes, resulting in
oxidative stress and accumulation of lipid peroxides
followed by membrane damage and cell lysis [82]. Cell
death occurring exclusively by ferroptosis correlates
with the accumulation of lipid peroxidation markers
and can be suppressed by iron chelators, lipophilic
antioxidants, inhibitors of lipid peroxidation, and de-
pletion of polyunsaturated fatty acids (PUFAs).
Ferroptosis play a particularly important role in
BCa due to the unique metabolic and oxidative en-
vironment of tumor cells. Several ferroptosis induc-
ers, such as erastin, artemisinin, conjugated polymer
nanoparticles, and quinazolinyl-arylurea derivatives,
have been shown to sensitize BCa cells to anti-cancer
treatment [83]. A combination of these compounds
with standard anticancer drugs, along with the ferro-
ptosis-related m
6
A factors and genes, could overcome
the resistance to therapy by targeting ferroptosis-relat-
ed vulnerabilities of BCa cells. This approach, which
leverages both conventional and ferroptosis-inducing
agents, presents a promising direction in improving
the efficacy of treatment, especially in patients with
therapy-resistant BCa.
m
6
A-RNA METHYLATION IN BLADDER CANCER DEVELOPMENT 657
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Ferroptosis is regulated by various pathways
and factors, including NRF2 (nuclear factor eryth-
roid  2- related factor  2), a transcription factor that
plays an essential role in cellular defense against
oxidative stress [84]. NRF2 regulates expression of
antioxidant proteins, such as solute carrier family  7
member 11 (SLC7A11)  [85] and glutathione peroxi-
dase  4 (GPX4) [86], that can prevent ferroptosis. Con-
versely, NRF2 has been shown to sensitize cancer cells
to ferroptosis by upregulating genes involved in iron
metabolism and redox homeostasis, e.g., the gene for
the multidrug resistance-associated protein  1 (ABCC1-
MRP1) [87]. Ferroptosis can be a mechanism of cell
death in tumors. Tumor cells, which often contain
high levels of reactive oxygen species (ROS), can un-
dergo ferroptosis due to the iron reaction with exces-
sive hydrogen peroxide, leading to the production of
hydroxyl radicals [88]. For example, activation of the
p62-Keap1-NRF2 pathway promoted resistance to fer-
roptosis in hepatocellular carcinoma cells, highlighting
a complex regulatory mechanism involved in the cell
death pathways in cancer [89]. In BCa, the NRF2-de-
pendent antioxidant response promotes tumor growth
through the p62-KEAP1-NRF2 pathway [90]. A num-
ber of m
6
A-associated ferroptosis-related genes (FRGs)
linked to BCa have been identified. Thus, myosin bind-
ing protein H (MYBPH), sclerostin (SOST), small pro-
line-rich protein 2A (SPRR2A), and cornulin (CRNN)
were suggested as potential oncogenes, while CYP4F8
(cytochrome P450 family  4 subfamily  F member  8),
PDZD3 (PDZ domain containing  3), CRTAC1 (cartilage
acidic protein  1), and LRTM1 (leucine rich repeats and
transmembrane domains  1) were proposed as tumor
suppressors. The mechanisms of interaction between
these proteins and NRF2 remain largely unknown
[91]. An aberrant expression of NRF2 in BCa is medi-
ated in an m
6
A-related manner, resulting in resistance
to ferroptosis. WTAP introduces m
6
A modifications
in the 3′-UTR of the NRF2 mRNA, making it a target
for YTHDF1, which stabilizes this transcript and pro-
motes its translation [92]. This process is associated
with the accelerated cell proliferation and repression
of the erastin-induced ferroptosis, thus ensuring poor
disease prognosis. The authors used BCa and adjacent
non-cancerous tissues from 45 paired samples ob-
tained from patients who underwent nephrectomy, as
well BCa cell lines (J82, UM-UC-3) and SV-HUC-1 cells
(control). Neither sex or age of patients, nor the ex-
pression of WTAP or NRF2 in healthy individuals were
taken into account in this study The authors focused
on the mechanistic aspects of m
6
A modification and
did not explore potential therapeutic interventions
targeting WTAP or NRF2 in vivo.
As reported in [93], WTAP is upregulated in BCa
cells and its high expression correlates with a poor
disease prognosis. The study included 48 males and
14 females in the BCa group and 14 males and 6 fe-
males in the control group. The average age in the
control group was 57  ±  19 years vs. 52  ±  13 years in
the BCa group. Statistical comparisons indicated no
significant differences in sex and age between the
groups (p >  0.05), suggesting that sex and age were not
confounding factors in the analysis. Normal bladder
mucosa tissue was used as a control. The authors did
not differentiate between the BCa molecular subtypes,
which made it unclear whether the expression of
WTAP varied between them, neither did they consid-
er potential confounding variables, such as smoking
status, comorbidities, or genetic predisposition, all of
which could influence WTAP expression. The down-
stream targets modified by WTAP and the potential of
specific m
6
A sites to serve as independent prognostic
markers were not discussed as well. Complementary
evidence comes from study [92], which elucidates the
WTAP-induced m
6
A-modification of NRF2 and links
this axis to ferroptosis resistance and adverse out-
come.
m
6
A methylation in BCa-related ncRNAs. Vari-
ous ncRNAs can also undergo m
6
A methylation. Only
around a third of transcribed genes in the human ge-
nome encode proteins, while the remaining two-thirds
are non-coding genes. Some genes are transcribed into
lncRNAs that regulate transcriptional and post-tran-
scriptional processes by modulating gene expression
via affecting chromatin remodeling. lncRNAs can
serve as signal, decoy, guide, or scaffold molecules
[94]. lncRNAs and circRNAs (covalently closed con-
tinuous loops) interact with miRNAs, resulting in the
so-called ‘sponge’ effect, a specific mechanism of RNA
interference in the regulation of gene expression [95].
Both lncRNAs and circRNAs have been implicated in
the development and progression of various diseases
due to their ability to act as miRNA sponges, which
emphasizes their significance in biological process-
es and as potential therapeutic targets. Because of
their abnormal expression, lncRNAs are of great sig-
nificance in the early diagnostics of cancer. For ex-
ample, lncRNA CASC11 (cancer susceptibility  11) has
been identified as an oncogenic lncRNA at the early
stage of BCa development. It was found to activate
cell proliferation through miRNA‐150 [96] and to pro-
mote tumor cell growth and metastasis in colorec-
tal cancer by activating the WNT-β-catenin signaling
pathway [97].
Huang et al. [98] identified 50 m
6
A-lncRNAs with
a potential prognostic value that were methylated
mostly by METTL3 and RBM15 methyltransferases.
A risk score model has been developed based on 11
lncRNAs with the prognostic significance. The knock-
down of METTL3 and RBM15 in BCa cells inhibited
proliferation, invasion, and migration of tumor cells
in vitro and in vivo. The study did not fully address
SINYAGOVSKAYA et al.658
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
the heterogeneity of BCa, as the authors did not dif-
ferentiate between the tumor molecular subtypes,
which may impact the generalizability of the devel-
oped prognostic model. Also, no specific m
6
A sites
have been identified in the lncRNAs.
miRNA-221 and miRNA-222 play the oncogenic
role in BCa by binding to the 3′-UTR of the tumor
suppressor PTEN (phosphatase and tensin homolog)
mRNA, inhibiting its expression and promoting cell
proliferation in vitro and in vivo [99]. METTL3 was
found to induce miRNA221/222 maturation in an
m
6
A-dependent manner, which correlated with can-
cer progression [99]. The authors used BCa and paired
normal tissues from patients who had been diagnosed
with BCa and underwent surgery (180 cases), two BCa
cell lines, and normal urinary epithelial cell line (con-
trol) for in vitro experiments; the results if these ex-
periments were validated in vivo. The patients were
categorized based on age and sex (<65 and ≥65 years
old; male and female), however, the relation of the
obtained data to those factors was not analyzed. The
study was focused exclusively on BCa and did not de-
tail whether the patients had any non-cancerous con-
dition; METTL3 expression and miR221/222 maturation
were studied exclusively in the context of BCa devel-
opment. Moreover, no multivariate analysis was per-
formed to determine whether the METTL3-miR221/222
axis could be used as an independent prognostic fac-
tor when adjusted for confounding variables, such
as tumor stage, grade, or molecular subtype. While
technically challenging, targeting ncRNAs has become
increasingly feasible with the CRISPR RNA-targeting
systems like Cas13. However, low delivery efficiency,
required specificity, and small size of miRNA targets
remain significant obstacles.
BCa-associated lncRNA BLACAT3 (BLCa-associated
transcript 3) is implicated in the promotion of angio-
genesis and hematogenous metastasis by activating
the downstream NF-κB signaling in an m
6
A-dependent
manner [100]. The authors assessed clinical samples
of tumor and adjacent normal tissues from 107 pa-
tients with MIBC who underwent radical cystectomy,
although no stratification by sex or age was per-
formed. Invitro experiments were carried in RT4 cells
(NMIBC cell line), MIBC-derived cell lines (UM-UC-3,
5637, T24, J82, SW780), and SV-HUC-1 cells (control).
The study provided experimental evidence linking
epitranscriptomic modifications (m
6
A) to cancer pro-
gression via lncRNAs (an emerging field in oncology).
The authors demonstrated the following mechanis-
tic pathway: m
6
A-modified BLACAT3 recruits YBX3
(DNA- and RNA-binding protein involved in the tran-
scription regulation, RNA stabilization, and transla-
tion), leading to the upregulation of NCF2 (a subunit
of the NADPH oxidase complex), which promotes an-
giogenesis and metastasis. The study suggested that
BLACAT3 might be specific to MIBC, as no significant
differences were observed between the RT4 cells
(NMIBC) and normal urothelial cells.
A recent study by Liu et al. [101] unveiled a
molecular mechanism of the interaction between
LINC01106 (long intergenic non-protein coding RNA
1106), miR-3148, and adaptor protein DAB1 (Dis-
abled-1). It was found that LINC01106 and miR-3148
competitively bind to the DAB1 mRNA. LINC01106
stabilizes the transcript, leading to a better disease
prognosis, while miR-3148, on the opposite, inhibits
its translation, resulting in a poor overall outcome of
BCa. Interestingly, CRISPR-mediated hypermethylation
of LINC01106 facilitated by dCas13b-METTL3-METTL14
increased its affinity to DAB1. The authors analyzed
four BCa cell lines and SVHUC-1 cells (control), as well
as 30 paired BCa tissue samples (malignant and adja-
cent normal tissues) obtained from BCa patients that
underwent surgery.
circRNAs are generally known to play the on-
cogenic role. Thus, the IGF2BP3 reader protein was
found to stabilize m
6
A-methylated circPSMA7, result-
ing in the enhanced expression of mitogen-activated
protein kinase  1 (MAPK1) and BCa progression and
metastasis. circPSMA7 acts a sponge for miR-128-3p
that exhibits the tumor suppressor activity by target-
ing the MAPK1 mRNA [102]. The study analyzed 33
paired samples of fresh BCa and adjacent normal tis-
sues. BCa cell lines UM-UC3 and T24, SV-HUC-1 cells,
and 293T human embryonic kidney cells were used
in in vitro experiments. The authors predicted eight
potential m
6
A modification sites in circPSMA7 and
carried out methylated RNA immunoprecipitation
(MeRIP) assay to confirm the presence of m
6
A modifi-
cations in the transcript. However, no mutations were
introduced in the identified sites to validate each site
separately and to determine which sites contributed
to the IGF2BP3-mediated stabilization of circPSMA7.
It also remained unclear whether individual sites
could be used independently as prognostic markers.
METTL14 plays a significant role in the advance-
ment of BCa by promoting expression of lncDBET
(D4Z4 binding element transcript lncRNA). Five m
6
A
sites in lncDBET methylated by METTL14 were pre-
dicted in [103]. The elevated levels of lncDBET trig-
ger a cascade of events that ultimately contribute to
the development of BCa, in particular, activation of
the peroxisome proliferator-activated receptor (PPAR)
signaling pathway through recruitment of fatty acids,
which has a profound impact on the lipid metabo-
lism in cancer cells. The mechanism involves a direct
interaction between lncDBET and fatty acid-binding
protein  5 (FABP5). The knockdowns of METTL14,
lncDBET, or FABP5 suppressed tumor growth in vi-
tro and invivo. The study was conducted in SVHUC-1
cells and BCa cell lines (UMUC3, 5637, T24, J82,
m
6
A-RNA METHYLATION IN BLADDER CANCER DEVELOPMENT 659
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
and EJ-M3), as well as in fresh BCa tumor tissues
and adjacent non-tumor specimens collected from
patients subjected to radical cystectomy or transure-
thral resection and were used to evaluate the levels
of METTL14, lncDBET, and FABP5. The patients were
not stratified based on the cancer stage or subtype.
Because of cancer heterogeneity, it is unclear whether
all BCa subtypes exhibited similar METTL14-lncDBET-
FABP5 expression patterns. The authors proposed the
METTL14-lncDBET-FABP5 axis as a potential thera-
peutic target, but did not address if such approach
would be clinically feasible without disruption of es-
sential pathways and unwanted off-target effects. The
prognostic value of this axis still has to be elucidated,
as m
6
A patterns and expression profiles of m
6
A fac-
tors may help stratification of patients into subgroups
and prediction of disease progression.
Interestingly, an elevated expression of METTL14
was discovered to be characteristic of more ‘aggres-
sive’ cell lines [104]. The authors analyzed 120 forma-
lin-fixed paraffin-embedded bladder urothelial carci-
noma samples (60 NMIBC and 60 MIBC), 40 normal
urothelial tract samples from nephrectomy specimens,
and 7  BCa cell lines. The knockdown of METTL14
caused a significant decrease in the m
6
A modification
levels in the cells, leading to a substantial inhibition
of cancer cell migration and invasion. Since METTL14
and METTL3 form a heterodimer that acts as a key
m
6
A ‘writer’ complex, downregulation of METTL14 led
to the disruption of its catalytic activity  [104]. How-
ever, others authors have reported that the METTL14
upregulation in BCa cells suppressed cell prolifera-
tion and migration of BCa cell lines [105,  106]. This
discrepancy may arise from the tumor heterogeneity,
as bladder tumors contain subpopulations of cells
with different molecular profiles  [107], i.e., different
regions of the same tumor may have varying levels
of METTL14. In  [104], the samples were not strati-
fied into subgroups based on the m
6
A profiles, which
makes it unclear whether METTL14 had a stronger
impact in certain groups of patients. The authors did
not explore potential downstream targets of METTL14,
so it remains obscure how METTL14 contributes to
the tumor progression or aggressiveness in BCa.
In summary, m
6
A plays a multifaceted role in the
regulation of both oncogenes and tumor-suppressors
in BCa. The levels of m
6
A methylation influence the
transcriptome fate in a diverse and somewhat unpre-
dictable way: either high or low methylation levels
may either up- or downregulate transcript expression.
m
6
A is a highly dynamic modification, and methyl-
ation patterns may vary at different disease stages,
which can be potentially used to precisely differenti-
ate the stage of disease development and predict its
progression, outcome, and possibility of recurrence.
Figure  1 illustrates m
6
A-related factors that play onco-
genic role in the development of BCa, as well as the
mechanisms of their action.
Factors associated with m
6
A that exert tumor sup-
pressive functions in the BCa development are illus-
trated in Fig. 2.
This novel approach has been extensively stud-
ied in a number of works that have proposed various
targets and potential applications in cancer therapy.
However, a deeper understanding of m
6
A modifica-
tion mechanisms and the role of this modification as a
prognostic biomarker is still required, e.g., for the use
of m
6
A biomarkers for correcting existing manage-
ment of patients. Many ongoing studies have limita-
tions that need to be resolved. For example, no links
have been established between the m
6
A profiles or
expression patterns of m
6
A factors and tumor stage,
grade, or other risk factors, even if it could help clini-
cians to stratify patients into groups. In most studies,
no association between the expression of m
6
A factors
and patients’ age and sex have been investigated (al-
though many authors state that these parameters do
not confound the analysis) [44, 93]. Often, the studies
do not specify whether the patients had chronic dis-
eases, comorbidities, or polymorbidities, which raises
the possibility that the expression of m
6
A factors could
be associated with such concurrent conditions. Most
studies are performed in BCa tissues and do not ex-
plore the possibility of m
6
A factor assessment in the
blood, urine, or other biological fluids, which would be
more clinically relevant for non-invasive diagnostics.
m
6
A-mediated drug resistance. Several studies
have highlighted the role of m
6
A modification in drug
resistance, including resistance to chemotherapy, im-
munotherapy, and specific drugs (e.g., cisplatin and
anthracyclines) in different cancers, including BCa
[108-110].
With the discovery of cancer immune checkpoints
and checkpoint inhibitors, such as programmed cell
death protein 1 (PD-1) and programmed death-ligand1
(PD-L1), the possibilities for using novel treatment
strategies have considerably expanded. For example,
METTL3 and METTL14 were reported to suppress in-
nate immune response to cancer cells in colorectal
carcinoma and melanoma by introducing m
6
A in the
3′-UTRs of transcripts of key factors involved in the
immune response signaling pathways, namely STAT1
(signal transducer and activator of transcription pro-
tein family) and IRF1 (interferon regulatory factor1).
METTL3-14 depletion caused a noticeable increase in
the production of cytokines and chemokines, as well
as activation of IFN‐γ signaling, suggesting m
6
A to
be a key factor in the regulation of cell sensitivity
to immunotherapy. Moreover, METTL3-14-depleted
cells also exhibited profound sensitivity to anti-PD-1
treatment [111]. It was suggested that METTL3 ex-
pression is regulated by JNK1, a core component
SINYAGOVSKAYA et al.660
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig. 1. Oncogenic effect of m
6
A modification of RNA in bladder cancer biology. a) m
6
A methylation of ITGA6 tran-
script mediated by METTL3 and YTHDF1/YTHDF3 reader proteins, promotes its translation and tumor progression [55].
b)METTL3-m
6
A-CDCP1 axis regulates CDCP1 expression and translation and promotes tumor progression, cell migration, and
metastasis [63]. c)m
6
A-eraser FTO demethylase regulates expression and stability of PTPN6, a key phosphatase involved in
cell signaling and cancer progression, suggesting its prognostic potential in BCa [42]. d) NRF2 promotes tumor growth and
resistance to ferroptosis through the m
6
A-mediated stabilization by WTAP and YTHDF1 [92]. e)FTO acts as an oncogene by
reducing m
6
A methylation of the MALAT1 lncRNA, enhancing its stability and expression, which promotes tumor growth,
since MALAT1 sponges miR-384 leading to the increased MAL2 levels; higher FTO expression correlates with advanced
cancer stages [43]. f) IGF2BP2 stabilizes m
6
A-modified NRP1 transcript, leading to the M2 macrophage polarization and
promotion of BCa progression [46]. g) FTO promotes BCa progression by modifying the maturation of pri-miR-576 through
the m
6
A-dependent mechanisms and regulates the miR-576-CDK6 pathway; higher FTO expression correlates with advanced
TNM stages [44]. h)METTL3 promotes miRNA221/222 maturation through m
6
A modification, facilitating cancer progression
via inhibition of the tumor suppressor PTEN [99] i) METTL14 promotes BCa progression by increasing lncDBET expres-
sion, which activates the PPAR signaling pathway and alters lipid metabolism through direct interaction with FABP5 [103].
j) m
6
A-methylated circPSMA7 is stabilized by IGF2BP3, which enhances MAPK1 stability and promotes tumor progression;
miR-128-3p can potentially reverse this effect [102]. k) IGF2BP3 interacts with m
6
A-methylated HMGB1 mRNA, influencing
the immune microenvironment of BCa tumor and affecting immunotherapy outcome [47].
of the JNK signaling pathway, which results in the
m
6
A methylation of PD-L1 mRNA, leading to its in-
creased expression in BCa cells [112].
Casein kinase 2 (CK2) is a serine/threonine pro-
tein kinase that regulates glycolysis in cancer cells.
Increased lactate production and glucose utilization
contribute to cancer progression. Demethylase ALKBH5
decreases the stability of the CK2 mRNA by removing
m
6
A from the transcript. ALKBH5 is downregulated in
BCa cells, resulting in more pronounced cell prolifer-
ation and tumor growth. Overexpression of ALKBH5
increases the sensitivity of BCa cells to cisplatin via
the CK2a-mediated glycolytic pathway [113, 114].
Some circRNAs play a major role in facilitating
chemoresistance in cancers (including BCa) in an
m
6
A-dependent manner. Xu et  al. [115] demonstrat-
ed that the expression of circ104797 in BCa tissues
was elevated compared to adjacent healthy tissues,
and the underlying mechanism of this upregulation
is m
6
A-dependent. The content of m
6
A-methylated
circ104797 was increased in cisplatin-resistant cell
lines, while targeted demethylation using the CRISPR-
dCas13b-ALKBH5 system significantly reduced it.
circ104797 is known to act as a sponge for miR-103a
and miR-660-3p that modulate cell sensitivity to drugs
by controlling the ability to expel chemotherapeutic
m
6
A-RNA METHYLATION IN BLADDER CANCER DEVELOPMENT 661
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig. 2. Tumor suppressive effect of m
6
A modification of RNA in bladder cancer biology. a) METTL14-m
6
A-Notch1 axis in-
hibits self-renewal of tumor-inducing cells (TICs) [71]. b) SYTL1 enhances anti-tumor immune response by activating NK
cells; m
6
A methyltransferase WTAP downregulates SYTL1 expression through m
6
A modification, leading to the transcript
degradation by YTHDF2 [81]. c) METTL3-introduced m
6
A modification is recognized by YTHDF2 and leads to the decay
of mRNA for the tumor-suppressor factor SETD7 [80] d) KLF4 is a tumor suppressor downregulated in BCa cells and
linked to poor survival and recurrence risk. KLF4 overexpression inhibits cell proliferation and induces cell cycle arrest
in G1 phase. METTL3 m
6
A-methylates the KLF mRNA, causing it recognition by the YTHDF2 reader protein and further
degradation [80]. e) LINC01106 stabilizes DAB1 mRNA, improving BCa prognosis, while miR-3148 inhibits its translation,
leading to poorer disease outcome. CRISPR-mediated hypermethylation of LINC01106 enhances its affinity to DAB1 [101].
agents and regulating genes involved in the DNA dam-
age response and DNA repair, respectively. The authors
suggested that circ104797 can be used as a biomarker
for predicting cell response to the anti-cancer treat-
ment by measuring its expression levels or levels of
its m
6
A-methylation, or as a target of treatment aimed
to disrupt circ104797 interaction with miRNAs to im-
prove the treatment outcome [115].
Another circRNA upregulated in BCa cells is
circ0008399. It binds WTAP to promote formation
of the WTAP-METTL3-METTL14 m
6
A methyltransfer-
ase complex that induces m
6
A modification in tumor
cells. Moreover, it facilitates the posttranscriptional
regulation of TNFAIP3 (tumor necrosis factor, alpha-
induced protein  3), an anti-apoptotic protein that in-
hibits TNF-induced apoptosis [116], thus promoting
cell resistance to chemotherapy [117].
Interestingly, key m
6
A regulators may also act as
tumor suppressors. Thus, lower levels of METTL16
were found to correlate with a poor outcome in BCa
patients [118]. Furthermore, METTL16 exerted a po-
tent anti-cancer effect in BCa cells both in in vitro
and invivo by reducing cell proliferation and increas-
ing the sensitivity to drugs (cisplatin). These effects
were achieved through a complex molecular path-
way involving HIF-2α (hypoxia-inducible factor  2α),
PMEPA1 (prostate transmembrane protein, androgen
induced 1), and autophagy regulated by the m
6
A
methylation of the respective mRNAs. These findings
open new prospects for developing targeted therapies
against BCa and selecting prognostic biomarkers for
prediction of patients’ response to chemotherapeutics.
There has long been a demand for more effi-
cient methods for predicting potential therapeutic
resistance and evaluating the treatment outcomes.
Abnormal expression of m
6
A regulators is typically
associated with various forms of resistance, offering
a basis for predicting cancer response to treatment.
First, there is a number of effective small-molecule
inhibitors for almost all major groups of m
6
A path-
way factors [109], which can be used to overcome
the drug resistance caused by the expression of these
factors. Second, expression levels of the m
6
A pathway
factors and the levels of m
6
A methylation of certain
transcripts associated with the drug resistance can be
used as diagnostic markers for predicting resistance
to therapy.
Mutations in m
6
A motifs and m
6
A factors.
m
6
A modifications are prevalent in certain mRNA re-
gions, such as stop codons, 3′-UTRs, and long inner
SINYAGOVSKAYA et al.662
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig. 3. The workflow for developing personalized treatment strategies based on the m
6
A RNA modification profiling in a pa-
tient population. a)Tissue samples are collected from the patients, including those with different BCa stages. m
6
A sequencing
is performed to assess m
6
A modification patterns in RNA, which may vary depending on the disease severity. b) Based on
m
6
A patterns, patients are classified into distinct subgroups. c) Patients’ survival probability is assessed based on the m
6
A
profiles to provide insights into the correlation between the m
6
A modification of RNA and clinical outcomes and to help
predict patients’ response to specific treatments. d)Personalized treatment strategy is designed based on the assessed risks.
exon regions. They are typically located in the con-
sensus motifs including UGACA, RRACH (R  =  G or A;
H  =  A, C, or U), and DRACH (where D  =  A, G or U;
R  =  A or G; H  =  A, C or U), as has been verified by
high-throughput sequencing. m
6
A modification is a
major molecular mechanism regulating the fate of
RNAs in response to various stimuli [119-121]. Func-
tional m
6
A sites are highly evolutionary conserved,
whereas “accidental” nonfunctional m
6
A modifica-
tions occurring as a result of methyltransferase off-
site targeting, are typically purged by natural selection
[122]. Mutations within the consensus motifs can be
detrimental, as they could potentially prevent meth-
ylation of adenosines and attenuate or even abolish
the enhanced translation of the corresponding tran-
scripts. Cancer cells seem to be generally resistant
to mutations within m
6
A consensus motifs [123].
SNPs (single nucleotide polymorphisms) affect-
ing m
6
A-methylation can occur in two primary loca-
tions – near and directly at m
6
A-modification sites
and significantly impact various biological processes
and pathways. Some SNPs cause mutations, potential-
ly interfering with the transcription and translation;
others are located in the UTRs or near stop codons,
affecting mRNA interaction with transcriptional reg-
ulators and RNA-binding proteins. These alterations
can influence mRNA stability and efficiency of its
nuclear transport [124]. As a result, gene expression
and overall cellular functions can be affected. The ef-
fects of these m
6
A-SNPs can vary depending on their
exact location and genes involved [125]. Further re-
search is needed to fully understand the implications
of these genetic variations in cellular processes and
disease mechanisms.
Identification of m
6
A-related SNPs can be partic-
ularly important in molecular diagnostics. One of the
examples of such m
6
A-SNPs is rs3088107 in the RNFT2
(ring finger protein, transmembrane  2) oncogene. Nor-
mally, expression of RNFT2 is elevated in BCa cells,
which promotes proliferation and metastasis of cancer
cells. rs3088107 SNP abolishes RNFT2 expression in
an m
6
A-mediated manner [125].
Mutations in the genes coding for m
6
A-related
factors may not only affect m
6
A methylation and
the fate of respective transcripts, but also result in
the production of proteins with an altered substrate
specificity. Indeed, the gain-of-function missense mu-
tation R298P in METTL14, which is responsible for the
recognition of the non-canonical sequence of GGAU
instead of canonical GGAC, is frequently found in can-
cer patients. The effects of the R298P mutation are
similar to those of the METTL3-METTL14 complex
overexpression linked to cancer cell growth and pro-
liferation [126].
In conclusion, since the m
6
A methylation pathway
plays a role in both tumorigenesis and tumor suppres-
sion at the levels of mRNA expression and ncRNAs,
and because mutations in m
6
A domains lead to
changes in the expression of m
6
A factors, m
6
A meth-
ylation can serve as a diagnostic marker of disease
m
6
A-RNA METHYLATION IN BLADDER CANCER DEVELOPMENT 663
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
progression and development rate. Figure 3 presents
a potential workflow for m
6
A profiling as a novel
prognostic biomarker for the stratification of BCa pa-
tients. RNAs or m
6
A-methylated RNAs can be extracted
from the patients’ tissues or blood using specific m
6
A-
targeting antibodies, sequenced, and subjected to bio-
informatic analysis, that would allow for the assess-
ment of expression levels of mRNAs and ncRNAs, as
well as the levels of m
6
A methylation of target RNAs.
Based on the results of analysis, predictions can be
made regarding the rate of disease progression, like-
lihood of tumor recurrence, and therapy efficacy to
help with the development of disease management
strategy for individual patients.
CONCLUSION
BCa presents a significant challenge to healthcare
because one of the highest prevalence among cancers,
as well as high recurrence and mortality rates. Timely
and accurate diagnostics and selection of appropriate
treatment significantly affect the BCa prognosis in pa-
tients. While the m
6
A-based molecular diagnostics is
still at its inception, the usefulness of m
6
A biomarkers
have been demonstrated for many cancers. Numer-
ous prospective m
6
A biomarkers of BCa have been
described, although in a limited number of patients.
There is a current need for identification of new BCa
biomarkers that could be conveniently analyzed, and
not only in tumor cells, but also in patients’ urine,
blood cells, or circulation (e.g., exosomal biomarkers
or free nucleic acids). The lack of valid and reliable
molecular biomarkers for predicting BCa development,
NMIBC-to-MIBC progression, and response to cisplatin
and other drugs (including PD-1/PD-1L) negatively af-
fects the prognostics in BCa patients.
The levels of m
6
A methylation and SNPs in mR-
NAs and ncRNAs, as well as the expression levels of
m
6
A factors, may serve as valuable biomarkers to
shed light on tumor behavior and patient prognosis
and to define the optimal treatment options. Dis-
tinct m
6
A patterns may act as early biomarkers for
cancer detection and risk assessment. Identification
of new m
6
A-related genes and evaluation of their
significance through the assessment of m
6
A scores,
clustering, enrichment analysis, and disease outcome
prediction could help to pinpoint potential targets for
precision therapy, evaluate the risk of cancer relapse,
and diagnose BCa at the earliest possible stage to en-
sure high survival rates. m
6
A profiling could be used
for stratifying the patients according to cancer sub-
types and clinicopathological characteristics, predict-
ing their response to treatment, and evaluating the
outcomes. The use of m
6
A machinery for therapeutic
and prognostic purposes is currently at its infancy.
However, this topic has been studied extensively,
and several prognostic models based on m
6
A profiles
have been already developed. A proposed prognostic
m
6
A-driven marker panel for renal cell carcinoma
(RCC) based on the transcriptome-wide m
6
A-seq data
can identify dysregulated m
6
A-modified target genes,
such as NDUFA4L2 (NDUFA4 mitochondrial complex
associated like  2, 4-like), NXPH4 (neurexophilin  4),
and UMOD (uromodulin), associated with significantly
poorer overall survival in RCC patients [127]. A novel
m
6
A classifier has recently been designed to predict
the treatment efficacy and response to treatment in
BCa patients [128]. All things considered, m
6
A meth-
ylation may serve as a prospective non-invasive diag-
nostic biomarker in the diagnostics and prognostics
of BCa.
Abbreviations. BCa, bladder cancer; METTL,
methyltransferase like protein; MIBC, muscle-invasive
bladder cancer; NMIBC, non-muscle-invasive bladder
cancer.
Acknowledgments. The authors thank Priority
2030 Research Program (Sechenov University).
Contributions. T.S., D.K., and A.K. developed the
study concept; T.S., Y.L., N.V-K., A.A., N.P., S.B., and I.G.
wrote the original draft; T.S., A.D., A.V., D.K., and A.K.
edited the manuscript; T.S. prepared the figures; V.C.,
A.D., A.V., and D.K. supervised the project; D.K. and
A.K. provided project administration; A.K. acquired
the funding. All authors have read and agreed to the
published version of the manuscript.
Funding. The study was supported by the Russian
Science Foundation (project no.22-75-10032).
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 de-
clare that they have no conflicts of interest.
REFERENCES
1. Ferlay, J., Ervik, M., Lam, F., Laversanne, M., Colombet,M.,
Mery, L., Piñeros, M., Znaor, A., Soerjomataram, I., and
Bray, F. (2024) Global cancer observatory: cancer today,
Int. Agency Res., URL: https://gco.iarc.who.int/today.
2. Burger, M., Catto, J. W. F., Dalbagni, G., Grossman,
H.B., Herr,H., Karakiewicz,P., Kassouf,W., Kiemeney,
L. A., La Vecchia, C., Shariat, S., and Lotan, Y. (2013)
Epidemiology and risk factors of urothelial bladder
cancer, Eur. Urol., 63, 234-241, https://doi.org/10.1016/
j.eururo.2012.07.033.
3. Aldousari, S., and Kassouf, W. (2010) Update on the
management of non-muscle invasive bladder cancer,
Can. Urol. Assoc.J., 4, 56-64, https://doi.org/10.5489/
cuaj.777.
SINYAGOVSKAYA et al.664
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
4. Shalata, A.T., Shehata,M., Van Bogaert,E., Ali, K.M.,
Alksas, A., Mahmoud, A., El-Gendy, E. M., Mohamed,
M.A., Giridharan, G.A., Contractor, S., and El-Baz, A.
(2022) Predicting recurrence of non-muscle-inva-
sive bladder cancer: current techniques and future
trends, Cancers, 14, 5019, https://doi.org/10.3390/can-
cers14205019.
5. Jeong,S., Han, J.H., Jeong, C.W., Kim, H.H., Kwak,C.,
Yuk, H.D., and Ku, J. H. (2022) Clinical determinants
of recurrence in pTa bladder cancer following trans-
urethral resection of bladder tumor, BMC Cancer, 22,
631, https://doi.org/10.1186/s12885-022-09733-8.
6. D′Andrea, D., Abufaraj, M., Susani, M., Ristl, R.,
Foerster,B., Kimura,S., Mari,A., Soria,F., Briganti,A.,
Karakiewicz, P. I., Gust, K. M., Rouprêt, M., and
Shariat, S. F. (2018) Accurate prediction of progres-
sion to muscle-invasive disease in patients with
pT1G3 bladder cancer: a clinical decision-making tool,
Urol. Oncol., 36, 239.e1-239.e7, https://doi.org/10.1016/
j.urolonc.2018.01.018.
7. Lorenz, D.A., Sathe,S., Einstein, J.M., and Yeo, G.W.
(2020) Direct RNA sequencing enables m(6)A detec-
tion in endogenous transcript isoforms at base-specif-
ic resolution, RNA, 26, 19-28, https://doi.org/10.1261/
rna.072785.119.
8. Shu,L., Huang,X., Cheng,X., and Li,X. (2021) Emerg-
ing roles of N6-methyladenosine modification in
neurodevelopment and neurodegeneration, Cells, 10,
2694, https://doi.org/10.3390/cells10102694.
9. Sokpor, G., Xie, Y., Nguyen, H. P., and Tuoc, T. (2021)
Emerging role of m(6)A methylome in brain devel-
opment: implications for neurological disorders and
potential treatment, Front. Cell Dev. Biol., 9, 656849,
https://doi.org/10.3389/fcell.2021.656849.
10. Qin, Y., Li, L., Luo, E., Hou, J., Yan, G., Wang, D.,
Qiao, Y., and Tang, C. (2020) Role of m6A RNA meth-
ylation in cardiovascular disease (review), Int. J. Mol.
Med., 46, 1958-1972, https://doi.org/10.3892/ijmm.
2020.4746.
11. Kumari,R., Ranjan,P., Suleiman, Z.G., Goswami, S.K.,
Li,J., Prasad,R., and Verma, S.K. (2022) mRNA mod-
ifications in cardiovascular biology and disease: with
a focus on m6A modification, Cardiovasc. Res., 118,
1680-1692, https://doi.org/10.1093/cvr/cvab160.
12. Williams, G. D., Gokhale, N. S., and Horner, S. M.
(2019) Regulation of viral infection by the RNA mod-
ification N6-methyladenosine, Annu. Rev. Virol., 6,
235-253, https://doi.org/10.1146/annurev-virology-
092818-015559.
13. Li, Y., Zhang, Q., Cui, G., Zhao, F., Tian, X., Sun, B.-F.,
Yang, Y., and Li, W. (2020) m(6)A regulates liver met-
abolic disorders and hepatogenous diabetes, Gen-
om. Proteom. Bioinform., 18, 371-383, https://doi.org/
10.1016/j.gpb.2020.06.003.
14. Zhang, C., Fu, J., and Zhou, Y. (2019) A review in
research progress concerning m6A Methylation
and Immunoregulation, Front. Immunol., 10, 922,
https://doi.org/10.3389/fimmu.2019.00922.
15. Tan, F., Zhao, M., Xiong, F., Wang, Y., Zhang, S.,
Gong, Z., Li, X., He, Y., Shi, L., Wang, F., Xiang, B.,
Zhou, M., Li, X., Li,Y., Li, G., Zeng, Z., Xiong, W., and
Guo,C. (2021) N6-methyladenosine-dependent signal-
ling in cancer progression and insights into cancer
therapies, J. Exp. Clin. Cancer Res., 40, 146, https://
doi.org/10.1186/s13046-021-01952-4.
16. Hong, K. (2018) Emerging function of N6-methylade-
nosine in cancer, Oncol. Lett., 16, 5519-5524, https://
doi.org/10.3892/ol.2018.9395.
17. Weiner, A. B., Desai, A. S., and Meeks, J. J. (2019)
Tumor location may predict adverse pathology and
survival following definitive treatment for blad-
der cancer: a national cohort study, Eur. Urol.
Oncol., 2, 304-310, https://doi.org/10.1016/j.euo.
2018.08.018.
18. Kassouf, W., Spiess, P. E., Brown, G. A., Liu, P.,
Grossman, H. B., Dinney, C. P. N., and Kamat, A. M.
(2008) Prostatic urethral biopsy has limited use-
fulness in counseling patients regarding final ure-
thral margin status during orthotopic neobladder
reconstruction, J. Urol., 180, 164-167, https://doi.org/
10.1016/j.juro.2008.03.037.
19. Gontero, P., Birtle, A., Capoun, O., Compérat, E.,
Dominguez-Escrig, J. L., Liedberg, F., Mariappan, P.,
Masson-Lecomte,A., Mostafid, H. A., Pradere,B., Rai,
B. P., van Rhijn, B. W. G., Seisen, T., Shariat, S. F.,
Soria, F., Soukup, V., Wood, R., and Xylinas, E. N.
(2024) European association of urology guidelines on
non-muscle-invasive bladder cancer (TaT1 and car-
cinoma in situ) – a summary of the 2024 guidelines
update, Eur. Urol., 86, 531-549, https://doi.org/10.1016/
j.eururo.2024.07.027.
20. Flores Monar, G.V., Reynolds,T., Gordon,M., Moon,D.,
and Moon, C. (2023) Molecular markers for bladder
cancer screening: an insight into bladder cancer
and FDA-approved biomarkers, Int. J. Mol. Sci., 24,
14374, https://doi.org/10.3390/ijms241814374.
21. Fan, J., Chen, B., Luo, Q., Li, J., Huang, Y., Zhu, M.,
Chen, Z., Li, J., Wang, J., Liu, L., Wei, Q., and Cao, D.
(2024) Potential molecular biomarkers for the di-
agnosis and prognosis of bladder cancer, Biomed.
Pharmacother., 173, 116312, https://doi.org/10.1016/
j.biopha.2024.116312.
22. Hu,X., Li,G., and Wu,S. (2022) Advances in diagnosis
and therapy for bladder cancer, Cancers (Basel), 14,
3181, https://doi.org/10.3390/cancers14133181.
23. Batista, R., Vinagre, N., Meireles, S., Vinagre, J.,
Prazeres, H., Leão, R., Máximo, V., and Soares, P.
(2020) Biomarkers for bladder cancer diagnosis and
surveillance: a comprehensive review, Diagnostics,
10, 39, https://doi.org/10.3390/diagnostics10010039.
24. Soria, F., Droller, M. J., Lotan, Y., Gontero, P.,
D’Andrea, D., Gust, K. M., Rouprêt, M., Babjuk, M.,
m
6
A-RNA METHYLATION IN BLADDER CANCER DEVELOPMENT 665
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Palou, J., and Shariat, S. F. (2018) An up-to-date
catalog of available urinary biomarkers for the sur-
veillance of non-muscle invasive bladder cancer,
World J. Urol., 36, 1981-1995, https://doi.org/10.1007/
s00345-018-2380-x.
25. Sylvester, R. J., Oosterlinck, W., Holmang, S., Sydes,
M. R., Birtle, A., Gudjonsson, S., De Nunzio, C.,
Okamura,K., Kaasinen,E., Solsona,E., Ali-El-Dein,B.,
Tatar, C.A., Inman, B.A., N’Dow,J., Oddens, J.R., and
Babjuk, M. (2016) Systematic review and individu-
al patient data meta-analysis of randomized trials
comparing a single immediate instillation of chemo-
therapy after transurethral resection with transure-
thral resection alone in patients with stage pTa-pT1
urothelial carcinoma, Eur. Urol., 69, 231-244, https://
doi.org/10.1016/j.eururo.2015.05.050.
26. Lidagoster, S., Ben-David, R., De Leon, B., and
Sfakianos, J.P. (2024) BCG and alternative therapies to
BCG therapy for non-muscle-invasive bladder cancer,
Curr. Oncol., 31, 1063-1078, https://doi.org/10.3390/
curroncol31020079.
27. Oerum,S., Meynier,V., Catala,M., and Tisné,C. (2021)
A comprehensive review of m6A/m6Am RNA meth-
yltransferase structures, Nucleic Acids Res., 49, 7239-
7255, https://doi.org/10.1093/nar/gkab378.
28. Wang,T., Kong,S., Tao,M., and Ju,S. (2020) The poten-
tial role of RNA N6-methyladenosine in Cancer pro-
gression, Mol. Cancer, 19, 88, https://doi.org/10.1186/
s12943-020-01204-7.
29. Qu, J., Yan, H., Hou, Y., Cao, W., Liu, Y., Zhang, E.,
He, J., and Cai, Z. (2022) RNA demethylase ALKBH5
in cancer: from mechanisms to therapeutic poten-
tial, J. Hematol. Oncol., 15, 8, https://doi.org/10.1186/
s13045-022-01224-4.
30. Azzam, S. K., Alsafar, H., and Sajini, A. A. (2022) FTO
m6A demethylase in obesity and cancer: implications
and underlying molecular mechanisms, Int. J. Mol.
Sci., 23, 3800, https://doi.org/10.3390/ijms23073800.
31. 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. (2014) N6-methyladenosine-dependent regula-
tion of messenger RNA stability, Nature, 505, 117-120,
https://doi.org/10.1038/nature12730.
32. Liu, N., Dai, Q., Zheng, G., He, C., Parisien, M., and
Pan, T. (2015) N(6)-methyladenosine-dependent RNA
structural switches regulate RNA-protein interac-
tions, Nature, 518, 560-564, https://doi.org/10.1038/
nature14234.
33. Huang,H., Weng,H., Sun,W., Qin,X., Shi,H., Wu,H.,
Zhao, B. S., Mesquita, A., Liu, C., Yuan, C. L., Hu,
Y.-C., Hüttelmaier, S., Skibbe, J. R., Su, R., Deng, X.,
Dong, L., Sun, M., Li, C., Nachtergaele, S., Wang, Y.,
Hu, C., Ferchen, K., Greis, K. D., Jiang, X., Wei, M.,
Qu,L., Guan, J.-L., He,C., Yang,J., and Chen,J. (2018)
Recognition of RNA N(6)-methyladenosine by IGF2BP
proteins enhances mRNA stability and translation,
Nat. Cell Biol., 20, 285-295, https://doi.org/10.1038/
s41556-018-0045-z.
34. Choe,J., Lin,S., Zhang,W., Liu,Q., Wang,L., Ramirez-
Moya,J., Du,P., Kim,W., Tang,S., Sliz,P., Santisteban,P.,
George, R.E., Richards, W.G., Wong, K.-K., Locker,N.,
Slack, F. J., and Gregory, R. I. (2018) mRNA circu-
larization by METTL3-eIF3h enhances translation
and promotes oncogenesis, Nature, 561, 556-560,
https://doi.org/10.1038/s41586-018-0538-8.
35. Uddin, M. B., Wang, Z., and Yang, C. (2021) The
m(6)A RNA methylation regulates oncogenic signal-
ing pathways driving cell malignant transforma-
tion and carcinogenesis, Mol. Cancer, 20, 61, https://
doi.org/10.1186/s12943-021-01356-0.
36. Deng, X., Su, R., Weng, H., Huang, H., Li, Z., and
Chen, J. (2018) RNA N(6)-methyladenosine modifi-
cation in cancers: current status and perspectives,
Cell Res., 28, 507-517, https://doi.org/10.1038/s41422-
018-0034-6.
37. Wang, S., Chai, P., Jia, R., and Jia, R. (2018) Novel in-
sights on m(6)A RNA methylation in tumorigenesis:
a double-edged sword, Mol. Cancer, 17, 101, https://
doi.org/10.1186/s12943-018-0847-4.
38. Liu, Q. (2021) Current advances in N6-methylad-
enosine methylation modification during bladder
cancer, Front. Genet., 12, 825109, https://doi.org/
10.3389/fgene.2021.825109.
39. Chen, M., Nie, Z., Wen, X., Gao, Y., Cao, H., and
Zhang, S. (2019) m6A RNA methylation regulators
can contribute to malignant progression and im-
pact the prognosis of bladder cancer, Biosci. Rep., 39,
BSR20192892, https://doi.org/10.1042/BSR20192892.
40. Wu,J., Wang,X., Xu,H., Tian,J., Ji,H., Zhu,J., Guo,H.,
and Chen, Z. (2022) Bioinformatics analysis of the
correlation between m6A RNA methylation regula-
tors and the immune infiltration and prognosis of
bladder cancer, Ann. Transl. Med., 10, 1386, https://
doi.org/10.21037/atm-22-5993.
41. Shen, C., Liu, J., Wang, J., Yang, X., Niu, H., and
Wang, Y. (2020) The analysis of PTPN6 for bladder
cancer: an exploratory study based on TCGA, Dis.
Markers, 2020, 4312629, https://doi.org/10.1155/2020/
4312629.
42. Wu, N., Sun, Y., Xue, D., and He, X. (2024) FTO pro-
motes the progression of bladder cancer via de-
methylating m6A modifications in PTPN6 mRNA,
Heliyon, 10, e34031, https://doi.org/10.1016/j.heliyon.
2024.e34031.
43. Tao, L., Mu, X., Chen, H., Jin, D., Zhang, R., Zhao, Y.,
Fan, J., Cao, M., and Zhou, Z. (2021) FTO modifies
the m6A level of MALAT and promotes bladder can-
cer progression, Clin. Transl. Med., 11, e310, https://
doi.org/10.1002/ctm2.310.
44. Zhou,G., Yan,K., Liu,J., Gao,L., Jiang,X., and Fan,Y.
(2021) FTO promotes tumour proliferation in blad-
der cancer via the FTO/miR-576/CDK6 axis in an
SINYAGOVSKAYA et al.666
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
m6A- dependent manner, Cell Death Discov., 7, 329,
https://doi.org/10.1038/s41420-021-00762-z.
45. Huang, W., Zhu, L., Huang, H., Li, Y., Wang, G., and
Zhang, C. (2023) IGF2BP3 overexpression predicts
poor prognosis and correlates with immune infiltra-
tion in bladder cancer, BMC Cancer, 23, 116, https://
doi.org/10.1186/s12885-022-10353-5.
46. Fu, D., Shi, X., Yi, X., Wu, D., He, H., Zhou, W., and
Cheng, W. (2024) m6A reader IGF2BP2 promotes M2
macrophage polarization and malignant biological
behavior of bladder cancer by stabilizing NRP1 mRNA
expression, BMC Urol., 24, 147, https://doi.org/10.1186/
s12894-024-01534-4.
47. Lv,L., Wei,Q., Zhang,J., Dong,Y., Shan,Z., Chang,N.,
Zhao, Y., Bian, P., and Yi, Q. (2024) IGF2BP3 pre-
vent HMGB1 mRNA decay in bladder cancer and
development, Cell. Mol. Biol. Lett., 29, 39, https://
doi.org/10.1186/s11658-024-00545-1.
48. Watt, F. M. (2002) Role of integrins in regulating
epidermal adhesion, growth and differentiation,
EMBOJ., 21, 3919-3926, https://doi.org/10.1093/emboj/
cdf399.
49. Hynes, R. O. (2002) Integrins: bidirectional, al-
losteric signaling machines, Cell, 110, 673-687, https://
doi.org/10.1016/S0092-8674(02)00971-6.
50. Stewart, R. L., West, D., Wang, C., Weiss, H. L.,
Gal, T., Durbin, E. B., O’Connor, W., Chen, M., and
O’Connor, K. L. (2016) Elevated integrin α6β4 ex-
pression is associated with venous invasion and de-
creased overall survival in non-small cell lung can-
cer, Hum. Pathol., 54, 174-183, https://doi.org/10.1016/
j.humpath.2016.04.003.
51. Ammothumkandy, A., Maliekal, T. T., Bose, M. V.,
Rajkumar,T., Shirley,S., Thejaswini,B., Giri, V.G., and
Krishna, S. (2016) CD66 and CD49f expressing cells
are associated with distinct neoplastic phenotypes
and progression in human cervical cancer, Eur. J.
Cancer, 60, 166-178, https://doi.org/10.1016/j.ejca.
2016.03.072.
52. Duan,X., Li,H., Wang,M., Ju,S., Li,F., Chen,P., Lu,H.,
Han, X., and Ren, J. (2021) PSMC2/ITGA6 axis plays
critical role in the development and progression of
hepatocellular carcinoma, Cell Death Discov., 7, 217,
https://doi.org/10.1038/s41420-021-00585-y.
53. Wei, L., Yin,F., Chen, C., and Li,L. (2019) Expression
of integrin α-6 is associated with multi drug resis-
tance and prognosis in ovarian cancer, Oncol. Lett.,
17, 3974-3980, https://doi.org/10.3892/ol.2019.10056.
54. Yamakawa, N., Kaneda,K., Saito, Y., Ichihara,E., and
Morishita,K. (2012) The increased expression of integ-
rin α6 (ITGA6) enhances drug resistance in EVI1(high)
leukemia, PLoS One, 7, e30706, https://doi.org/
10.1371/journal.pone.0030706.
55. Jin,H., Ying,X., Que,B., Wang,X., Chao,Y., Zhang,H.,
Yuan, Z., Qi, D., Lin, S., Min, W., Yang, M., and Ji, W.
(2019) N6-methyladenosine modification of ITGA6
mRNA promotes the development and progression
of bladder cancer, EBioMedicine, 47, 195-207, https://
doi.org/10.1016/j.ebiom.2019.07.068.
56. Ying, X., Huang, Y., Liu, B., Hu, W. Y., Ji, D., Chen, C.,
Zhang,H., Liang,Y., Lv, Y., and Ji, W. (2024) Targeted
m6A demethylation of ITGA6 mRNA by a multisite
dCasRx-m6A editor inhibits bladder cancer develop-
ment, J. Adv. Res., 56, 57-68, https://doi.org/10.1016/
j.jare.2023.03.010.
57. Law, M. E., Ferreira, R. B., Davis, B. J., Higgins, P. J.,
Kim, J.-S., Castellano, R. K., Chen, S., Luesch, H., and
Law, B. K. (2016) CUB domain-containing protein 1
and the epidermal growth factor receptor cooperate
to induce cell detachment, Breast Cancer Res., 18, 80,
https://doi.org/10.1186/s13058-016-0741-1.
58. Scherl-Mostageer,M., Sommergruber,W., Abseher,R.,
Hauptmann, R., Ambros, P., and Schweifer, N. (2001)
Identification of a novel gene, CDCP1, overexpressed
in human colorectal cancer, Oncogene, 20, 4402-4408,
https://doi.org/10.1038/sj.onc.1204566.
59. Perry, S. E., Robinson, P., Melcher, A., Quirke, P.,
Bühring, H.-J., Cook, G.P., and Blair, G.E. (2007) Expres-
sion of the CUB domain containing protein 1 (CDCP1)
gene in colorectal tumour cells, FEBS Lett., 581,
1137-1142, https://doi.org/10.1016/j.febslet.2007.02.025.
60. Ikeda, J., Oda, T., Inoue, M., Uekita, T., Sakai, R.,
Okumura, M., Aozasa, K., and Morii, E. (2009) Ex-
pression of CUB domain containing protein (CDCP1)
is correlated with prognosis and survival of patients
with adenocarcinoma of lung, Cancer Sci., 100, 429-
433, https://doi.org/10.1111/j.1349-7006.2008.01066.x.
61. Awakura, Y., Nakamura, E., Takahashi, T., Kotani, H.,
Mikami,Y., Kadowaki,T., Myoumoto,A., Akiyama,H.,
Ito, N., Kamoto, T., Manabe, T., Nobumasa, H.,
Tsujimoto,G., and Ogawa,O. (2008) Microarray-based
identification of CUB-domain containing protein1 as
a potential prognostic marker in conventional renal
cell carcinoma, J.Cancer Res. Clin. Oncol., 134, 1363-
1369, https://doi.org/10.1007/s00432-008-0412-4.
62. Casar, B., Rimann, I., Kato, H., Shattil, S. J., Quigley,
J.P., and Deryugina, E.I. (2014) In vivo cleaved CDCP1
promotes early tumor dissemination via complexing
with activated β1 integrin and induction of FAK/
PI3K/Akt motility signaling, Oncogene, 33, 255-268,
https://doi.org/10.1038/onc.2012.547.
63. Yang, F., Jin, H., Que,B., Chao, Y., Zhang, H., Ying,X.,
Zhou, Z., Yuan, Z., Su, J., Wu, B., Zhang, W., Qi, D.,
Chen, D., Min, W., Lin, S., and Ji, W. (2019) Dy-
namic m6A mRNA methylation reveals the role
of METTL3-m6A-CDCP1 signaling axis in chemical
carcinogenesis, Oncogene, 38, 4755-4772, https://
doi.org/10.1038/s41388-019-0755-0.
64. Ying,X., Jiang,X., Zhang,H., Liu,B., Huang,Y., Zhu,X.,
Qi, D., Yuan, G., Luo, J., and Ji, W. (2020) Program-
mable N6-methyladenosine modification of CDCP1
mRNA by RCas9-methyltransferase like 3 conjugates
m
6
A-RNA METHYLATION IN BLADDER CANCER DEVELOPMENT 667
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
promotes bladder cancer development, Mol. Cancer,
19, 169, https://doi.org/10.1186/s12943-020-01289-0.
65. Chen, X., Xu, W., Pan, J., Yang, H., Li, Y., Chen, X.,
Sun, Y., Liu, Q., and Qiu, S. (2024) m6A methylation
profiling as a prognostic marker in nasopharyn-
geal carcinoma: insights from MeRIP-Seq and RNA-
Seq, Front. Immunol., 15, 1492648, https://doi.org/
10.3389/fimmu.2024.1492648.
66. Li, A., Gan, Y., Cao, C., Ma, B., Zhang, Q., Zhang, Q.,
and Yao, L. (2021) Transcriptome-wide map of
N6-methyladenosine methylome profiling in human
bladder cancer, Front. Oncol., 11, 717622, https://
doi.org/10.3389/fonc.2021.717622.
67. Zhu, D.-H., Su, K.-K., Ou-Yang, X.-X., Zhang, Y.-H., Yu,
X.-P., Li, Z.-H., Ahmadi-Nishaboori, S.-S., and Li, L.-J.
(2024) Mechanisms and clinical landscape of N6-meth-
yladenosine (m6A) RNA modification in gastrointesti-
nal tract cancers, Mol. Cell. Biochem., 479, 1553-1570,
https://doi.org/10.1007/s11010-024-05040-x.
68. Gavas, S., Quazi, S., and Karpiński, T. M. (2021)
Nanoparticles for cancer therapy: current progress
and challenges, Nanoscale Res. Lett., 16, 173, https://
doi.org/10.1186/s11671-021-03628-6.
69. Palakurthi, S. S., Shah, B., Kapre, S., Charbe, N.,
Immanuel, S., Pasham, S., Thalla, M., Jain, A., and
Palakurthi, S. (2024) A comprehensive review of
challenges and advances in exosome-based drug de-
livery systems, Nanoscale Adv., 6, 5803-5826, https://
doi.org/10.1039/D4NA00501E.
70. Dogbey, D.M., Torres, V.E.S., Fajemisin,E., Mpondo,L.,
Ngwenya,T., Akinrinmade, O.A., Perriman, A.W., and
Barth,S. (2023) Technological advances in the use of
viral and non-viral vectors for delivering genetic and
non-genetic cargos for cancer therapy, Drug Deliv.
Transl. Res., 13, 2719-2738, https://doi.org/10.1007/
s13346-023-01362-3.
71. Gu, C., Wang, Z., Zhou, N., Li, G., Kou, Y., Luo, Y.,
Wang, Y., Yang, J., and Tian, F. (2019) Mettl14 in-
hibits bladder TIC self-renewal and bladder tumor-
igenesis through N(6)-methyladenosine of Notch1,
Mol. Cancer, 18, 168, https://doi.org/10.1186/s12943-
019-1084-1.
72. Cao, L., Ren, Y., Guo, X., Wang, L., Zhang, Q., Li, X.,
Wu, X., Meng, Z., and Xu, K. (2020) Downregulation
of SETD7 promotes migration and invasion of lung
cancer cells via JAK2/STAT3 pathway, Int. J. Mol.
Med., 45, 1616-1626, https://doi.org/10.3892/ijmm.
2020.4523.
73. Akiyama, Y., Koda, Y., Byeon, S.-J., Shimada, S.,
Nishikawaji, T., Sakamoto, A., Chen, Y., Kojima, K.,
Kawano, T., Eishi, Y., Deng, D., Kim, W. H., Zhu,
W.-G., Yuasa, Y., and Tanaka, S. (2016) Reduced ex-
pression of SET7/9, a histone mono-methyltransfer-
ase, is associated with gastric cancer progression,
Oncotarget, 7, 3966-3983, https://doi.org/10.18632/
oncotarget.6681.
74. Gu, Y., Wang, X., Liu, H., Li, G., Yu, W., and Ma, Q.
(2018) SET7/9 promotes hepatocellular carcinoma
progression through regulation of E2F1, Oncol. Rep.,
40, 1863-1874, https://doi.org/10.3892/or.2018.6621.
75. Monteiro, F. L., Williams, C., and Helguero, L. A.
(2022) A Systematic Review to Define the Multi-Facet-
ed Role of Lysine Methyltransferase SETD7 in Cancer,
Cancers (Basel)., 14, 1414, https://doi.org/10.3390/can-
cers14061414.
76. Xie, Y., Zhao, J., Liang, Y., Chen, M., Luo, Y., Cui, X.,
Jiang,B., Peng,L., and Wang,X. (2019) MicroRNA-10b
controls the metastasis and proliferation of colorectal
cancer cells by regulating Krüppel-like factor 4, Artif.
Cells Nanomed., Biotechnol., 47, 1722-1729, https://
doi.org/10.1080/21691401.2019.1606006.
77. Qi, X.-T., Li, Y.-L., Zhang, Y.-Q., Xu, T., Lu, B., Fang,L.,
Gao, J.-Q., Yu, L.-S., Zhu, D.-F., Yang, B., He, Q.-J., and
Ying, M.-D. (2019) KLF4 functions as an oncogene in
promoting cancer stem cell-like characteristics in os-
teosarcoma cells, Acta Pharmacol. Sin., 40, 546-555,
https://doi.org/10.1038/s41401-018-0050-6.
78. Li, H., Wang, J., Xiao, W., Xia, D., Lang, B., Wang, T.,
Guo, X., Hu, Z., Ye, Z., and Xu, H. (2014) Epigenetic
inactivation of KLF4 is associated with urothelial can-
cer progression and early recurrence, J. Urol., 191,
493-501, https://doi.org/10.1016/j.juro.2013.08.087.
79. Xu, X., Li, J., Zhu, Y., Xie, B., Wang, X., Wang, S.,
Xie, H., Yan, H., Ying, Y., Lin, Y., Liu, B., Wang, W.,
and Zheng,X. (2017) CRISPR-ON-Mediated KLF4 over-
expression inhibits the proliferation, migration and
invasion of urothelial bladder cancer in vitro and
invivo, Oncotarget, 8, 102078-102087, https://doi.org/
10.18632/oncotarget.22158.
80. Xie, H., Li, J., Ying, Y., Yan, H., Jin, K., Ma, X., He, L.,
Xu,X., Liu, B., Wang, X., Zheng, X., and Xie,L. (2020)
METTL3/YTHDF2 m6A axis promotes tumorigene-
sis by degrading SETD7 and KLF4 mRNAs in blad-
der cancer, J. Cell. Mol. Med., 24, 4092-4410, https://
doi.org/10.1111/jcmm.15063.
81. Wang, J., Luo, J., Wu, X., and Li, Z. (2024) WTAP en-
hances the instability of SYTL1 mRNA caused by YT-
HDF2 in bladder cancer, Histol. Histopathol., 39, 633-
646, https://doi.org/10.14670/HH-18-671.
82. Riegman, M., Sagie, L., Galed, C., Levin, T., Stein-
berg, N., Dixon, S. J., Wiesner, U., Bradbury, M. S.,
Niethammer, P., Zaritsky, A., and Overholtzer, M.
(2020) Ferroptosis occurs through an osmotic mech-
anism and propagates independently of cell rupture,
Nat. Cell Biol., 22, 1042-1048, https://doi.org/10.1038/
s41556-020-0565-1.
83. An,W., Gupta,R., Zhai,K., Wang,Y., Xu,W., and Cui,Y.
(2024) Current and Potential Roles of Ferroptosis in
Bladder Cancer, Curr. Med. Sci., 44, 51-63, https://
doi.org/10.1007/s11596-023-2814-6.
84. Dodson, M., Castro-Portuguez, R., and Zhang, D. D.
(2019) NRF2 plays a critical role in mitigating lipid
SINYAGOVSKAYA et al.668
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
peroxidation and ferroptosis, Redox Biol., 23, 101107,
https://doi.org/10.1016/j.redox.2019.101107.
85. Koppula, P., Zhuang, L., and Gan, B. (2021) Cystine
transporter SLC7A11/xCT in cancer: ferroptosis, nutri-
ent dependency, and cancer therapy, Protein Cell, 12,
599-620, https://doi.org/10.1007/s13238-020-00789-5.
86. Ingold,I., Berndt,C., Schmitt,S., Doll,S., Poschmann,G.,
Buday, K., Roveri, A., Peng, X., Porto Freitas, F.,
Seibt, T., Mehr, L., Aichler, M., Walch, A., Lamp, D.,
Jastroch,M., Miyamoto,S., Wurst,W., Ursini,F., Arnér,
E. S. J., Fradejas-Villar, N., Schweizer, U., Zischka, H.,
Friedmann Angeli, J. P., and Conrad, M. (2018) Sele-
nium utilization by GPX4 is required to prevent hy-
droperoxide-induced ferroptosis, Cell, 172, 409-422.
e21, https://doi.org/10.1016/j.cell.2017.11.048.
87. de Souza, I., Monteiro, L. K. S., Guedes, C. B., Silva,
M. M., Andrade-Tomaz, M., Contieri, B., Latancia,
M. T., Mendes, D., Porchia, B. F. M. M., Lazarini, M.,
Gomes, L. R., and Rocha, C. R. R. (2022) High levels
of NRF2 sensitize temozolomide-resistant glioblasto-
ma cells to ferroptosis via ABCC1/MRP1 upregulation,
Cell Death Dis., 13, 591, https://doi.org/10.1038/s41419-
022-05044-9.
88. Wang, D., Tang,L., Zhang, Y., Ge, G., Jiang,X., Mo, Y.,
Wu, P., Deng, X., Li, L., Zuo, S., Yan, Q., Zhang, S.,
Wang, F., Shi, L., Li, X., Xiang, B., Zhou, M., Liao, Q.,
Guo, C., Zeng, Z., Xiong, W., and Gong, Z. (2022) Reg-
ulatory pathways and drugs associated with fer-
roptosis in tumors, Cell Death Dis., 13, 544, https://
doi.org/10.1038/s41419-022-04927-1.
89. Sun, X., Ou, Z., Chen, R., Niu, X., Chen, D., Kang, R.,
and Tang,D. (2016) Activation of the p62-Keap1-NRF2
pathway protects against ferroptosis in hepatocellu-
lar carcinoma cells, Hepatology, 63, 173-184, https://
doi.org/10.1002/hep.28251.
90. Li,T., Jiang,D., and Wu,K. (2020) p62 promotes blad-
der cancer cell growth by activating KEAP1/NRF2-de-
pendent antioxidative response, Cancer Sci., 111,
1156-1164, https://doi.org/10.1111/cas.14321.
91. Ma, J., Hu, J., Zhao, L., Wu, Z., Li, R., and Deng, W.
(2024) Identification of clinical prognostic factors
and analysis of ferroptosis-related gene signatures in
the bladder cancer immune microenvironment, BMC
Urol., 24, 6, https://doi.org/10.1186/s12894-023-01354-y.
92. Wang,K., Wang,G., Li,G., Zhang,W., Wang,Y., Lin,X.,
Han,C., Chen,H., Shi,L., Reheman,A., Li,J., Li,Z., and
Yang,X. (2023) m6A writer WTAP targets NRF2 to ac-
celerate bladder cancer malignancy via m6A-depen-
dent ferroptosis regulation, Apoptosis, 28, 627-638,
https://doi.org/10.1007/s10495-023-01817-5.
93. Chen, L., and Wang, X. (2018) Relationship between
the genetic expression of WTAP and bladder cancer
and patient prognosis, Oncol. Lett., 16, 6966-6970,
https://doi.org/10.3892/ol.2018.9554.
94. Gao, N., Li, Y., Li, J., Gao, Z., Yang, Z., Li, Y., Liu, H.,
and Fan,T. (2020) Long non-coding RNAs: the regula-
tory mechanisms, research strategies, and future di-
rections in cancers, Front. Oncol., 10, 598817, https://
doi.org/10.3389/fonc.2020.598817.
95. Qin, T., Li, J., and Zhang, K.-Q. (2020) Structure, reg-
ulation, and function of linear and circular long
non-coding RNAs, Front. Genet., 11, 150, https://
doi.org/10.3389/fgene.2020.00150.
96. Luo, H., Xu, C., Le, W., Ge, B., and Wang, T. (2019)
lncRNA CASC11 promotes cancer cell proliferation in
bladder cancer through miRNA-150, J. Cell. Biochem.,
120, 13487-13493, https://doi.org/10.1002/jcb.28622.
97. Zhang, Z., Zhou, C., Chang, Y., Zhang, Z., Hu, Y.,
Zhang,F., Lu,Y., Zheng,L., Zhang,W., Li,X., and Li,X.
(2016) Long non-coding RNA CASC11 interacts with
hnRNP-K and activates the WNT/β-catenin pathway
to promote growth and metastasis in colorectal can-
cer, Cancer Lett., 376, 62-73, https://doi.org/10.1016/
j.canlet.2016.03.022.
98. Huang,Y., Lv,Y., Yang,B., Zhang,S., Liu,B., Zhang,C.,
Hu, W., Jiang, L., Chen, C., Ji, D., Xiong, C., Liang, Y.,
Liu, M., Ying, X., and Ji, W. (2024) Enhancing m(6)A
modification of lncRNA through METTL3 and RBM15
to promote malignant progression in bladder can-
cer, Heliyon, 10, e28165, https://doi.org/10.1016/
j.heliyon.2024.e28165.
99. Han,J., Wang, J.Z., Yang,X., Yu,H., Zhou,R., Lu, H.C.,
Yuan, W.B., Lu, J.C., Zhou, Z.J., Lu,Q., Wei, J.F., and
Yang,H. (2019) METTL3 promote tumor proliferation
of bladder cancer by accelerating pri-miR221/222
maturation in m6A-dependent manner, Mol. Cancer,
18, 110, https://doi.org/10.1186/s12943-019-1036-9.
100. Xie,J., Zhang,H., Wang,K., Ni,J., Ma,X., Khoury, C.J.,
Prifti,V., Hoard,B., Cerenzia, E.G., Yin,L., Zhang,H.,
Wang, R., Zhuo, D., Mao, W., and Peng, B. (2023)
M6A-mediated-upregulation of lncRNA BLACAT3 pro-
motes bladder cancer angiogenesis and hematoge-
nous metastasis through YBX3 nuclear shuttling and
enhancing NCF2 transcription, Oncogene, 42, 2956-
2970, https://doi.org/10.1038/s41388-023-02814-3.
101. Liu,J., Tian,C., Qiao,J., Deng,K., Ye,X., and Xiong,L.
(2024) m6A Methylation-Mediated Stabilization of
LINC01106 Suppresses Bladder Cancer Progression by
Regulating the miR-3148/DAB1 Axis, Biomedicines, 12,
114, https://doi.org/10.3390/biomedicines12010114.
102. Yi,J., Ma,X., Ying,Y., Liu, Z., Tang,Y., Shu,X., Sun,J.,
Wu, Y., Lu, D., Wang, X., Luo, J., Liu, B., Zheng, X.,
Lin, Y., Li, J., and Xie, L. (2024) N6-methyladenosine-
modified CircPSMA7 enhances bladder cancer ma-
lignancy through the miR-128-3p/MAPK1 axis, Can-
cer Lett., 585, 216613, https://doi.org/10.1016/j.canlet.
2024.216613.
103. Liu,P., Fan,B., Othmane,B., Hu,J., Li,H., Cui,Y., Ou,Z.,
Chen,J., and Zu, X. (2022) M6A-induced lncDBET pro-
motes the malignant progression of bladder cancer
through FABP5-mediated lipid metabolism, Theranos-
tics, 12, 6291-6307, https://doi.org/10.7150/thno.71456.
m
6
A-RNA METHYLATION IN BLADDER CANCER DEVELOPMENT 669
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
104. Guimarães-Teixeira,C., Lobo,J., Miranda-Gonçalves,V.,
Barros-Silva, D., Martins-Lima, C., Monteiro-Reis, S.,
Sequeira, J.P., Carneiro,I., Correia, M.P., Henrique,R.,
and Jerónimo,C. (2022) Downregulation of m6A writ-
er complex member METTL14 in bladder urothe-
lial carcinoma suppresses tumor aggressiveness,
Mol. Oncol., 16, 1841-1856, https://doi.org/10.1002/
1878-0261.13181.
105. Zhang,N., Hua,X., Tu,H., Li,J., Zhang,Z., and Max,C.
(2021) Isorhapontigenin (ISO) inhibits EMT through
FOXO3A/METTL14/VIMENTIN pathway in blad-
der cancer cells, Cancer Lett., 520, 400-408, https://
doi.org/10.1016/j.canlet.2021.07.041.
106. Xie, M., and Zhang, H. (2024) METTL14-mediate the
biological effects of EMT in bladder Cancer cells by
methylating SOX4 mRNA with m6A, Indian J. Pharm.
Educ. Res., 58, 814-821, https://doi.org/10.5530/
ijper.58.3.89.
107. Lavallee, E., Sfakianos, J. P., and Mulholland, D. J.
(2021) Tumor heterogeneity and consequences for
bladder cancer treatment, Cancers (Basel)., 13, 5297,
https://doi.org/10.3390/cancers13215297.
108. Wang, C., Ma, D., Yu, H., Zhuo, Z., and Ye, Z. (2023)
N6-methyladenosine (m6A) as a regulator of car-
cinogenesis and drug resistance by targeting epithe-
lial-mesenchymal transition and cancer stem cells,
Heliyon, 9, e14001, https://doi.org/10.1016/j.heliyon.
2023.e14001.
109. Liu, W.-W., Zhang, Z.-Y., Wang,F., and Wang,H. (2023)
Emerging roles of m6A RNA modification in cancer
therapeutic resistance, Exp. Hematol. Oncol., 12, 21,
https://doi.org/10.1186/s40164-023-00386-2.
110. Liu, Z., Zou, H., Dang, Q., Xu, H., Liu, L., Zhang, Y.,
Lv, J., Li, H., Zhou, Z., and Han, X. (2022) Biological
and pharmacological roles of m(6)A modifications in
cancer drug resistance, Mol. Cancer, 21, 220, https://
doi.org/10.1186/s12943-022-01680-z.
111. Wang,L., Hui,H., Agrawal,K., Kang,Y., Li,N., Tang,R.,
Yuan, J., and Rana, T. M. (2020) m(6) A RNA methyl-
transferases METTL3/14 regulate immune responses
to anti-PD-1 therapy, EMBO J., 39, e104514, https://
doi.org/10.15252/embj.2020104514.
112. Ni, Z., Sun, P., Zheng, J., Wu, M., Yang, C., Cheng, M.,
Yin,M., Cui, C., Wang,G., Yuan,L., Gao,Q., and Li,Y.
(2022) JNK Signaling Promotes Bladder Cancer Im-
mune Escape by Regulating METTL3-Mediated m6A
Modification of PD-L1 mRNA, Cancer Res., 82, 1789-
1802, https://doi.org/10.1158/0008-5472.CAN-21-1323.
113. Yu,H., Yang,X., Tang,J., Si,S., Zhou,Z., Lu,J., Han,J.,
Yuan,B., Wu,Q., Lu,Q., and Yang,H. (2021) ALKBH5
inhibited cell proliferation and sensitized bladder
cancer cells to cisplatin by m6A-CK2α-mediated gly-
colysis, Mol. Ther. Nucleic Acids, 23, 27-41, https://
doi.org/10.1016/j.omtn.2020.10.031.
114. Zhang,X., Yang,X., Yang,C., Li,P., Yuan,W., Deng,X.,
Cheng, Y., Li, P., Yang, H., Tao, J., and Lu, Q. (2016)
Targeting protein kinase CK2 suppresses bladder can-
cer cell survival via the glucose metabolic pathway,
Oncotarget, 7, 87361-87372, https://doi.org/10.18632/
oncotarget.13571.
115. Xu,C., Zhou, J., Zhang, X., Kang, X., Liu, S., Song, M.,
Chang,C., Lin,Y., and Wang,Y. (2024) N(6)-methylad-
enosine-modified circ_104797 sustains cisplatin resis-
tance in bladder cancer through acting as RNA spong-
es, Cell. Mol. Biol. Lett., 29, 28, https://doi.org/10.1186/
s11658-024-00543-3.
116. Priem, D., Devos, M., Druwé, S., Martens, A.,
Slowicka,K., Ting, A. T., Pasparakis,M., Declercq, W.,
Vandenabeele, P., van Loo, G., and Bertrand, M. J. M.
(2019) A20 protects cells from TNF-induced apoptosis
through linear ubiquitin-dependent and -indepen-
dent mechanisms, Cell Death Dis., 10, 692, https://
doi.org/10.1038/s41419-019-1937-y.
117. Wei, W., Sun, J., Zhang, H., Xiao, X., Huang, C.,
Wang,L., Zhong,H., Jiang,Y., Zhang,X., and Jiang,G.
(2021) Circ0008399 Interaction with WTAP promotes
assembly and activity of the m(6)A methyltransfer-
ase complex and promotes cisplatin resistance in
bladder cancer, Cancer Res., 81, 6142-6156, https://
doi.org/10.1158/0008-5472.CAN-21-1518.
118. Yu, H., Zhuang, J., Zhou, Z., Song, Q., Lv, J., Yang, X.,
Yang, H., and Lu, Q. (2024) METTL16 suppressed
the proliferation and cisplatin-chemoresistance of
bladder cancer by degrading PMEPA1 mRNA in
a m6A manner through autophagy pathway, Int.
J. Biol. Sci., 20, 1471-1491, https://doi.org/10.7150/
ijbs.86719.
119. Meyer, K. D., Saletore, Y., Zumbo, P., Elemento, O.,
Mason, C.E., and Jaffrey, S. R. (2012) Comprehensive
analysis of mRNA methylation reveals enrichment in
3’ UTRs and near stop codons, Cell, 149, 1635-1646,
https://doi.org/10.1016/j.cell.2012.05.003.
120. Dominissini,D., Moshitch-Moshkovitz,S., Schwartz,S.,
Salmon-Divon,M., Ungar,L., Osenberg,S., Cesarkas,K.,
Jacob-Hirsch, J., Amariglio, N., Kupiec, M., Sorek, R.,
and Rechavi, G. (2012) Topology of the human and
mouse m6A RNA methylomes revealed by m6A-
seq, Nature, 485, 201-206, https://doi.org/10.1038/
nature11112.
121. Bayoumi, M., and Munir, M. (2021) Evolutionary
conservation of the DRACH signatures of potential
N6-methyladenosine (m(6)A) sites among influenza
A viruses, Sci. Rep., 11, 4548, https://doi.org/10.1038/
s41598-021-84007-0.
122. Liu,Z., and Zhang,J. (2018) Most m6A RNA modifica-
tions in protein-coding regions are evolutionarily un-
conserved and likely nonfunctional, Mol. Biol. Evol.,
35, 666-675, https://doi.org/10.1093/molbev/msx320.
123. An,M., Wang,H., and Zhu,Y. (2020) Mutations in m6A
consensus motifs are suppressed in the m6A modified
genes in human cancer cells, PLoS One, 15, e0236882,
https://doi.org/10.1371/journal.pone.0236882.
SINYAGOVSKAYA et al.670
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
124. Deng, N., Zhou, H., Fan, H., and Yuan, Y. (2017) Sin-
gle nucleotide polymorphisms and cancer suscepti-
bility, Oncotarget, 8, 110635-110649, https://doi.org/
10.18632/oncotarget.22372.
125. Lv,J., Song,Q., Bai,K., Han,J., Yu,H., Li,K., Zhuang,J.,
Yang,X., et al. (2022) N6-methyladenosine-related sin-
gle-nucleotide polymorphism analyses identify onco-
gene RNFT2 in bladder cancer, Cancer Cell Int., 22,
301, https://doi.org/10.1186/s12935-022-02701-z.
126. Zhang,C., Tunes,L., Hsieh, M.-H., Wang,P., Kumar,A.,
Khadgi, B. B., Yang, Y., Doxtader, K. A., Herrell, E.,
Koczy, O., Setlem, R., Zhang, X., Evers, B., Wang, Y.,
Xing, C., Zhu, H., and Nam, Y. (2023) Cancer muta-
tions rewire the RNA methylation specificity of
METTL3-METTL14, bioRxiv Prepr. Serv. Biol., 10,
eads4750, https://doi.org/10.1101/2023.03.16.532618.
127. Waldbillig, F., Bormann,F., Neuberger,M., Ellinger,J.,
Erben,P., Kriegmair, M.C., Michel, M.S., Nuhn,P., and
Nientiedt,M. (2023) An m6A-driven prognostic mark-
er panel for renal cell carcinoma based on the first
transcriptome-wide m6A-seq, Diagnostics, 13, 823,
https://doi.org/10.3390/diagnostics13050823.
128. Zhang, G., Yang, J., Fang, J., Yu, R., Yin, Z., Chen, G.,
Tai, P., He, D., Cao, K., and Jiang, J. (2024) Develop-
ment of an m6A subtype classifier to guide precision
therapy for patients with bladder cancer, J. Cancer,
15, 5204-5217, https://doi.org/10.7150/jca.99483.
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.