Skip to main content
Erschienen in: BMC Cancer 1/2024

Open Access 01.12.2024 | Review

The role of the methyltransferase METTL3 in prostate cancer: a potential therapeutic target

verfasst von: Xuming Zhou, Keqiang Chai, Hezhen Zhu, Cong Luo, Xiaofeng Zou, Junrong Zou, Guoxi Zhang

Erschienen in: BMC Cancer | Ausgabe 1/2024

Abstract

The incidence of prostate cancer (PCa), the most prevalent malignancy, is currently at the forefront. RNA modification is a subfield of the booming field of epigenetics. To date, more than 170 types of RNA modifications have been described, and N6-methyladenosine (m6A) is the most abundant and well-characterized internal modification of mRNAs involved in various aspects of cancer progression. METTL3, the first identified key methyltransferase, regulates human mRNA and non-coding RNA expression in an m6A-dependent manner. This review elucidates the biological function and role of METTL3 in PCa and discusses the implications of METTL3 as a potential therapeutic target for future research directions and clinical applications.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Prostate cancer (PCa) is one of the most prevalent malignant tumors in the world, with the second-highest incidence rate after lung cancer and the eighth-highest mortality rate. It is also a leading cause of cancer-related deaths in men. The incidence rates of PCa have substantial geographical and ethnic differences. Australia/New Zealand, North America, and Europe have a higher incidence rate of 85/100,000, while Asia has the lowest incidence rate of 4.5/100,000–10.5/100,000 [14]. There is no standard treatment protocol for high-risk patients because of the highly aggressive nature of tumors and the complex tumor microenvironment in high-risk patients; androgen deprivation therapy (ADT) is often combined with novel endocrine therapy [58]. Therefore, there is an urgent need to explore the mechanisms of PCa development and therapeutic targets. The rapid development of epigenetics in recent years has provided new avenues to search for the mechanisms of PCa progression, metastasis, and potential therapeutic targets. N6-methyladenosine (m6A), a well-known post-transcriptional modification first identified in 1974, is thought to be the most frequent internal modification in mammalian mRNAs and also occurs in small ncRNAs and lncRNAs in eukaryotic species [912]. This modification is enriched in 3′ UTRs, near-stop codons, long inner exons, intergenic regions, introns, and 5′ UTRs [13, 14]. m6A methyltransferases mainly comprise METTL3, METTL5, METTL14, METTL16, RBM15, WTAP, VIRMA, and ZCCHC “writer” proteins. METTL3 is the only subunit that binds to the methyl donor S-adenosylmethionine (SAM) and catalyzes the methyltransferase domain responsible for converting adenosine to m6A (See Fig. 1) [1518]. Furthermore, there are “erasers” proteins with demethylation capabilities, including FTO and ALKBH5 [19, 20], and unique protein binding recognition elements “readers,” including YTHDC1/2 and YTHDF1/2/3 [21, 22].
Studies have demonstrated that METTL3 expression is upregulated in various tumors, including breast, lung, liver, stomach, colorectal, and pancreatic cancers [2328].In PCa, METTL3 upregulation appears to play an important role. METTL3 expression is upregulated in PCa cell lines, and METTL3 knockdown induces apoptosis in cancer cells [29].METTL3 upregulation is also associated with poor prognosis in PCa patients, and METTL3 expression is upregulated in PCa tissues, particularly bone metastases [30, 31].These studies suggest that exploring the specific mechanisms of METTL3 in PCa genesis and metastasis through m6A modification helps us gain a deeper understanding and that identifying and targeting these essential genes involved in PCa metastasis play a key role in the future treatment of metastatic PCa. The influence of current research findings on the clinical translation of PCa and whether they can contribute to the clinical treatment of PCa are discussed in the current study.

m6A modifications

To date, more than 100 types of RNA chemical modifications have been identified for modifying coding and non-coding RNAs [32].As research on these modifications is emerging, they have significantly affected human diseases [3335]. m6A is the most abundant and well-characterized internal modification of mRNAs, which regulates self-renewal in embryonic stem and cancer cells and facilitates cell survival after heat shock or DNA damage [3638]. In addition to their role in mRNAs, m6A modifications are also present in non-coding RNAs, such as miRNAs, lncRNAs, and circRNAs, which regulate their biological functions [3944]. RNA modifications precisely regulate the biological functions of numerous molecules, diversifying genetic information. A protein group has been identified, thereby influencing the outcome of RNA [45].We referred to these proteins that specifically deposit, remove, and recognize RNA as “writer,” “eraser,” and “reader” proteins, respectively (See Table 1 and Fig. 2).
Table 1
m6A modification-related factors
Type
Factors
Function
Ref.
Writer
METTL3/14, WTAP, VIRMA, RBM15
It catalyzes the m6A modification of adenylate on mRNA
[4648]
 
METTL16
Methylated snRNA, pre-mRNA, and ncRNA
[4952]
 
ZC3H13
Connecting WTAP and RBM15
[53]
Eraser
FTO
Demethylation of m6A
[5456]
 
ALKBH5
Demethylation of m6A
[57]
Reader
YTHDC1
Alternative splicing and RNA export
[21]
 
YTHDC2
mRNA degradation and translation initiation
[22]
 
YTHDF1
Promote translation and RNA degradation
[58]
 
YTHDF2
Promotes RNA degradation
[58]
 
YTHDF3
Promotes mRNA translation and degradation
[58]
 
IGFBP1/2/3
Promotes RNA stability
[59]
 
HnRNPG/C
Regulation of mRNA splicing
[60]

m6A writers

During transcription, m6A is deposited in the nascent pre-mRNAs by the methyltransferase complex in the nucleus. These complexes are composed of METTL3, METTL14, METTL16, RBM15, WTAP, VIRMA, and ZC3H13 [4648, 61].Among them, METTL3 catalyzes the conversion of adenosine to m6A through its methyltransferase domain. METTL14 is responsible for recognizing RNA substrates, RBM15 is responsible for the initial recruitment of the complex at target sites in mRNA, and WTAP and VIRMA are responsible for complex formation. ZC3H13 acts as a linker between the aptamers RBM15 and WTAP. METTL16 mainly methylates snRNA, some intron sites of pre-mRNA, in addition to other ncRNAs [44, 4953, 62].

m6A erasers

Methylation modification of m6A is reversible and involves the combined participation of methyltransferases, demethyltransferases, and methylated reading proteins. The identified m6A demethylases include FTO and ALKBH5 [2, 19, 20, 63, 64]. FTO proteins are similar to members of the ALKB protein family in their core structural domains. However, the unique long loop at the C-terminus differs from that of the ALKB protein family, and this unique structural domain allows FTO proteins to demethylate single-stranded DNA or RNA that undergoes methylation modification [5456].Once the transcriptional level of the FTO gene is abnormal, it can cause many diseases [65].ALKHB5 is another important demethylase that demethylates mRNA in the nucleus, with an alanine-rich region at the N-terminal end and a unique coiled-coil structure [57].The level of m6A modification in mRNA was significantly elevated after ALKBH5 knockdown in cell lines [66].

m6A readers

A specific RNA-binding protein is required for m6A-modified mRNA to perform specific biological functions [67, 68].Various “reader” proteins have been identified, including YTH structural domain proteins such as YTHDC1, YTHDC2, YTHDF1, YTHDF2, YTHDF3, hnRNPC, hnRNPG, IGF2BP1, IGF2BP2, and IGF2BP3 [21, 22, 39, 5860, 69, 70].The main functions of these “reader” proteins include regulating RNA stability, translation efficiency, RNA splicing, and RNA export [67, 7178].

Expression of writers, erasers, and readers in prostate cancer

The biological functions of METTL3 upregulation in PCa play an important role in cancer progression. METTL3 expression is upregulated in PCa cell lines, and knockdown of METTL3 induces apoptosis in cancer cells [29]. METTL3 upregulation was also associated with poor prognosis in PCa patients, and its expression was upregulated in PCa tissues, especially in bone metastases [30, 31]. Among other methyltransferase components, high VIRMA expression may be associated with poor PCa prognosis [79]. It was also found that METTL14 promotes PCa proliferation in an m6A-dependent manner by inhibiting THBS6 expression, a glycoprotein that inhibits angiogenesis [80].
The discovery of FTO and ALKBH5 demonstrates that RNA modifications are reversible. The current study shows that FTO is commonly downregulated in PCa tissues and cell lines and that patients with lower FTO expression have a more advanced tumor stage as well as higher Gleason scores [81, 82]. Li et al. found that FTO inhibited PCa progression by downregulating melanocortin receptor 4 (MC4R) expression [83]. A recent study showed that ALKBH5 expression is downregulated in PCa tissues and inhibits the growth of PCa cell lines [84]. Overall, studies on m6A erasers in PCa are limited and require further exploration.
m6A readers also play an important role in the progression of prostate cancer. YTHDC1 was found to bind and co-localize with the oncogene MET adhesin in subnuclear patches and affect PCa proliferation [85]. YTHDC2 expression was upregulated in PCa tissues and cell lines and was significantly correlated with PSA levels and Gleason scores, while YTHDC2 overexpression promoted proliferation and invasion in PCa cell lines [86]. YTHDF1/2 is overexpressed in PCa, and PLK1, a key factor in the cell cycle, is a direct target of YTHDF1 in PCa cells. ELK1-activated YTHDF1 controls PLK1 translation efficiency in an m6A-dependent manner, enabling the activation of the PI3K/AKT signaling pathway, leading to prostate cancer progression. yTHDF1 can also contribute to prostate cancer progression by regulating TRIM44 to promote PCa cell proliferation and migration [87, 88]. In contrast, YTHDF2 leads to PCa progression by mediating the degradation of the tumor suppressors LHPP and NKX3–1 and activating the AKT signaling pathway [89]. YTHDF2 is also a direct target of miR - 495 and miR - 493 - 3p. On the lysine demethylase 5a (KDM5a)/miRNA495/YTHDF2/m6AMOB3b axis, YTHDF2 recognizes m6A of MOB3b mRNA, induces MOB3b mRNA degradation and suppresses its expression. miR - 493 - 3p suppresses YTHDF2 expression, thereby increasing the level of m6A [90, 91]. Therefore, high expression levels of YTHDF2 promote the proliferation, migration, and invasion of PCa cells. hnRNPA2B1 is highly expressed in CRPC cells and promotes proliferation, leading to a worse prognosis of PCa [92]. Regarding bone metastasis, IGF2BP2 promotes PCAT6 upregulation in an m6A-dependent manner. In addition, PCAT6 enhances IGF1R mRNA stability via the PCAT6/IGF2BP2/IGF1R RNA-protein trimer, thereby upregulating IGF1R expression and promoting PCa bone metastasis and tumor growth [93]. Clinical case studies have revealed that IGF2BP3 is associated with infiltrative tumor recurrence [94]. IGF2BP3 also binds cyclic RNA hsa_circ_0003258 in the cytoplasm, enhances the stability of HDAC4 mRNA, activates the ERK pathway, and triggers EMT to accelerate PCa metastasis [95].

METTL3 structure and function

The human METTL3 gene is located in the 14q11.2 region of the chromosome and contains 580 amino acids. METTL3 is the only catalytic subunit in the entire complex, whereas METTL14 has no enzymatic activity because of its closed conformation of the catalytic structure and inability to bind SAM [96, 97].METTL3, the only catalytic subunit, contains the leader helix (LH), nuclear localization signal (NLS), CCCH-type zinc finger domain (ZFD), and SAM structure-binding domain-containing methyltransferase domain (MTD) (See Fig. 3) [49, 98101]. LH and NLS enable METTL14 to bind to METTL3 and synergize with WTAP’s functional NLS to mediate the nucleation of the methyltransferase complex and play an overall methyltransferase role [102].The species-conserved ZnF1 and ZnF2 sequences form the ZFD CCCH-type zinc-finger structural domain. In the absence of ZnF1, the heterodimer formed by METTL3-METTL14 is inactivated, and ZFD can act as a target recognition domain to specifically bind RNA containing the 5′-GGACU-3′ shared sequence [99]. Zinc fingers are responsible for RNA-specific recognition, enabling METTL3 to exert its methyltransferase activity. MTD mediates the METTL3-METTL14 interaction for the binding domain of bound SAM [49].It also possesses several phosphorylation sites in METTL3, including S2, S43, S48, S50, S219, S243, T348, and S350. A comparative analysis of METTL3 from different species revealed that S2, S43, S48, S50, S219, and S243 are conserved in vertebrates but not Drosophila. s350 is conserved in mammals but absent in Drosophila and zebrafish, whereas T348 is not conserved [102].The post-translational phosphorylation modification of METTL3 enables METTL3 to form complexes with other proteins and to be functional.

Regulation of METTL3 expression and m6A deposition

METTL3 expression in cancer cells is regulated by various mechanisms (See Fig. 4). Wang et al. [103] found that P300 mediates histone H3 lysine 27 acetylation (H3K27ac) and promotes METTL3 transcription in gastric cancer. In pancreatic cancer, cigarette smoke condensates induce hypomethylation of the METTL3 promoter, which subsequently recruits the transcription factor NFIC for overexpression [27]. In addition, non-coding RNAs can participate in tumor progression by regulating METTL3 expression. miRNAs are specific transcription factors that regulate METTL3 and can reduce the expression and function of METTL3, thereby altering the tumor-promoting effects of METTL3 [23, 28, 104]. lncRNAs are also involved in the regulation of METTL3. For example, LINC00470 interacts with METTL3 to promote PTEN mRNA degradation, promoting gastric cancer (GC) progression [105]. In addition, lncRNA RHO GTPase-activating kinase 5 (ARHG AP5)-AS1 recruits METTL3 to enhance the stability of AR HGAP5 mRNA, leading to poor prognosis and chemotherapy resistance in GC [106]. It has also been found that SUMOylation of METTL3 inhibits its methyltransferase activity without affecting the stability of the protein [107].
m6A deposition can be regulated by modifying the sequence number and structure of the sites, but the specific mechanism requires further study [108]. It has been shown that histone H3 lysine 36 trimethylation (H3K36me3) can regulate m6A modification by directly interacting with METTL14 and recruiting MTC complexes, leading to selective deposition of m6A in the CDS and 3′ UTR [109]. Zinc finger protein 217 (ZFP217) blocks METTL3 and inhibits m6A deposition on stem-related transcripts [110]. In addition, SMAD family member 2(SMAD2/3) recruits the METTL3/14 complex to a population of transcripts involved in early cell fate decisions [111]. Another transcription factor, CAATT-box-binding protein (CEBPZ), directly recruits METTL3 to chromatin [111]. AN et al. performed a large-scale computer screen to identify cell-specific trans-regulators of m6A and found that TRA2A and CAPRIN1 interact with METTL3 [112]. Fish et al. found that the RNA-binding protein TARBP2 recruits METLL3 and deposits m6A on introns of target mRNAs, thereby regulating RNA splicing and stabilization [113].

METTL3 is involved in PCa

Studies have demonstrated that METTL3 is involved in various progressive processes in PCa, including proliferation, migration, apoptosis, drug resistance, and maintenance of glycolipid metabolism. The latest findings on METTL3 in PCa are summarized in the following sections (See Fig. 5).

Role of METTL3 in PCa proliferation

The infinite proliferation and anti-apoptotic behavior of cancer are essential reasons for its development, which is also the main characteristic of cancer [114]. METTL3 promotes cancer cell proliferation and anti-apoptosis by regulating various targets and pathways, including miRNAs and non-coding RNAs [115119], which are essential for PCa. METTL3 can induce methylation of pri-miRNA in mammalian cells, and the tagged pri-miRNA can be recognized and processed by the double-stranded RNA-binding protein DGCR8 [40]. Wang et al. [120] found that METTL3 is necessary for DGCR8 to regulate pri-miRNAs in PCa, and their experiments demonstrated that m6A modification-dependent METTL3 could interact with DGCR8 to enhance the recognition of pri-miR-182 in PCa, promote the maturation of pri-miRNAs, and lead to PCa. METTL3 can also promote PCa progression by mediating the m6A modification of KIF3C mRNA [121]. A study on the metabolic effects of METTL3 on long-stranded non-coding RNAs found that METTL3-mediated m6A-modified lncRNA MALAT1 promoted PCa proliferation by activating the PI3K/AKT signaling pathway and that METTL3-mediated lncRNA PVT1 could regulate the miR-27b-3b/BLM signaling pathway [122, 123]. Abnormal METTL3 expression can also cause abnormal activation of proto-oncogenes to exert oncogenic effects. For instance, METTL3-mediated m6A modification in PCa can lead to abnormal expression of Myc mRNA and promote PCa proliferation [30]. Overall, METTL3 regulates the stabilization, metabolism, and maturation of miRNAs and ncRNAs and plays a crucial role in PCa proliferation.

Role of METTL3 in PCa migration and invasion

One of the main characteristics of cancer is its invasion and distant metastasis [114, 124, 125]. The leading cause of death in PCa is late, unavoidable distant metastasis [126].m6A modification can recruit specific “reader” proteins to regulate mRNA processing, stabilization, and translation [127, 128]. YTHDF2—the first identified m6A “reader” protein—can regulate mRNA degradation and cell viability [129131]; the binding site of m6A is located in the 3′UTR of mRNA [129]. YTHDF2 can specifically bind to mRNA with m6A methylation marks at the 5′ UTR to promote protein translation [132]. Li et al. demonstrated that METTL3 is frequently upregulated in PCa as an upstream cooperating factor for YTHDF2. Analyses of MeRIP-seq, mRNA-seq, and databases have identified LHPP and NKX3–1 as the main targets of YTHDF2, and both LHPP and NKX3–1 are tumor suppressors that regulate tumor progression by inhibiting AKT phosphorylation [133136]. YTHDF2 directly binds to m6A-containing LHPP and NKX3–1 to induce mRNA decay. The mechanism may be that YTHDF2 induces the degradation of tumor suppressors LHPP and NKX3–1 to upregulate p-PKA and promote PCa progression by binding to METTL3-mediated m6A sites [89]. Cai et al. also observed elevated levels of METTL3 in PCa cells, which promoted PCa growth by regulating the hedgehog pathway [29]. In the Wnt pathway, METTL3 can affect Wnt/β-catenin signaling through m6A methylation of LEF1 mRNA to promote PCa proliferation and migration [137].
Two essential phenomena in developing cancer metastasis are epithelial-mesenchymal transition (EMT) and cancer cell migration [138140]. Chen et al. [141] reported that METTL3 could directly affect the expression of ARHGDIA, a key migration-related protein that regulates the growth, migration, and polarity of tumor cells [142]. Ectopic expression of ARHGDIA effectively attenuated the effect of METTL3 knockdown on the invasive ability of PCa cells, and that METTL3 promoted PCa metastasis by upregulating ARHGDIA expression through m6A modifications.
Remarkably, these studies focused on METTL3 regulation of downstream genes to promote PCa progression. However, whether METTL3 directly affects molecular studies related to proliferation and migration has received less attention and warrants further investigation. Taken together, these findings suggest that METTL3 plays an essential role as an oncogene in PCa progression and metastasis.

METTL3 promotes PCa bone metastasis

Bone metastases from PCa are the main cause of death in PCa patients, and the overall 5-year survival rate for patients with bone metastases is approximately 30%, with many serious complications, such as bone pain, spinal cord compression, and pathological fractures, affecting the quality of life and survival rate of patients [143, 144]. However, the mechanism underlying PCa bone-specific metastasis remains unclear, and the role of m6A modification in bone metastasis may provide new insights. Lang et al. identified a novel molecular mechanism of bone metastasis in which METTL3-mediated m6A modification promotes PCAT6 upregulation in an IGF2BP2-dependent manner. Furthermore, PCAT6 enhances IGF1R mRNA stability via the PCAT6/IGF2BP2/IGF1R RNA-protein trimer, thereby upregulating IGF1R expression and promoting PCa bone metastasis and tumor growth [93]. METTL3 increased the mRNA level of the adhesion molecule ITGB1 and adhesion to bone marrow stromal type I collagen in PCa cells through m6A modification, thereby increasing the possibility of bone metastasis in PCa [31].

Role of METTL3 in PCa glucose metabolism

As a hallmark of cancer, metabolic reprogramming maximizes energy consumption and production, contributing to tumor growth, invasion, and metastasis [145, 146]. Glycolysis is the preferred pathway for cancer cells to obtain energy, but glycolysis is not a hallmark of primary PCa and only plays a key role in advanced tumors [146148]. METTL3 enhances the stability of SNHG7 and recruits SRSF1 to regulate c-Myc expression by regulating m6A modification of SNHG7, further promoting glycolysis in PCa cells [149]. Current METTL3 research on glucose metabolism in cancer mainly focuses on gastrointestinal tumors, including gastric, liver, and colorectal cancers [103, 150, 151]. Further research is required to determine whether METTL3 acts as a regulator of PCa to target other related molecules that affect glucose metabolism.

Role of METTL3 in PCa drug-resistance

Clinical resistance to PCa is mainly noted in metastatic depot-resistant PCa (mCRPC), and resistance to late treatment with enzalutamide and abiraterone, second-generation androgen receptor (AR) inhibitors, cannot be avoided [152, 153].Low levels of METTL3 are associated with dysregulation of AR signaling and render PCa cells resistant to AR inhibitors in an AR-independent manner via upregulation of NR5A2 [154].This finding suggests that METTL3-mediated m6A modification may regulate the therapeutic sensitivity of AR inhibitors and that patients with varying METTL3 expression levels may respond differently to AR inhibitors, which warrants further investigation.

Targeting of METTL3 for potential clinical application

METTL3 plays a crucial role in cancer progression, and METTL3 inhibition has attracted the attention of pharmaceutical companies. The research and development of m6A-modified inhibitors as therapeutic targets is receiving increasing attention [155].Based on the multiple roles of METTL3, targeting METTL3 may offer new hope for individualized tumor treatment.

Nucleoside METTL3 inhibitors

Most studies on METTL3 inhibitors are still in the early stages, are limited in number, and are mainly divided into two types: nucleoside and non-nucleoside analogs. Bedi et al. [156] identified an N-substituted amide adenosine analog of ribonucleic acid as a potent METTL3 inhibitor after screening 4000 adenosine partial analogs and derivatives of SAM by in silico high-throughput docking. This binding mode was validated by protein crystallography and demonstrated a good ligand efficiency. However, its anticancer effects have not yet been tested.

Non-nucleoside METTL3 inhibitors

As adenosine analogs are less cell-permeable and less binding, non-nucleoside-selective METTL3 inhibitors can compensate for these disadvantages. UZH1a is a high-nanomolar inhibitor discovered through protein structure-based optimization and potency assessment of compounds in HTRF. UZH1a inhibits the activity of METTL3 by occupying its SAM-binding site [157], resulting in a dependent decrease in mRNA m6A methylation levels in leukemic MOLM-13 cells, osteosarcoma U2OS cells, and human embryonic kidney immortalized cells [157].Dolbois et al. optimized the UZH1a analog to obtain a more potent METTL3 inhibitor—UZH2—with a decrease of m6A in UZH2 polyadenylated RNA of 0.7 and 2.5 mM in MOLM-13 and PC-3 cell lines, respectively. For other m6A “writer” proteins, RNA methyltransferases were selectively probed, and no off-target was found [158].
STM2457 is a non-nucleoside METTL3 inhibitor developed for treating hematologic malignancies. Yankova et al. identified STM1760 as a non-SAM-related analog through high-throughput screening of 250,000 drug compounds, including pharmacodynamic optimization and ex vivo studies to obtain STM2457. STM2457 has been revealed to block proliferation and colony formation in MOLM-13 cell lines, promote apoptosis, and not affect normal hematopoietic function. Regarding In vivo studies, STM2457 inhibited the proliferation of acute myeloid leukemia (AML) in patient-derived xenograft and leukemia mouse models [159].

Oral small molecule inhibitors

Rosenfeld et al. invented STC-15, a novel oral small-molecule inhibitor of METTL3. In preclinical cancer models, STC-15 treatment results in the activation of innate immune pathways, inhibition of tumor growth, and enhancement of the anti-tumor properties of anti-PD-1 therapy to produce a durable anti-tumor immune response [160].

Diagnostic and prognostic biomarkers

METTL3 plays a key role in many biological processes, particularly tumorigenesis and development. In most cases, METTL3 functions as an oncogene in cancers. This causes alterations in mRNA translation, leading to tumor progression. METTL3 expression is higher in many tumor tissues than in normal tissues. Therefore, it is a potential clinical diagnostic and prognostic biomarker of cancer. In most cancers, high METTL3 expression predicts a poor prognosis. Examples include osteosarcoma [161], glioblastoma [162, 163], gastric cancer (GC) [26, 103, 162, 164, 165], colorectal cancer (CRC) [166], ovarian cancer [167], bladder cancer (BCa) [168], and pancreatic cancer [169]. In contrast, METTL3 is usually associated with tumor drug resistance [106, 170172]. METTL3-induced chemoresistance has been detected in several tumors, suggesting that functional inhibition of METTL3 may restore tumor chemosensitivity [156]. Furthermore, the knockdown of METTL3 could enhance the efficacy of anti-PD-1 therapy by activating the IFN signaling pathway [173]. Therefore, METTL3 is expected to be a novel target for tumor-targeted therapy.

Discussion

Most cancer studies on the role of METTL3 have focused on regulating the oncogenic effects of its downstream factors, whereas upstream regulators that lead to abnormal METTL3 expression have received little attention. The following studies have suggested that histone modifications and ncRNAs may play regulatory roles. In bladder cancer, activated JNK signaling is associated with increased METTL3 expression, and JNK knockdown impairs the binding of c-Jun to the METTL3 promoter, thereby reducing RNA m6A expression levels [174].In pancreatic cancer, a model of smoke condensate-induced malignant transformation of pancreatic ductal epithelial cells demonstrated that smoke condensate-induced METTL3 promoter hypermethylation leads to elevated METTL3 levels [27]. In gastrointestinal tumors, miR-4429 has been reported to reduce METTL3 expression in gastric cancer [26], and another study reported that HOXA10 increases Smad2/3 expression in the nucleus and promotes METTL3 deposition to regulate the progression of EMT in gastric cancer [175]. However, most studies in this area have focused on gastrointestinal tumors, and the cause of abnormal METTL3 expression in PCa remains unclear. These findings suggest that these upstream regulators can also affect METTL3 expression, leading to tumor progression. Studying the epigenetic modifications that cause abnormal METTL3 expression would help us better understand the biological functions of METTL3 in PCa.
m6A modifications have various biological functions in developing various types of cancer. METTL3, a crucial regulator of m6A, has been widely studied [176]. METTL3 is involved in all aspects of tumor progression, including cancer cell proliferation, migration, invasion, apoptosis, metastasis, angiogenesis, drug resistance, glycolipid metabolism, and tumor stem cell maintenance [177].Recent METTL3 studies in PCa have addressed biological functions, including proliferation, migration, invasion, metastasis, drug resistance, and glucose metabolism (See Table 2), whereas tumor angiogenesis, lipid metabolism, and tumor stem cell maintenance have hardly been investigated (See Fig. 6). Other cancer studies in this area have reported that METTL3 mediates the upregulation of the mRNA levels of tumor angiogenesis-related cytokines and angiogenic factors. The stability of ncRNAs leads to tumor angiogenesis in an m6A-dependent manner [103, 178180]. METTL3-mediated m6A methylation renders the mRNA of lipid metabolism-related genes unstable and affects downstream lipid accumulation [181].This instability may lead to dysregulation of lipid metabolism and facilitate tumor cell growth and immune escape. m6A mRNA modification is essential for cancer stem cell self-renewal and tumor metastasis, enhancing the frequency of tumor stem cell self-renewal, and cancer cell genesis and initiation by promoting the expression of SOX2 mRNA, a cancer stem cell marker [182184]. In summary, whether METTL3 is involved in tumor angiogenesis, lipid metabolism, and tumor stem cells in PCa is a worthy target for investigation, which will help us better understand the specific mechanisms of PCa genesis and metastasis.METTL3 plays a crucial role in the progression of PCa, suggesting that it may be a promising molecular biomarker for clinical diagnosis and prognostic relevance. Ji et al. reported that the overexpression of m6A methylation regulators resulted in a worse survival benefit for patients with high levels of mRNA methylation by affecting the subcellular localization of proteins in PCa [185].METTL3 is also associated with higher tumor stage and poorer prognosis in PCa [29],although more studies are required to demonstrate its feasibility.
Table 2
Roles of METTL3 in PCa
Target
Signaling pathways
Mechanism
Biological functions
Ref.
PVT1
miR-27b-3b/BLM
Enhance expression of PVT1 mRNA
Proliferation of PCa
[142]
MALAT
PI3K/AKT
Enhance expression of MALAT mRNA
Proliferation of PCa
[141]
KIF6C
IGF2BP3/KIF3C
Enhance KIF6C mRNA stability
Proliferation of PCa
[140]
DGCR8
Wnt/β-catenin
Promote the maturation of pri-miR-182
Proliferation of PCa
[139]
Myc
 
Activate Myc
Proliferation of PCa
[30]
ARHGDIA
 
EMT
Migration and invasion
[132]
LEF1
Wnt/β-catenin
Enhance expression of LEF1 mRNA
Migration and invasion
[128]
YTHDF2
cAMP
Inhibition expression of LHPP mRNA and NEX3–1 mRNA
Migration and invasion
[125]
GLI1
c-Myc/Cyclin D1
Enhance expression of GLI1 mRNA
Migration and invasion
[29]
IGFBP2
PCAT6/IGF2BP2/IGF1R
Enhance expression of PCAT6 mRNA, Enhance IGF1R stability
Bone metastasis
[93]
IGFBP1
 
Enhance expression of ITGB1 mRNA
Bone metastasis
[31]
SNHG7
c-Myc
Enhance SNHG7 mRNA stability, recruit SRSF1
Glycolysis
[149]
NR5A2
 
Enhance expression of NR5A2 mRNA
Drug-resistance
[154]
The androgen receptor (AR) plays a crucial role in PCa pathogenesis, and METTL3 may play a functional role. Roy et al. [186] found that METTL3 expression was higher in AR-expressing PCa cell lines than in AR-negative PCa cell lines, and similar findings were observed at the protein level. This finding suggests a potential interaction between METTL3, which is elevated at the onset of PCa, and androgen signaling. Notably, the expression of the AR target gene NKX3.1 was increased after METTL3 knockdown, whereas the expression of prostate-specific antigen (PSA) decreased, suggesting a direct role of METTL3 in AR expression [186]. METTL3 knockdown also leads to the elevation of key regulatory factors, such as KDM1A, which is involved in PCa initiation and progression and regulates AR expression and function [187189]. Further studies on the effect of METTL3 deletion on overall androgen signaling are needed. Because of the role of m6A methylation in the splicing process [190, 191], future research must investigate whether METTL3 functions in the progression of PCa to CRPC because of the AR splicing process.
METTL3 has an oncogenic function in most cancers but has also been shown to be a tumor suppressor in some cases [192]. For example, lower METTL3 expression was detected in renal cell carcinoma (RCC) tissues, suggesting that higher METTL3 expression may predict a better prognosis for RCC patients, possibly due to the inhibition of tumor growth by promoting cell cycle arrest in the G1 phase [193]. It has also been shown that the self-renewal of glioblastoma stem cells (GSC) is regulated by m6A mRNA modification, and METTL3 downregulation significantly promotes tumor progression [182]. Similar results were found in melanoma studies, where Jia et al. found that METTL3 downregulation led to reduced m6A levels in melanoma, predicting early recurrence and enhanced aggressiveness, and verified that METTL3-mediated m6A modification promoted the translation of the tumor suppressor gene HINT2 [194]. The opposing roles of METTL3 in different cancers may be related to tumor heterogeneity and METTL3 complex physiological functions, and METTL3 produces inconsistent effects in the same type of cancer. For example, METTL3 elevation promotes the progression of non-small cell lung cancer (NSCLC), but METTL3 also inhibits tumorigenesis in NSCLC [195, 196]. In HCC, METTL3 and METTL14 have opposing effects on the migration of hepatocellular carcinoma cells [24, 42]. These results suggest that some functions of METTL3 are independent of m6A modifications, and the potential mechanisms need further exploration.

Conclusions and prospects

In recent years, RNA m6A modification has emerged as a prominent field in cancer research. Dysregulation of is frequently observed across various types of cancer, exerting significant influence on cancer progression by modulating the expression of oncogenes and tumor suppressor genes. Aberrant m6A modification is closely associated with tumor progression and the prognosis of cancer patients, highlighting the potential for targeting m6A regulators as a promising approach for cancer therapy. However, despite the identification of numerous m6A modification modulators, only a limited number have demonstrated efficacy and actionable targets for cancer treatment. None of the reported inhibitors or activators targeting m6A modification have been approved for clinical use in treating cancer. Thorough investigation through pre trials is necessary before these targeted therapies can be approved for clinical application.
Currently, the m6A content in RNA can be detected by various methods, including two-dimensional thin-layer chromatography [197, 198], m6A dot-blot [19], and high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) [19, 20]. However, these methods are unsuitable for extensively characterizing modification sites [199]. Before the development of methylated RNA immunoprecipitation followed by high-throughput sequencing (MeRIP-seq), m6A distribution throughout the transcriptome was unknown, and this method attracted much attention for its accuracy and reproducibility [13, 200]. However, because MeRIP-seq relies on RNA fragments with a resolution of approximately 100–200 nt, it is not possible to detect methylation sites with single-nucleotide resolution [201]. Other methods such as photo-crosslinking-assisted m6A-sequencing (PA-m6A-Seq), site-specific cleavage, and radioactive-labelling followed by ligation-assisted extraction and thin-layer chromatography (SCARLET) have been used, but they are time-consuming and unsuitable for high-throughput applications [202, 203]. However, a new method called m6A individual nucleotide resolution crosslinking immunoprecipitation (miCLIP), which accurately detects m6A modification sites, is an important step in this field [204]. In addition, CRISPR-based genetically engineered groups can directly detect the effects of altering m6A modification sites in many organisms [205]. As a complementary method, it is valuable to study the function of m6A methylation. Although many methods for detecting m6A methylation have been developed, several challenges and difficulties remain.
As a malignant tumor with the highest incidence in men worldwide, late metastasis is fatal and incurable for patients, and the relationship between RNA modification and PCa may lead to novel strategies for treating PCa. Current research on METTL3 in PCa mainly involves its biological functions and mechanisms, but some functions have not been further investigated. Future research on METTL3 in PCa should mainly focus on tumor angiogenesis, glycolipid metabolism, maintenance of tumor stem cells, and its effect on the tumor microenvironment. In clinical applications, although studies have incorporated METTL3 into tumor biomarkers and inhibitor development with great potential, they remain in the early stages and require continued attention.

Acknowledgments

We thank Home for Researchers editorial team (www.​home-forresearchers.​com) for language editing service.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef
2.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.PubMedCrossRef Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.PubMedCrossRef
3.
Zurück zum Zitat Dy GW, Gore JL, Forouzanfar MH, Naghavi M, Fitzmaurice C. Global burden of urologic cancers, 1990-2013. Eur Urol. 2017;71(3):437–46.PubMedCrossRef Dy GW, Gore JL, Forouzanfar MH, Naghavi M, Fitzmaurice C. Global burden of urologic cancers, 1990-2013. Eur Urol. 2017;71(3):437–46.PubMedCrossRef
4.
Zurück zum Zitat Wong MC, Goggins WB, Wang HH, et al. Global incidence and mortality for prostate cancer: analysis of temporal patterns and trends in 36 countries. Eur Urol. 2016;70(5):862–74.PubMedCrossRef Wong MC, Goggins WB, Wang HH, et al. Global incidence and mortality for prostate cancer: analysis of temporal patterns and trends in 36 countries. Eur Urol. 2016;70(5):862–74.PubMedCrossRef
5.
Zurück zum Zitat Gillessen S, Armstrong A, Attard G, et al. Management of patients with advanced prostate cancer: report from the advanced prostate cancer consensus conference 2021. Eur Urol. 2022;82(1):115–41.PubMedCrossRef Gillessen S, Armstrong A, Attard G, et al. Management of patients with advanced prostate cancer: report from the advanced prostate cancer consensus conference 2021. Eur Urol. 2022;82(1):115–41.PubMedCrossRef
7.
9.
Zurück zum Zitat Desrosiers R, Friderici K, Rottman F. Identification of methylated nucleosides in messenger rna from novikoff hepatoma cells. Proc Natl Acad Sci U S A. 1974;71(10):3971–5.PubMedPubMedCentralCrossRef Desrosiers R, Friderici K, Rottman F. Identification of methylated nucleosides in messenger rna from novikoff hepatoma cells. Proc Natl Acad Sci U S A. 1974;71(10):3971–5.PubMedPubMedCentralCrossRef
10.
11.
Zurück zum Zitat Ding Z, Liu Y, Huang Q, Cheng C, Song L, Zhang C, et al. M6a-and immune-related lncrna signature confers robust predictive power for immune efficacy in lung squamous cell carcinoma. View. 2023;4(3):20220083.CrossRef Ding Z, Liu Y, Huang Q, Cheng C, Song L, Zhang C, et al. M6a-and immune-related lncrna signature confers robust predictive power for immune efficacy in lung squamous cell carcinoma. View. 2023;4(3):20220083.CrossRef
14.
Zurück zum Zitat Geula S, Moshitch-Moshkovitz S, Dominissini D, et al. Stem cells. M6a mrna methylation facilitates resolution of naïve pluripotency toward differentiation. Science. 2015;347(6225):1002–6.PubMedCrossRef Geula S, Moshitch-Moshkovitz S, Dominissini D, et al. Stem cells. M6a mrna methylation facilitates resolution of naïve pluripotency toward differentiation. Science. 2015;347(6225):1002–6.PubMedCrossRef
15.
Zurück zum Zitat Horiuchi K, Kawamura T, Iwanari H, et al. Identification of wilms' tumor 1-associating protein complex and its role in alternative splicing and the cell cycle. J Biol Chem. 2013;288(46):33292–302.PubMedPubMedCentralCrossRef Horiuchi K, Kawamura T, Iwanari H, et al. Identification of wilms' tumor 1-associating protein complex and its role in alternative splicing and the cell cycle. J Biol Chem. 2013;288(46):33292–302.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Schwartz S, Mumbach M, Jovanovic M, et al. Perturbation of m6a writers reveals two distinct classes of mrna methylation at internal and 5’ sites. Cell Rep. 2014;8(1):284–96.PubMedPubMedCentralCrossRef Schwartz S, Mumbach M, Jovanovic M, et al. Perturbation of m6a writers reveals two distinct classes of mrna methylation at internal and 5’ sites. Cell Rep. 2014;8(1):284–96.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Huang H, Weng H, Chen J. M(6)a modification in coding and non-coding rnas: roles and therapeutic implications in cancer. Cancer Cell. 2020;37(3):270–88.PubMedPubMedCentralCrossRef Huang H, Weng H, Chen J. M(6)a modification in coding and non-coding rnas: roles and therapeutic implications in cancer. Cancer Cell. 2020;37(3):270–88.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Xu C, Wang X, Liu K, et al. Structural basis for selective binding of m6a rna by the ythdc1 yth domain. Nat Chem Biol. 2014;10(11):927–9.PubMedCrossRef Xu C, Wang X, Liu K, et al. Structural basis for selective binding of m6a rna by the ythdc1 yth domain. Nat Chem Biol. 2014;10(11):927–9.PubMedCrossRef
22.
Zurück zum Zitat Li F, Zhao D, Wu J, Shi Y. Structure of the yth domain of human ythdf2 in complex with an m(6)a mononucleotide reveals an aromatic cage for m(6)a recognition. Cell Res. 2014;24(12):1490–2.PubMedPubMedCentralCrossRef Li F, Zhao D, Wu J, Shi Y. Structure of the yth domain of human ythdf2 in complex with an m(6)a mononucleotide reveals an aromatic cage for m(6)a recognition. Cell Res. 2014;24(12):1490–2.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Li T, Hu PS, Zuo Z, et al. Mettl3 facilitates tumor progression via an m(6)a-igf2bp2-dependent mechanism in colorectal carcinoma. Mol Cancer. 2019;18(1):112.PubMedPubMedCentralCrossRef Li T, Hu PS, Zuo Z, et al. Mettl3 facilitates tumor progression via an m(6)a-igf2bp2-dependent mechanism in colorectal carcinoma. Mol Cancer. 2019;18(1):112.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Li E, Wei B, Wang X, Kang R. Mettl3 enhances cell adhesion through stabilizing integrin β1 mrna via an m6a-hur-dependent mechanism in prostatic carcinoma. Am J Cancer Res. 2020;10:1012–25.PubMedPubMedCentral Li E, Wei B, Wang X, Kang R. Mettl3 enhances cell adhesion through stabilizing integrin β1 mrna via an m6a-hur-dependent mechanism in prostatic carcinoma. Am J Cancer Res. 2020;10:1012–25.PubMedPubMedCentral
32.
Zurück zum Zitat Boccaletto P, Stefaniak F, Ray A, et al. Modomics: a database of rna modification pathways. 2021 update. Nucleic Acids Res. 2022;50(d1):d231–5.PubMedCrossRef Boccaletto P, Stefaniak F, Ray A, et al. Modomics: a database of rna modification pathways. 2021 update. Nucleic Acids Res. 2022;50(d1):d231–5.PubMedCrossRef
33.
Zurück zum Zitat Barbieri I, Kouzarides T. Role of rna modifications in cancer. Nat Rev Cancer. 2020;20(6):303–22.PubMedCrossRef Barbieri I, Kouzarides T. Role of rna modifications in cancer. Nat Rev Cancer. 2020;20(6):303–22.PubMedCrossRef
34.
36.
Zurück zum Zitat Davalos V, Blanco S, Esteller M. Snapshot: messenger rna modifications. Cell. 2018;174(2):498–498.e1.PubMedCrossRef Davalos V, Blanco S, Esteller M. Snapshot: messenger rna modifications. Cell. 2018;174(2):498–498.e1.PubMedCrossRef
38.
39.
Zurück zum Zitat Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. Hnrnpa2b1 is a mediator of m(6)a-dependent nuclear rna processing events. Cell. 2015;162(6):1299–308.PubMedPubMedCentralCrossRef Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. Hnrnpa2b1 is a mediator of m(6)a-dependent nuclear rna processing events. Cell. 2015;162(6):1299–308.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Alarcón CR, Lee H, Goodarzi H, Halberg N, Tavazoie SF. N6-methyladenosine marks primary micrornas for processing. Nature. 2015;519(7544):482–5.PubMedPubMedCentralCrossRef Alarcón CR, Lee H, Goodarzi H, Halberg N, Tavazoie SF. N6-methyladenosine marks primary micrornas for processing. Nature. 2015;519(7544):482–5.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Liu N, Dai Q, Zheng G, He C, Parisien M, Pan T. N(6)-methyladenosine-dependent rna structural switches regulate rna-protein interactions. Nature. 2015;518(7540):560–4.PubMedPubMedCentralCrossRef Liu N, Dai Q, Zheng G, He C, Parisien M, Pan T. N(6)-methyladenosine-dependent rna structural switches regulate rna-protein interactions. Nature. 2015;518(7540):560–4.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat As W, Kretschmer J, Hackert P, et al. Human mettl16 is a n(6)-methyladenosine (m(6)a) methyltransferase that targets pre-mrnas and various non-coding rnas. EMBO Rep. 2017;18(11):2004–14.CrossRef As W, Kretschmer J, Hackert P, et al. Human mettl16 is a n(6)-methyladenosine (m(6)a) methyltransferase that targets pre-mrnas and various non-coding rnas. EMBO Rep. 2017;18(11):2004–14.CrossRef
45.
46.
Zurück zum Zitat Niu Y, Zhao X, Wu YS, Li MM, Wang XJ, Yang YG. N6-methyl-adenosine (m6a) in rna: an old modification with a novel epigenetic function. Genom Proteom Bioinform. 2013;11(1):8–17.CrossRef Niu Y, Zhao X, Wu YS, Li MM, Wang XJ, Yang YG. N6-methyl-adenosine (m6a) in rna: an old modification with a novel epigenetic function. Genom Proteom Bioinform. 2013;11(1):8–17.CrossRef
47.
48.
Zurück zum Zitat Chen Z, Zhong X, Xia M, Zhong J. The roles and mechanisms of the m6a reader protein ythdf1 in tumor biology and human diseases. Mol Ther Nucleic Acids. 2021;26:1270–9.PubMedPubMedCentralCrossRef Chen Z, Zhong X, Xia M, Zhong J. The roles and mechanisms of the m6a reader protein ythdf1 in tumor biology and human diseases. Mol Ther Nucleic Acids. 2021;26:1270–9.PubMedPubMedCentralCrossRef
49.
50.
Zurück zum Zitat Ping XL, Sun BF, Wang L, et al. Mammalian wtap is a regulatory subunit of the rna n6-methyladenosine methyltransferase. Cell Res. 2014;24(2):177–89.PubMedPubMedCentralCrossRef Ping XL, Sun BF, Wang L, et al. Mammalian wtap is a regulatory subunit of the rna n6-methyladenosine methyltransferase. Cell Res. 2014;24(2):177–89.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Patil DP, Chen CK, Pickering BF, et al. M(6)a rna methylation promotes xist-mediated transcriptional repression. Nature. 2016;537(7620):369–73.PubMedPubMedCentralCrossRef Patil DP, Chen CK, Pickering BF, et al. M(6)a rna methylation promotes xist-mediated transcriptional repression. Nature. 2016;537(7620):369–73.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Knuckles P, Lence T, Haussmann IU, et al. Zc3h13/flacc is required for adenosine methylation by bridging the mrna-binding factor rbm15/spenito to the m(6)a machinery component wtap/fl(2)d. Genes Dev. 2018;32(5–6):415–29.PubMedPubMedCentralCrossRef Knuckles P, Lence T, Haussmann IU, et al. Zc3h13/flacc is required for adenosine methylation by bridging the mrna-binding factor rbm15/spenito to the m(6)a machinery component wtap/fl(2)d. Genes Dev. 2018;32(5–6):415–29.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Wen J, Lv R, Ma H, et al. Zc3h13 regulates nuclear rna m(6)a methylation and mouse embryonic stem cell self-renewal. Mol Cell. 2018;69(6):1028–1038.e6.PubMedPubMedCentralCrossRef Wen J, Lv R, Ma H, et al. Zc3h13 regulates nuclear rna m(6)a methylation and mouse embryonic stem cell self-renewal. Mol Cell. 2018;69(6):1028–1038.e6.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Wei J, Liu F, Lu Z, et al. Differential m(6)a, m(6)a(m), and m(1)a demethylation mediated by fto in the cell nucleus and cytoplasm. Mol Cell. 2018;71(6):973–985.e5.PubMedPubMedCentralCrossRef Wei J, Liu F, Lu Z, et al. Differential m(6)a, m(6)a(m), and m(1)a demethylation mediated by fto in the cell nucleus and cytoplasm. Mol Cell. 2018;71(6):973–985.e5.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Liu J, Jia G. Methylation modifications in eukaryotic messenger rna. J Genet Genomics. 2014;41(1):21–33.PubMedCrossRef Liu J, Jia G. Methylation modifications in eukaryotic messenger rna. J Genet Genomics. 2014;41(1):21–33.PubMedCrossRef
56.
Zurück zum Zitat Zou S, Toh JD, Wong KH, Gao YG, Hong W, Woon EC. N(6)-methyladenosine: a conformational marker that regulates the substrate specificity of human demethylases fto and alkbh5. Sci Rep. 2016;6:25677.PubMedPubMedCentralCrossRef Zou S, Toh JD, Wong KH, Gao YG, Hong W, Woon EC. N(6)-methyladenosine: a conformational marker that regulates the substrate specificity of human demethylases fto and alkbh5. Sci Rep. 2016;6:25677.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Ueda Y, Ooshio I, Fusamae Y, et al. Alkb homolog 3-mediated trna demethylation promotes protein synthesis in cancer cells. Sci Rep. 2017;7:42271.PubMedPubMedCentralCrossRef Ueda Y, Ooshio I, Fusamae Y, et al. Alkb homolog 3-mediated trna demethylation promotes protein synthesis in cancer cells. Sci Rep. 2017;7:42271.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Cd A, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet. 2016;17(8):487–500.CrossRef Cd A, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet. 2016;17(8):487–500.CrossRef
59.
Zurück zum Zitat Huang H, Weng H, Sun W, et al. Recognition of rna n(6)-methyladenosine by igf2bp proteins enhances mrna stability and translation. Nat Cell Biol. 2018;20(3):285–95.PubMedPubMedCentralCrossRef Huang H, Weng H, Sun W, et al. Recognition of rna n(6)-methyladenosine by igf2bp proteins enhances mrna stability and translation. Nat Cell Biol. 2018;20(3):285–95.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Liu N, Zhou KI, Parisien M, Dai Q, Diatchenko L, Pan T. N6-methyladenosine alters rna structure to regulate binding of a low-complexity protein. Nucleic Acids Res. 2017;45(10):6051–63.PubMedPubMedCentralCrossRef Liu N, Zhou KI, Parisien M, Dai Q, Diatchenko L, Pan T. N6-methyladenosine alters rna structure to regulate binding of a low-complexity protein. Nucleic Acids Res. 2017;45(10):6051–63.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Petri BJ, Klinge CM. M6a readers, writers, erasers, and the m6a epitranscriptome in breast cancer. J Mol Endocrinol. 2023;70(2) Petri BJ, Klinge CM. M6a readers, writers, erasers, and the m6a epitranscriptome in breast cancer. J Mol Endocrinol. 2023;70(2)
62.
Zurück zum Zitat Sun T, Wu R, Ming L. The role of m6a rna methylation in cancer. Biomed Pharmacother. 2019;112:108613.PubMedCrossRef Sun T, Wu R, Ming L. The role of m6a rna methylation in cancer. Biomed Pharmacother. 2019;112:108613.PubMedCrossRef
65.
Zurück zum Zitat Li Y, Su R, Deng X, Chen Y, Chen J. Fto in cancer: functions, molecular mechanisms, and therapeutic implications. Trends Cancer. 2022;8(7):598–614.PubMedCrossRef Li Y, Su R, Deng X, Chen Y, Chen J. Fto in cancer: functions, molecular mechanisms, and therapeutic implications. Trends Cancer. 2022;8(7):598–614.PubMedCrossRef
66.
67.
Zurück zum Zitat Zaccara S, Ries Rj J Sr. Reading, writing and erasing mrna methylation. Nat Rev Mol Cell Biol. 2019;20(10):608–24.PubMedCrossRef Zaccara S, Ries Rj J Sr. Reading, writing and erasing mrna methylation. Nat Rev Mol Cell Biol. 2019;20(10):608–24.PubMedCrossRef
68.
Zurück zum Zitat Shi H, Wei J, He C. Where, when, and how: context-dependent functions of rna methylation writers, readers, and erasers. Mol Cell. 2019;74(4):640–50.PubMedPubMedCentralCrossRef Shi H, Wei J, He C. Where, when, and how: context-dependent functions of rna methylation writers, readers, and erasers. Mol Cell. 2019;74(4):640–50.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Wu R, Li A, Sun B, et al. A novel m(6)a reader prrc2a controls oligodendroglial specification and myelination. Cell Res. 2019;29(1):23–41.PubMedCrossRef Wu R, Li A, Sun B, et al. A novel m(6)a reader prrc2a controls oligodendroglial specification and myelination. Cell Res. 2019;29(1):23–41.PubMedCrossRef
70.
Zurück zum Zitat Zhang F, Kang Y, Wang M, et al. Fragile x mental retardation protein modulates the stability of its m6a-marked messenger rna targets. Hum Mol Genet. 2018;27(22):3936–50.PubMedPubMedCentral Zhang F, Kang Y, Wang M, et al. Fragile x mental retardation protein modulates the stability of its m6a-marked messenger rna targets. Hum Mol Genet. 2018;27(22):3936–50.PubMedPubMedCentral
71.
73.
Zurück zum Zitat Liao S, Sun H, Xu C. Yth domain: a family of n(6)-methyladenosine (m(6)a) readers. Genom Proteom Bioinform. 2018;16(2):99–107.CrossRef Liao S, Sun H, Xu C. Yth domain: a family of n(6)-methyladenosine (m(6)a) readers. Genom Proteom Bioinform. 2018;16(2):99–107.CrossRef
74.
Zurück zum Zitat Patil DP, Pickering BF, Jaffrey SR. Reading m(6)a in the transcriptome: m(6)a-binding proteins. Trends Cell Biol. 2018;28(2):113–27.PubMedCrossRef Patil DP, Pickering BF, Jaffrey SR. Reading m(6)a in the transcriptome: m(6)a-binding proteins. Trends Cell Biol. 2018;28(2):113–27.PubMedCrossRef
75.
Zurück zum Zitat Yang Y, Hsu PJ, Ys C, Yang YG. Dynamic transcriptomic m(6)a decoration: writers, erasers, readers and functions in rna metabolism. Cell Res. 2018;28(6):616–24.PubMedPubMedCentralCrossRef Yang Y, Hsu PJ, Ys C, Yang YG. Dynamic transcriptomic m(6)a decoration: writers, erasers, readers and functions in rna metabolism. Cell Res. 2018;28(6):616–24.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Peer E, Moshitch-Moshkovitz S, Rechavi G, Dominissini D. The epitranscriptome in translation regulation. Cold Spring Harb Perspect Biol. 2019;11(8) Peer E, Moshitch-Moshkovitz S, Rechavi G, Dominissini D. The epitranscriptome in translation regulation. Cold Spring Harb Perspect Biol. 2019;11(8)
77.
Zurück zum Zitat Livneh I, Moshitch-Moshkovitz S, Amariglio N, Rechavi G, Dominissini D. The m(6)a epitranscriptome: transcriptome plasticity in brain development and function. Nat Rev Neurosci. 2020;21(1):36–51.PubMedCrossRef Livneh I, Moshitch-Moshkovitz S, Amariglio N, Rechavi G, Dominissini D. The m(6)a epitranscriptome: transcriptome plasticity in brain development and function. Nat Rev Neurosci. 2020;21(1):36–51.PubMedCrossRef
78.
85.
Zurück zum Zitat Luxton HJ, Simpson BS, Mills IG, Nr B, Ahmed Z, Stavrinides V, et al. The oncogene metadherin interacts with the known splicing proteins ythdc1, sam68 and t-star and plays a novel role in alternative mrna splicing. Cancers (basel). 2019;11 https://doi.org/10.3390/cancers11091233. Luxton HJ, Simpson BS, Mills IG, Nr B, Ahmed Z, Stavrinides V, et al. The oncogene metadherin interacts with the known splicing proteins ythdc1, sam68 and t-star and plays a novel role in alternative mrna splicing. Cancers (basel). 2019;11 https://​doi.​org/​10.​3390/​cancers11091233.
86.
Zurück zum Zitat Song J, You G, Yin X, Zhu G, Wang W, Yu Y, et al. Overexpression of ythdc2 contributes to the progression of prostate cancer and predicts poor outcomes in patients with prostate cancer. J Biochem Mol Toxicol. 2023:e23308. https://doi.org/10.1002/jbt.23308. Song J, You G, Yin X, Zhu G, Wang W, Yu Y, et al. Overexpression of ythdc2 contributes to the progression of prostate cancer and predicts poor outcomes in patients with prostate cancer. J Biochem Mol Toxicol. 2023:e23308. https://​doi.​org/​10.​1002/​jbt.​23308.
96.
Zurück zum Zitat Zhou KI, Pan T. Structures of the m(6)a methyltransferase complex: two subunits with distinct but coordinated roles. Mol Cell. 2016;63(2):183–5.PubMedPubMedCentralCrossRef Zhou KI, Pan T. Structures of the m(6)a methyltransferase complex: two subunits with distinct but coordinated roles. Mol Cell. 2016;63(2):183–5.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Oerum S, Meynier V, Catala M, Tisné C. A comprehensive review of m6a/m6am rna methyltransferase structures. Nucleic Acids Res. 2021;49(13):7239–55.PubMedPubMedCentralCrossRef Oerum S, Meynier V, Catala M, Tisné C. A comprehensive review of m6a/m6am rna methyltransferase structures. Nucleic Acids Res. 2021;49(13):7239–55.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Śledź P, Jinek M. Structural insights into the molecular mechanism of the m(6)a writer complex. Elife. 2016;5 Śledź P, Jinek M. Structural insights into the molecular mechanism of the m(6)a writer complex. Elife. 2016;5
99.
Zurück zum Zitat Huang J, Dong X, Gong Z, et al. Solution structure of the rna recognition domain of mettl3-mettl14 n(6)-methyladenosine methyltransferase. Protein Cell. 2019;10(4):272–84.PubMedCrossRef Huang J, Dong X, Gong Z, et al. Solution structure of the rna recognition domain of mettl3-mettl14 n(6)-methyladenosine methyltransferase. Protein Cell. 2019;10(4):272–84.PubMedCrossRef
100.
Zurück zum Zitat Jones DT. Protein secondary structure prediction based on position-specific scoring matrices. J Mol Biol. 1999;292(2):195–202.PubMedCrossRef Jones DT. Protein secondary structure prediction based on position-specific scoring matrices. J Mol Biol. 1999;292(2):195–202.PubMedCrossRef
101.
Zurück zum Zitat Ward JJ, Mcguffin LJ, Bryson K, Buxton BF, Jones DT. The disopred server for the prediction of protein disorder. Bioinformatics. 2004;20(13):2138–9.PubMedCrossRef Ward JJ, Mcguffin LJ, Bryson K, Buxton BF, Jones DT. The disopred server for the prediction of protein disorder. Bioinformatics. 2004;20(13):2138–9.PubMedCrossRef
102.
Zurück zum Zitat Schöller E, Weichmann F, Treiber T, et al. Interactions, localization, and phosphorylation of the m(6)a generating mettl3-mettl14-wtap complex. RNA. 2018;24(4):499–512.PubMedPubMedCentralCrossRef Schöller E, Weichmann F, Treiber T, et al. Interactions, localization, and phosphorylation of the m(6)a generating mettl3-mettl14-wtap complex. RNA. 2018;24(4):499–512.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef
115.
Zurück zum Zitat Yan R, Dai W, Wu R, Huang H, Shu M. Therapeutic targeting m6a-guided mir-146a-5p signaling contributes to the melittin-induced selective suppression of bladder cancer. Cancer Lett. 2022;534:215615.PubMedCrossRef Yan R, Dai W, Wu R, Huang H, Shu M. Therapeutic targeting m6a-guided mir-146a-5p signaling contributes to the melittin-induced selective suppression of bladder cancer. Cancer Lett. 2022;534:215615.PubMedCrossRef
116.
Zurück zum Zitat Bi X, Lv X, Liu D, et al. Mettl3 promotes the initiation and metastasis of ovarian cancer by inhibiting ccng2 expression via promoting the maturation of pri-microrna-1246. Cell Death Discov. 2021;7(1):237.PubMedPubMedCentralCrossRef Bi X, Lv X, Liu D, et al. Mettl3 promotes the initiation and metastasis of ovarian cancer by inhibiting ccng2 expression via promoting the maturation of pri-microrna-1246. Cell Death Discov. 2021;7(1):237.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Jiang X, Yuan Y, Tang L, et al. Comprehensive pan-cancer analysis of the prognostic and immunological roles of the mettl3/lncrna-snhg1/mirna-140-3p/ube2c axis. Front Cell Dev Biol. 2021;9:765772.PubMedPubMedCentralCrossRef Jiang X, Yuan Y, Tang L, et al. Comprehensive pan-cancer analysis of the prognostic and immunological roles of the mettl3/lncrna-snhg1/mirna-140-3p/ube2c axis. Front Cell Dev Biol. 2021;9:765772.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Ji F, Lu Y, Chen S, et al. M(6)a methyltransferase mettl3-mediated lncrna foxd2-as1 promotes the tumorigenesis of cervical cancer. Mol Ther Oncolytics. 2021;22:574–81.PubMedPubMedCentralCrossRef Ji F, Lu Y, Chen S, et al. M(6)a methyltransferase mettl3-mediated lncrna foxd2-as1 promotes the tumorigenesis of cervical cancer. Mol Ther Oncolytics. 2021;22:574–81.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Xue L, Li J, Lin Y, et al. M(6) a transferase mettl3-induced lncrna abhd11-as1 promotes the Warburg effect of non-small-cell lung cancer. J Cell Physiol. 2021;236(4):2649–58.PubMedCrossRef Xue L, Li J, Lin Y, et al. M(6) a transferase mettl3-induced lncrna abhd11-as1 promotes the Warburg effect of non-small-cell lung cancer. J Cell Physiol. 2021;236(4):2649–58.PubMedCrossRef
120.
Zurück zum Zitat Wang D, Wang X, Huang B, et al. Mettl3 promotes prostate cancer progression by regulating mir-182 maturation in m6a-dependent manner. Andrologia. 2022;54(7):1581–91.PubMedCrossRef Wang D, Wang X, Huang B, et al. Mettl3 promotes prostate cancer progression by regulating mir-182 maturation in m6a-dependent manner. Andrologia. 2022;54(7):1581–91.PubMedCrossRef
121.
Zurück zum Zitat Ma H, Zhang F, Zhong Q, Hou J. Mettl3-mediated m6a modification of kif3c-mrna promotes prostate cancer progression and is negatively regulated by mir-320d. Aging. 2021;13(18):22332–44.PubMedPubMedCentralCrossRef Ma H, Zhang F, Zhong Q, Hou J. Mettl3-mediated m6a modification of kif3c-mrna promotes prostate cancer progression and is negatively regulated by mir-320d. Aging. 2021;13(18):22332–44.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Mao Y, Li W, Weng Y, et al. Mettl3-mediated m(6)a modification of lncrna malat1 facilitates prostate cancer growth by activation of pi3k/akt signaling. Cell Transplant. 2022;31:9636897221122997.PubMedCrossRef Mao Y, Li W, Weng Y, et al. Mettl3-mediated m(6)a modification of lncrna malat1 facilitates prostate cancer growth by activation of pi3k/akt signaling. Cell Transplant. 2022;31:9636897221122997.PubMedCrossRef
123.
Zurück zum Zitat Chen B, Liu C, Long H, Bai G, Zhu Y, Xu H. N(6)-methyladenosine-induced long non-coding rna pvt1 regulates the mir-27b-3p/blm axis to promote prostate cancer progression. Int J Oncol. 2023;62(1) Chen B, Liu C, Long H, Bai G, Zhu Y, Xu H. N(6)-methyladenosine-induced long non-coding rna pvt1 regulates the mir-27b-3p/blm axis to promote prostate cancer progression. Int J Oncol. 2023;62(1)
125.
Zurück zum Zitat Qu S, Huang C, Zhu T, Wang K, Zhang H, Wang L, et al. Olfml3, as a potential predictor of prognosis and therapeutic target for glioma, is closely related to immune cell infiltration. View. 2023;4:20220052.CrossRef Qu S, Huang C, Zhu T, Wang K, Zhang H, Wang L, et al. Olfml3, as a potential predictor of prognosis and therapeutic target for glioma, is closely related to immune cell infiltration. View. 2023;4:20220052.CrossRef
126.
127.
Zurück zum Zitat Musselman CA, Me L, Côté J, Kutateladze TG. Perceiving the epigenetic landscape through histone readers. Nat Struct Mol Biol. 2012;19(12):1218–27.PubMedPubMedCentralCrossRef Musselman CA, Me L, Côté J, Kutateladze TG. Perceiving the epigenetic landscape through histone readers. Nat Struct Mol Biol. 2012;19(12):1218–27.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Wojtas MN, Pandey RR, Mendel M, Homolka D, Sachidanandam R, Pillai RS. Regulation of m6a transcripts by the 3’→5’ rna helicase ythdc2 is essential for a successful meiotic program in the mammalian germline. Mol Cell. 2017;68(2):374–387.e12.PubMedCrossRef Wojtas MN, Pandey RR, Mendel M, Homolka D, Sachidanandam R, Pillai RS. Regulation of m6a transcripts by the 3’→5’ rna helicase ythdc2 is essential for a successful meiotic program in the mammalian germline. Mol Cell. 2017;68(2):374–387.e12.PubMedCrossRef
129.
Zurück zum Zitat Du H, Zhao Y, He J, et al. Ythdf2 destabilizes m(6)a-containing rna through direct recruitment of the ccr4-not deadenylase complex. Nat Commun. 2016;7:12626.PubMedPubMedCentralCrossRef Du H, Zhao Y, He J, et al. Ythdf2 destabilizes m(6)a-containing rna through direct recruitment of the ccr4-not deadenylase complex. Nat Commun. 2016;7:12626.PubMedPubMedCentralCrossRef
130.
Zurück zum Zitat Wang X, Lu Z, Gomez A, et al. N6-methyladenosine-dependent regulation of messenger rna stability. Nature. 2014;505(7481):117–20.PubMedCrossRef Wang X, Lu Z, Gomez A, et al. N6-methyladenosine-dependent regulation of messenger rna stability. Nature. 2014;505(7481):117–20.PubMedCrossRef
131.
132.
Zurück zum Zitat Zhou J, Wan J, Gao X, Zhang X, Jaffrey SR, Qian SB. Dynamic m(6)a mrna methylation directs translational control of heat shock response. Nature. 2015;526(7574):591–4.PubMedPubMedCentralCrossRef Zhou J, Wan J, Gao X, Zhang X, Jaffrey SR, Qian SB. Dynamic m(6)a mrna methylation directs translational control of heat shock response. Nature. 2015;526(7574):591–4.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Ma XX, Cao ZG, Zhao SL. M6a methyltransferase mettl3 promotes the progression of prostate cancer via m6a-modified lef1. Eur Rev Med Pharmacol Sci. 2020;24(7):3565–71.PubMed Ma XX, Cao ZG, Zhao SL. M6a methyltransferase mettl3 promotes the progression of prostate cancer via m6a-modified lef1. Eur Rev Med Pharmacol Sci. 2020;24(7):3565–71.PubMed
138.
Zurück zum Zitat Babaei G, Aziz SG, Jaghi N. Emt, cancer stem cells and autophagy; the three main axes of metastasis. Biomed Pharmacother. 2021;133:110909.PubMedCrossRef Babaei G, Aziz SG, Jaghi N. Emt, cancer stem cells and autophagy; the three main axes of metastasis. Biomed Pharmacother. 2021;133:110909.PubMedCrossRef
139.
140.
141.
Zurück zum Zitat Chen Y, Pan C, Wang X, et al. Silencing of mettl3 effectively hinders invasion and metastasis of prostate cancer cells. Theranostics. 2021;11(16):7640–57.PubMedPubMedCentralCrossRef Chen Y, Pan C, Wang X, et al. Silencing of mettl3 effectively hinders invasion and metastasis of prostate cancer cells. Theranostics. 2021;11(16):7640–57.PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Liang L, Li Q, Huang LY, et al. Loss of arhgdia expression is associated with poor prognosis in hcc and promotes invasion and metastasis of hcc cells. Int J Oncol. 2014;45(2):659–66.PubMedCrossRef Liang L, Li Q, Huang LY, et al. Loss of arhgdia expression is associated with poor prognosis in hcc and promotes invasion and metastasis of hcc cells. Int J Oncol. 2014;45(2):659–66.PubMedCrossRef
144.
Zurück zum Zitat Klaassen Z, Howard LE, De Hoedt A, et al. Factors predicting skeletal-related events in patients with bone metastatic castration-resistant prostate cancer. Cancer. 2017;123(9):1528–35.PubMedCrossRef Klaassen Z, Howard LE, De Hoedt A, et al. Factors predicting skeletal-related events in patients with bone metastatic castration-resistant prostate cancer. Cancer. 2017;123(9):1528–35.PubMedCrossRef
146.
Zurück zum Zitat Carvalho TM, Cardoso HJ, Figueira MI, Vaz CV, Socorro S. The peculiarities of cancer cell metabolism: a route to metastasization and a target for therapy. Eur J Med Chem. 2019;171:343–63.PubMedCrossRef Carvalho TM, Cardoso HJ, Figueira MI, Vaz CV, Socorro S. The peculiarities of cancer cell metabolism: a route to metastasization and a target for therapy. Eur J Med Chem. 2019;171:343–63.PubMedCrossRef
147.
Zurück zum Zitat Sadeghi RN, Karami-Tehrani F, Salami S. Targeting prostate cancer cell metabolism: impact of hexokinase and cpt-1 enzymes. Tumour Biol. 2015;36(4):2893–905.PubMedCrossRef Sadeghi RN, Karami-Tehrani F, Salami S. Targeting prostate cancer cell metabolism: impact of hexokinase and cpt-1 enzymes. Tumour Biol. 2015;36(4):2893–905.PubMedCrossRef
149.
Zurück zum Zitat Liu J, Yuan JF, Wang YZ. Mettl3-stabilized lncrna snhg7 accelerates glycolysis in prostate cancer via srsf1/c-myc axis. Exp Cell Res. 2022;416(1):113149.PubMedCrossRef Liu J, Yuan JF, Wang YZ. Mettl3-stabilized lncrna snhg7 accelerates glycolysis in prostate cancer via srsf1/c-myc axis. Exp Cell Res. 2022;416(1):113149.PubMedCrossRef
151.
Zurück zum Zitat Lin Y, Wei X, Jian Z, Zhang X. Mettl3 expression is associated with glycolysis metabolism and sensitivity to glycolytic stress in hepatocellular carcinoma. Cancer Med. 2020;9(8):2859–67.PubMedPubMedCentralCrossRef Lin Y, Wei X, Jian Z, Zhang X. Mettl3 expression is associated with glycolysis metabolism and sensitivity to glycolytic stress in hepatocellular carcinoma. Cancer Med. 2020;9(8):2859–67.PubMedPubMedCentralCrossRef
152.
Zurück zum Zitat Wang Y, Chen J, Wu Z, et al. Mechanisms of enzalutamide resistance in castration-resistant prostate cancer and therapeutic strategies to overcome it. Br J Pharmacol. 2021;178(2):239–61.PubMedCrossRef Wang Y, Chen J, Wu Z, et al. Mechanisms of enzalutamide resistance in castration-resistant prostate cancer and therapeutic strategies to overcome it. Br J Pharmacol. 2021;178(2):239–61.PubMedCrossRef
153.
Zurück zum Zitat Antonarakis ES, Lu C, Wang H, et al. Ar-v7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371(11):1028–38.PubMedPubMedCentralCrossRef Antonarakis ES, Lu C, Wang H, et al. Ar-v7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371(11):1028–38.PubMedPubMedCentralCrossRef
154.
Zurück zum Zitat Cotter KA, Gallon J, Uebersax N, et al. Mapping of m(6)a and its regulatory targets in prostate cancer reveals a mettl3-low induction of therapy resistance. Mol Cancer Res. 2021;19(8):1398–411.PubMedPubMedCentralCrossRef Cotter KA, Gallon J, Uebersax N, et al. Mapping of m(6)a and its regulatory targets in prostate cancer reveals a mettl3-low induction of therapy resistance. Mol Cancer Res. 2021;19(8):1398–411.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Boriack-Sjodin PA, Ribich S, Copeland RA. Rna-modifying proteins as anticancer drug targets. Nat Rev Drug Discov. 2018;17(6):435–53.PubMedCrossRef Boriack-Sjodin PA, Ribich S, Copeland RA. Rna-modifying proteins as anticancer drug targets. Nat Rev Drug Discov. 2018;17(6):435–53.PubMedCrossRef
156.
Zurück zum Zitat Bedi RK, Huang D, Eberle SA, Wiedmer L, Śledź P, Caflisch A. Small-molecule inhibitors of mettl3, the major human epitranscriptomic writer. Chemmedchem. 2020;15(9):744–8.PubMedCrossRef Bedi RK, Huang D, Eberle SA, Wiedmer L, Śledź P, Caflisch A. Small-molecule inhibitors of mettl3, the major human epitranscriptomic writer. Chemmedchem. 2020;15(9):744–8.PubMedCrossRef
157.
Zurück zum Zitat Moroz-Omori EV, Huang D, Kumar Bedi R, et al. Mettl3 inhibitors for epitranscriptomic modulation of cellular processes. Chemmedchem. 2021;16(19):3035–43.PubMedPubMedCentralCrossRef Moroz-Omori EV, Huang D, Kumar Bedi R, et al. Mettl3 inhibitors for epitranscriptomic modulation of cellular processes. Chemmedchem. 2021;16(19):3035–43.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Dolbois A, Bedi RK, Bochenkova E, et al. 1,4,9-triazaspiro[5.5]undecan-2-one derivatives as potent and selective mettl3 inhibitors. J Med Chem. 2021;64(17):12738–60.PubMedCrossRef Dolbois A, Bedi RK, Bochenkova E, et al. 1,4,9-triazaspiro[5.5]undecan-2-one derivatives as potent and selective mettl3 inhibitors. J Med Chem. 2021;64(17):12738–60.PubMedCrossRef
159.
Zurück zum Zitat Yankova E, Blackaby W, Albertella M, et al. Small-molecule inhibition of mettl3 as a strategy against myeloid leukaemia. Nature. 2021;593(7860):597–601.PubMedPubMedCentralCrossRef Yankova E, Blackaby W, Albertella M, et al. Small-molecule inhibition of mettl3 as a strategy against myeloid leukaemia. Nature. 2021;593(7860):597–601.PubMedPubMedCentralCrossRef
160.
Zurück zum Zitat Ofir-Rosenfeld Y, Vasiliauskaitė L, Saunders C, et al. Stc-15, an oral small molecule inhibitor of the rna methyltransferase mettl3, inhibits tumour growth through activation of anti-cancer immune responses associated with increased interferon signalling, and synergises with t cell checkpoint blockade. Eur J Cancer. 2022;174:s123.CrossRef Ofir-Rosenfeld Y, Vasiliauskaitė L, Saunders C, et al. Stc-15, an oral small molecule inhibitor of the rna methyltransferase mettl3, inhibits tumour growth through activation of anti-cancer immune responses associated with increased interferon signalling, and synergises with t cell checkpoint blockade. Eur J Cancer. 2022;174:s123.CrossRef
174.
Zurück zum Zitat Ni Z, Sun P, Zheng J, et al. Jnk signaling promotes bladder cancer immune escape by regulating mettl3-mediated m6a modification of pd-l1 mrna. Cancer Res. 2022;82(9):1789–802.PubMedCrossRef Ni Z, Sun P, Zheng J, et al. Jnk signaling promotes bladder cancer immune escape by regulating mettl3-mediated m6a modification of pd-l1 mrna. Cancer Res. 2022;82(9):1789–802.PubMedCrossRef
175.
Zurück zum Zitat Song C, Zhou C. Hoxa10 mediates epithelial-mesenchymal transition to promote gastric cancer metastasis partly via modulation of tgfb2/smad/mettl3 signaling axis. J Exp Clin Cancer Res. 2021;40(1):62.PubMedPubMedCentralCrossRef Song C, Zhou C. Hoxa10 mediates epithelial-mesenchymal transition to promote gastric cancer metastasis partly via modulation of tgfb2/smad/mettl3 signaling axis. J Exp Clin Cancer Res. 2021;40(1):62.PubMedPubMedCentralCrossRef
176.
Zurück zum Zitat Meng W, Xiao H, Mei P, et al. Critical roles of mettl3 in translation regulation of cancer. Biomolecules. 2023;13(2) Meng W, Xiao H, Mei P, et al. Critical roles of mettl3 in translation regulation of cancer. Biomolecules. 2023;13(2)
178.
Zurück zum Zitat Chamorro-Jorganes A, Sweaad WK, Katare R, et al. Mettl3 regulates angiogenesis by modulating let-7e-5p and mirna-18a-5p expression in endothelial cells. Arterioscler Thromb Vasc Biol. 2021;41(6):e325–37.PubMedCrossRef Chamorro-Jorganes A, Sweaad WK, Katare R, et al. Mettl3 regulates angiogenesis by modulating let-7e-5p and mirna-18a-5p expression in endothelial cells. Arterioscler Thromb Vasc Biol. 2021;41(6):e325–37.PubMedCrossRef
179.
Zurück zum Zitat Zhang G, Wang T, Huang Z, et al. Mettl3 dual regulation of the stability of linc00662 and vegfa rnas promotes colorectal cancer angiogenesis. Discov Oncol. 2022;13(1):89.PubMedPubMedCentralCrossRef Zhang G, Wang T, Huang Z, et al. Mettl3 dual regulation of the stability of linc00662 and vegfa rnas promotes colorectal cancer angiogenesis. Discov Oncol. 2022;13(1):89.PubMedPubMedCentralCrossRef
180.
Zurück zum Zitat Wang G, Dai Y, Li K, et al. Deficiency of mettl3 in bladder cancer stem cells inhibits bladder cancer progression and angiogenesis. Front Cell Dev Biol. 2021;9:627706.PubMedPubMedCentralCrossRef Wang G, Dai Y, Li K, et al. Deficiency of mettl3 in bladder cancer stem cells inhibits bladder cancer progression and angiogenesis. Front Cell Dev Biol. 2021;9:627706.PubMedPubMedCentralCrossRef
181.
183.
Zurück zum Zitat Gao Q, Zheng J, Ni Z, et al. The m(6)a methylation-regulated aff4 promotes self-renewal of bladder cancer stem cells. Stem Cells Int. 2020;2020:8849218.PubMedPubMedCentralCrossRef Gao Q, Zheng J, Ni Z, et al. The m(6)a methylation-regulated aff4 promotes self-renewal of bladder cancer stem cells. Stem Cells Int. 2020;2020:8849218.PubMedPubMedCentralCrossRef
184.
Zurück zum Zitat Zhang C, Samanta D, Lu H, et al. Hypoxia induces the breast cancer stem cell phenotype by hif-dependent and alkbh5-mediated m6a-demethylation of nanog mrna. Proc Natl Acad Sci U S A. 2016;113(14):e2047–56.PubMedPubMedCentralCrossRef Zhang C, Samanta D, Lu H, et al. Hypoxia induces the breast cancer stem cell phenotype by hif-dependent and alkbh5-mediated m6a-demethylation of nanog mrna. Proc Natl Acad Sci U S A. 2016;113(14):e2047–56.PubMedPubMedCentralCrossRef
185.
Zurück zum Zitat Ji G, Huang C, He S, et al. Comprehensive analysis of m6a regulators prognostic value in prostate cancer. Aging (albany ny). 2020;12(14):14863–84.PubMedPubMedCentralCrossRef Ji G, Huang C, He S, et al. Comprehensive analysis of m6a regulators prognostic value in prostate cancer. Aging (albany ny). 2020;12(14):14863–84.PubMedPubMedCentralCrossRef
186.
Zurück zum Zitat Lothion-Roy J, Haigh DB, Harris AE, et al. Clinical and molecular significance of the rna m(6)a methyltransferase complex in prostate cancer. Front Genet. 2022;13:1096071.PubMedCrossRef Lothion-Roy J, Haigh DB, Harris AE, et al. Clinical and molecular significance of the rna m(6)a methyltransferase complex in prostate cancer. Front Genet. 2022;13:1096071.PubMedCrossRef
187.
Zurück zum Zitat Cai C, He HH, Chen S, et al. Androgen receptor gene expression in prostate cancer is directly suppressed by the androgen receptor through recruitment of lysine-specific demethylase 1. Cancer Cell. 2011;20(4):457–71.PubMedPubMedCentralCrossRef Cai C, He HH, Chen S, et al. Androgen receptor gene expression in prostate cancer is directly suppressed by the androgen receptor through recruitment of lysine-specific demethylase 1. Cancer Cell. 2011;20(4):457–71.PubMedPubMedCentralCrossRef
188.
Zurück zum Zitat Kashyap V, Ahmad S, Nilsson EM, et al. The lysine specific demethylase-1 (lsd1/kdm1a) regulates vegf-a expression in prostate cancer. Mol Oncol. 2013;7(3):555–66.PubMedPubMedCentralCrossRef Kashyap V, Ahmad S, Nilsson EM, et al. The lysine specific demethylase-1 (lsd1/kdm1a) regulates vegf-a expression in prostate cancer. Mol Oncol. 2013;7(3):555–66.PubMedPubMedCentralCrossRef
189.
Zurück zum Zitat Cai C, He HH, Gao S, et al. Lysine-specific demethylase 1 has dual functions as a major regulator of androgen receptor transcriptional activity. Cell Rep. 2014;9(5):1618–27.PubMedPubMedCentralCrossRef Cai C, He HH, Gao S, et al. Lysine-specific demethylase 1 has dual functions as a major regulator of androgen receptor transcriptional activity. Cell Rep. 2014;9(5):1618–27.PubMedPubMedCentralCrossRef
190.
Zurück zum Zitat Lence T, Akhtar J, Bayer M, et al. M(6)a modulates neuronal functions and sex determination in drosophila. Nature. 2016;540(7632):242–7.PubMedCrossRef Lence T, Akhtar J, Bayer M, et al. M(6)a modulates neuronal functions and sex determination in drosophila. Nature. 2016;540(7632):242–7.PubMedCrossRef
191.
Zurück zum Zitat Haussmann IU, Bodi Z, Sanchez-Moran E, et al. M(6)a potentiates sxl alternative pre-mrna splicing for robust drosophila sex determination. Nature. 2016;540(7632):301–4.PubMedCrossRef Haussmann IU, Bodi Z, Sanchez-Moran E, et al. M(6)a potentiates sxl alternative pre-mrna splicing for robust drosophila sex determination. Nature. 2016;540(7632):301–4.PubMedCrossRef
Metadaten
Titel
The role of the methyltransferase METTL3 in prostate cancer: a potential therapeutic target
verfasst von
Xuming Zhou
Keqiang Chai
Hezhen Zhu
Cong Luo
Xiaofeng Zou
Junrong Zou
Guoxi Zhang
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2024
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-11741-1

Weitere Artikel der Ausgabe 1/2024

BMC Cancer 1/2024 Zur Ausgabe

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Alter verschlechtert Prognose bei Endometriumkarzinom

11.05.2024 Endometriumkarzinom Nachrichten

Ein höheres Alter bei der Diagnose eines Endometriumkarzinoms ist mit aggressiveren Tumorcharakteristika assoziiert, scheint aber auch unabhängig von bekannten Risikofaktoren die Prognose der Erkrankung zu verschlimmern.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.