Functions and mechanisms of RNA m6A regulators in breast cancer (Review)
- Authors:
- Published online on: July 26, 2024 https://doi.org/10.3892/ijo.2024.5674
- Article Number: 86
-
Copyright : © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].
Abstract
Introduction
Breast cancer (BC) is a common malignant tumor and its incidence rate has shown an overall upward trend in the past decade (1). Despite the progress in both understanding and treating BC, nearly 30% of patients suffer from recurrence or metastasis due to the deficiency of effective treatment or prevention strategies, which is the main reason for BC-related mortality (2). The extensively used classification for BC comprises Luminal A, Luminal B, human epidermal growth factor receptor (EGFR) 2 (HER2) overexpression and triple-negative BC (TNBC) (3). Studies have indicated that with early diagnosis and timely treatment, the overall survival of nonmetastatic BC and de novo metastatic BC (MBC) has been evidently improved. However, for recurrent MBC and elderly patients, there has been no improvement in decades (4). Hence, investigating the molecular mechanisms underlying the onset and progression of BC, and enhancing the capacity for monitoring BC treatment efficacy or identifying promising therapeutic targets, are of immense importance for precise diagnosis, efficient stratified management and the development of more refined treatment strategies for BC.
N6-methyladenosine (m6A) is the most prevalent internal mRNA modification in eukaryotes, which is installed by the methyltransferase complex (MTC) and removed by demethylases (5). It has been established as a widespread regulatory mechanism that controls gene expression in diverse physiological and pathological processes, including cancer (6,7). As the most universal epigenetic RNA modification, m6A plays a crucial role in regulating RNA stability, decay, splicing, transport and translation, thus affecting tumor progression significantly (8,9).
Abnormal m6A regulators have been recognized as new anticancer drug targets according to the close correlation between aberrant m6A modification and the occurrence, progression and prognosis of tumors (10). As the m6A modification and its associated factors are significantly dysregulated in cancers, gaining a comprehensive understanding of their roles in tumorigenesis and cancer progression will provide in-depth insight into the development of new therapeutic strategies for cancer treatment. The present review aims to summarize the current understanding of m6A modification and its functions in biological processes and cancers, with a particular focus on its mechanisms and roles in BC progression. Furthermore, the functions of m6A in DNA damage response, genomic instability and metabolic reprogramming were summarized.
BC
BC originates from mammary duct epithelial cells, which is the malignant tumor type with the highest incidence and mortality rates for women worldwide, accounting for ~30% of cancers in females (11-13). Clinical manifestations of BC may include breast lumps, nipple discharge and breast skin changes. However, early symptoms of part of breast cancer are not obvious or characteristic, which increases the difficulty of early identification.
BC exhibits apparent heterogeneity. According to the status of hormone receptors [estrogen receptor (ER) and progesterone receptor (PR)] and HER2, BC can be classified into three primary subtypes: Luminal ER-positive and PR-positive, which can be further categorized as luminal A and B, HER2-positive BC and TNBC (14,15). This BC classification based on biomarkers provides a foundation for further research and more precise determination of prognosis and selection of personalized treatments (13). For instance, the systemic treatment of nonmetastatic BC typically varies based on the subtype: Hormone receptor-positive tumors generally exhibit improved outcomes with endocrine therapy, while erb-b2 receptor tyrosine kinase 2 (ERBB2)-positive tumors typically require ERBB2-targeted antibodies or small-molecule inhibitors in combination with chemotherapy. By contrast, patients with triple-negative tumors tend to display greater sensitivity to chemotherapy (16).
The prognosis for BC varies among the different subtypes, which may be the most significant factor. Luminal A is the molecular subtype with the highest proportion in BC, exhibiting the lowest malignancy and the highest 5-year survival rate. The prognosis of patients with luminal B is slightly worse than that of patients with luminal A according to statistics (17). Compared to other subtypes, patients who are HER2-positive and those with TNBC often exhibit greater invasiveness, higher potential for recurrence and metastasis, and a poorer prognosis. It poses a significant challenge in the treatment of BC (18-20). In addition, other factors such as advanced age at diagnosis, later stage of cancer progression, metastasis, genetic predisposition and even high parity may also contribute to a worse prognosis (17).
A large portion of BC cases can be attributed to reproductive and hormonal factors (early menarche, late menopause, later primiparity age) (21), as well as lifestyle factors (e.g. overweight, lack of exercise, alcohol intake and smoking) (22). It has been proved that long-term contraceptives or menopausal hormone therapy with a combination of estrogen and progesterone raise the risk of BC as well (23). Familial inheritance is another universally acknowledged formidable hazard factor for BC. Women with a family history have a 2-to-4-fold increased probability of suffering from BC compared to others, with younger diagnosis ages and even higher mortality rates (24,25). Germline mutations in genes such as ATM serine/threonine kinase, BRCA1 DNA repair associated (BRCA1), BRCA2, checkpoint kinase 2 and partner and localizer of BRCA2 are frequently associated with an increased risk of developing BC (26,27). However, such mutations are rare in the general population.
M6A
M6A refers to methylation modification on the 6th nitrogen atom of RNA adenylate (28), which is the most universal internal messenger RNA modification in eukaryotes. M6A methylation can be found in mRNA, micro (mi)RNAs and long non-coding (lnc)RNAs to monitor and regulate their stability, translation, splicing and transport (29,30). The majority of m6A-modified mRNAs contain only one m6A site, while others contain 20 or more. Sequence analysis indicated that these sites emerge uniformly on the consensus RRACH motif and are not randomly distributed throughout the entire transcript. Instead, they tend to be concentrated in the 3'-untranslated region (UTR) near the stop codon (31,32). Furthermore, m6A modification is dynamic and reversible, which is installed by methyltransferases ('writers'), recognized by RNA-binding proteins ('readers') and removed by demethylases ('erasers') (28). Methyltransferase complex, an enzyme that catalyzes the methylation of m6A, consists of several core proteins. The so-called 'reader' can identify and bind to m6A methylated targets to carry out follow-up biological processes (33). The erasers are in charge of removing relevant markers through dynamic regulation, but they exert activity only in specific tissues or under certain disease-relevant conditions. These proteases constitute a vital regulator system required for different stages of gene expression that affects specific biological processes (34). However, when these regulators are dysregulated, particularly in tumors, they may stimulate the occurrence of tumors, proliferation and metastasis of cancer cells (9,35). Hence, investigating disorders in m6A levels could be immensely significant for detecting cancer and monitoring its treatment.
m6A writers
m6A methyltransferase complex, which is named 'writers' as well, is composed of core proteins methyltransferase-like 3 (METTL3), METTL14, Wilms tumor 1-associated protein (WTAP) and other proteins. METTL3 and METTL14 form stable heterodimers, which are responsible for the majority of m6A sites in mRNA (34). Among them, METTL3 functions as the fundamental catalytic enzyme by binding to S-adenosylmethionine as a subunit, and is capable of enhancing the translation of most oncogenic mRNAs (31,36). METTL14 acts as an allosteric activator of METTL3, thereby stabilizing its structure and recognizing target RNAs (37). WTAP is essential for the proper localization of the METTL3-METTL14 complex to nuclear speckles and for the enhancement of its catalytic activity (38). Furthermore, m6A methylation is regulated by several other proteins. METTL16 has been demonstrated to function independently and regulate m6A modification in several RNAs. Specifically, in the nucleus, it deposits N6 into specific mRNA targets, while separately stimulating translation in the cytoplasm (39). KIAA1429 [also known as vir-like m6A methyltransferase-associated protein (VIRMA)], a newly confirmed ingredient of the m6A methyltransferase complex, is crucial in guiding the process of regioselective m6A deposition. It means that KIAA1429 can mediate preferential m6A deposition in the 3'UTR and around the termination codon (40). RNA binding motif protein 15/15B (RBM15/15B) can attract and bind cone proteins complexes, directing them to specific RNA positions (41). Zinc finger CCCH domain-containing protein 13 (ZC3H13) serves as an anchor for the complex (WTAP, VIRMA and Cbl proto-oncogene like 1) within the nucleus so as to promote m6A methylation and mouse embryonic stem cell self-renewal (42). The summarized functions of m6A writers are presented in Table I.
m6A erasers
The eraser enzyme is capable of facilitating m6A demethylation through the involvement of two proteins, namely Fat mass and obesity-associated protein (FTO) and AlkB homolog 3/5 RNA demethylase (ALKBH3/5) (33). FTO and ALKBH5, which belong to the α-ketoglutarate-dependent dioxygenase family, only work in the presence of oxygen, ferrous ions and α-ketoglutarate (43). They initiate the conversion of m6A into N6 hydroxymethyl adenosine and subsequently into N6 formyl adenosine (f6A). Ultimately, f6A is hydrolyzed to adenosine to complete the demethylation process (43). FTO is the first m6A demethylase and it exhibits a strong correlation with weight gain, obesity and other metabolic diseases in humans (44). FTO is situated in both the nucleus and cytoplasm, and directly participates in the regulation of multiple pre-nuclear mRNA processing pathways, as well as other processes such as mRNA splicing (45). The second eraser, ALKBH5, is located in the nucleus and regulates gene expression mainly by mediating the transportation, metabolism and assembly of mRNA (43). The expression level of FTO and ALKBH5 affects the level of m6A in mRNA. Numerous studies have reported that the overexpression of m6A erasers is closely related to the occurrence and development of cancer (43,44,46). The functions of m6A erasers are summarized in Table II.
m6A readers
The reader is another participant in the dynamic and reversible regulation of m6A methylation, which can recognize and bind to m6A targets. The readers comprise the YTH domain family of proteins (YTHDC1/2, YTHDF1/2/3), insulin-like growth factor 2 mRNA binding protein (IGF2BP1/2/3), the heterokaryotic nuclear RNA protein family [heterogeneous nuclear ribonucleoprotein C (HNRNPC), HNRNPG] and eukaryotic initiation factor 3 (eIF3) (47). Diverse species rely on different m6A readers to perform specific biological functions.
The YTH family members are the most vital readers with a conservative m6A binding domain. YTHDF2 recruits RNA decay mechanism factor (C-C motif chemokine receptor 4-NOT deaminase complex) directly, which has an important role in accelerating the degradation of m6A-modified RNA (48). Unlike YTHDF2, YTHDF1 may neither take part in mRNA decay directly nor alter the total methylation mRNA level, but it facilitates translation by interacting with translation initiation factors and ribosomes (49,50). YTHDF3 is considered an assistant to boost the translation or degradation of target RNA with two other YTHDF members (51). YTHDC1 facilitates exon inclusion in the nucleus by recruiting splicing factor 3 (SRSF3) and preventing SRSF10 from binding to mRNA (52). It can also promote the nuclear export of m6A-labeled mRNA by interacting with nuclear transport receptors, and it is involved in promoting the stability of mRNA transcripts (47,53). YTHDC2 has 3′→5′ RNA helicase activity and improves the translation efficiency of target mRNA (54).
In the HNRNP family, HNRNPA2/B1 contains two RNA-specific recognition motifs and governs the directional sorting of miRNAs, thereby promoting primary miRNA processing (55). HNRNPC and HNRNPG can modulate mRNA abundance and splicing (43). IGF2BPs have been proven to be a unique and conservative family of m6A readers, which can enhance translation efficiency in an m6A-dependent way by regulating alternative splicing and improving stability (56). In addition, eIF3 promotes cap (m7GPPPN)-independent and YTHDF1-dependent mRNA translation (57). The functions of m6A readers are summarized in Table III. The functions of m6A regulators are illustrated in Fig. 1.
m6A and BC
Studies have indicated that intricate signal transduction processes at genetic, transcriptomic and epigenetic levels influence the occurrence and progression of cancer, including BC, which is often characterized by genetic and epigenetic alterations (58).
M6A methylation has been proven to regulate post-transcriptional gene expression through diverse mechanisms. Different m6A readers, writers and erasers interact and crosstalk with each other to activate or inhibit multiple carcinogenic pathways by regulating different targets (59). The abnormal expression of m6A mediators in BC is related to different BC subtypes and functions. Changing the degree of m6A modification may alter the cell cycle of BC cells and stimulate the proliferation, metastasis and invasion of BC cells by affecting the activity of downstream targets and various signaling pathways, such as the B-cell lymphoma-2 (Bcl-2) and phosphatidylinositol 3-kinase/Protein Kinase B (PI3K/Akt) pathways (60). The disorder of m6A regulators is universally existing in BC tissues. The following summarizes the main roles of some important m6A regulators in the occurrence and progression of BC.
METTL3 in BC
As an m6A methyltransferase, METTL3 has a crucial role in cancer. Mostly, METTL3 stimulates the occurrence and progression of diverse cancers as an oncogene, through depositing m6A modification on key transcripts (37). Numerous studies have validated that METTL3 expression is elevated in diverse cancerous tissues. However, the mechanism by which METTL3 promotes carcinogenesis may differ across various cancer types. The reported mechanisms mainly entail activating multiple m6A-dependent signaling pathways, increasing m6A modification of carcinogenic primary miR-25 and mediating the binding of m6A-modified target transcripts with specific cytokines, so as to promote mRNA translation or degradation, and ultimately facilitate tumor-cell proliferation and migration (61-64).
However, in certain cases, contrary results have been reported for similar tumors, implying that METTL3 may at times function as a tumor suppressor (65). For instance, certain researchers have detected that METTL3 methylation of basic leucine zipper ATF-like transcription factor (BATF) mRNA inhibits its expression in gastric cancer (GC), and low expression of BATF mRNA is significantly associated with postoperative recurrence of GC (66). In addition, there have been reports indicating that the knockdown of METTL3 significantly hastened tumor progression and reduced the lifespan of animals implanted with glioblastoma stem cells (67). Other studies have demonstrated that METTL3 expression is decreased in certain cases of renal cell carcinoma and bladder cancer (68,69). Shi et al (70) found that a low level of METTL3 in TNBC is indicative of a poor prognosis, suggesting that the reduced presence of m6A markers contributes to the progression of TNBC.
To date, certain studies on the mechanism underlying the role of METTL3 in BC have been published. The present study only provides a summary of recent findings. Wan et al (71) discovered that METTL3 enhances the m6A modification of programmed cell death ligand 1 (PD-L1) mRNA in BC cells, thereby improving the stability and expression of PD-L1 mRNA. Knocking down METTL3 can boost anti-tumor immunity and reduce PD-L1 expression, thus alleviating the progression of BC. Cai et al (72) have shown that the expression levels of METTL3 and hepatitis B x-interacting protein (HBXIP) are very high in BC tissues. HBXIP increases the expression of METTL3 through restraining the expression of tumor suppressor let-7g, and METTL3 in turn upregulates HBXIP via m6A modification, thus forming a positive feedback regulatory loop of HBXIP/let-7g/METTL3/HBXIP, and ultimately causing the malignant growth of BC cells (72).
It has also been observed that the METTL3 level in BC is significantly higher than that in surrounding normal tissues, particularly in patients with T3-T4 BC or lymph node metastasis (73). Studies revealed that METTL3 overexpression can upregulate enhancer of zeste homolog 2 through m6A modification. This process results in the suppression of tumor suppressor genes and promotion of epithelial-mesenchymal transformation (EMT), which triggers the occurrence, migration and invasion of BC cells (74,75). In addition, another study indicated that METTL3 can accelerate the proliferation of BC by regulating the methylation of BCL-2 or the metastasis associated lung adenocarcinoma transcript 1 (MALAT1)/miR-26b/high mobility group AT-hook 2 axis (76).
To sum up, METTL3 has been observed to be overexpressed in most BC samples, and its expression level appears to be positively correlated with the malignancy and metastasis of BC. The specific mechanism of the connection between METTL3 and BC-cell proliferation may involve multiple signaling pathways, but the exact mechanism requires to be further studied and clarified. The functions of METTL3 in BC are shown in Fig. 2.
KIAA1429 in BC
KIAA1429 acts as a scaffold for bridging the core protein of methyltransferase and it is also involved in the positive regulation of diverse tumorigenesis. Certain studies have indicated that KIAA1429 promotes the proliferation and growth of BC in a way independent of m6A, and the overall survival period of patients with BC is positively associated with KIAA1429 (77,78).
Zhang et al (78) found that KIAA1429 can improve the stability of structural maintenance of chromosomes 1A (SMC1A) mRNA via binding to the motif of SMC1A mRNA. Subsequently, SMC1A further increases snail family transcriptional repressor 1 (SNAIL) expression via binding to the promoter region of the SNAIL gene, which promotes the migration and invasion of BC. Another study illustrated that KIAA1429 targets to regulate cyclin-dependent kinase 1 (CDK1) (77), which is an oncogene related to the proliferation and metastasis of BC. The functions of KIAA1429 in BC are illustrated in Fig. 3.
FTO in BC
It is known that FTO, as an obesity-related protein, can catalyze the demethylation of m6A. Numerous studies have indicated that FTO is significantly upregulated in various cancerous tissues, including but not limited to cervical squamous cell carcinoma (79), lung squamous cell carcinoma (80), gastric cancer (81) and pancreatic cancer (82). FTO is involved in the regulation of tumor progression by decreasing the abundance of m6A and activating specific signaling pathways, reducing the overall survival rate of patients afflicted with malignant tumors (83). In a significant proportion of BC specimens, an elevated expression of FTO was observed compared to the adjacent normal breast tissue. Furthermore, it has been strongly associated with tumor proliferation, invasion and metastasis (83-85).
Niu et al (84) reported that, in MCF7 and MDA-MB231 cells, the expression of FTO was negatively correlated with BCL2 interacting protein 3 (BNIP3) in BC. Due to the overexpression of FTO in BC, the level of BNIP3 is downregulated, which is necessary for cell apoptosis (84). This change inhibits the cleavage of apoptosis factor caspase-3 and promotes the expression of anti-apoptotic protein Bcl-2 (86,87), thus reducing cell apoptosis and promoting the proliferation and colony formation of BC cells.
Xu et al (85) demonstrated that in SKBR3 and MDA-MB453 cells, FTO overexpression decreased the expression of miR-181b-3p, increasing the expression of ADP ribosylation factor like GTPase 5B (ARL5B) directly and indirectly. ARL5B subsequently drives the migration and invasion of HER2+ BC tissue.
Liu et al (88) indicated that FTO overexpression promotes aerobic glycolysis and increases ATP production via improving the activity of pyruvate kinase and hexokinase. Subsequently, the PI3K/AKT signaling pathway is abnormally activated, thus accelerating the progression of BC.
In conclusion, deregulation of FTO is a tumorigenic factor that cannot be ignored. The FTO-m6A axis can be considered a potential new target for the treatment and diagnosis of BC. The functions of FTO in BC are presented in Fig. 4.
ALKBH5 in BC
A growing body of evidence indicates that ALKBH5 is commonly dysregulated in malignant tumors, which regulates the expression of multiple oncogenes and contributes to tumor immune evasion through post-transcriptional mechanisms (89). However, studies indicated that ALKBH5 has a dual role in cancer, as its expression is not consistently upregulated or downregulated across all cancer types. Certain studies have shown a positive association between ALKBH5 levels and BC (90-92).
Under anoxic conditions, ALKBH5 mediates the pluripotency factor Nanog homeobox (NANOG) to regulate the BC stem cell characteristic specification in a hypoxia-inducible factor-dependent manner. In other words, ALKBH5 enhances the demethylation of NANOG mRNA and upregulates NANOG, while knocking down ALKBH5 inhibits this pluripotency factor (93,94). Therefore, ALKBH5 disorder is considered to be an important link in the proliferation, metastasis and enhancement of the stem cell phenotype of BC.
In addition, ALKBH5 upregulates the expression of ubiquitin conjugating enzyme E2 C (UBE2C) and reduces that of p53 by modifying the m6A of the downstream target gene UBE2C (91). Among them, UBE2C has been proven to exert a carcinogenic effect (95). The upregulated p53 is conducive to decreasing cancer cells and preventing the occurrence of cancer (96). Therefore, the ALKBH5/UBE2C/p53 axis is regarded as a potential mechanism for promoting the tumorigenesis and metastasis of TNBC cells (91).
In general, before ALKBH5 can be utilized as a therapeutic target for BC, its expression and specific regulatory mechanism should be further clarified. The functions of ALKBH5 in BC are presented in Fig. 5.
YTHDFs in BC
As a m6A binding protein, YTHDF1 amplification is universal in cancer tissues. The level of YTHDF1 is negatively associated with survival and positively correlated with the degree of malignancy and metastasis (97-99). In the experimental report by Sun et al (98), YTHDF1 and its downstream target transcription factor 8 (E2F8) were indicated to promote the transition to S-phase by regulating cell cycle-related factors, and to be involved in DNA replication and DNA damage repair (DDR). Furthermore, YTHDF1 blocked the cleavage of E2F8 mRNA, which is dependent on METTL14. All of these findings indicate that YTHDF1 functions as a promoter of tumor growth. As reported by Chen et al (97), low YTHDF1 restrained the proliferation, invasion and EMT of BC cells, and blocked cell-cycle progression. YTHDF1 also accelerated the translation of forkhead box (FOX)M1 by combining with m6A-modified FOXM1 mRNA, thus promoting its carcinogenic effect.
YTHDF2 can selectively bind m6A-modified sites and promote mRNA decay, but its function in solid tumors is still controversial. Recent reports have mentioned that YTHDF2 can degrade tumor promoter and tumor suppressor gene mRNA and have a dual role in tumor progression (100,101). For instance, YTHDF2 acts as a cancer-promoting regulator in certain tumors, such as glioblastoma, acute myeloid leukemia and prostate cancer (102-104). However, it has a tumor suppressor function in other tumor types, such as melanoma and liver cancer (100). As Einstein et al (105) suggested, moderate expression of YTHDF2 is essential to maintain the survival of cells driven by MYC proto-oncogene, bHLH transcription factor (MYC). Depletion of YTHDF2 activates the EMT-specific pathway in BC cells, particularly in TNBC, leading to further activation of cancer-related translation initiation factors. However, in MYC-addicted cells, over-translation of these target mRNAs eventually activates programmed cell death, leading to TNBC tumor-cell apoptosis. This result proves the importance of YTHDF2 for the survival of TNBC cells and the feasibility of knocking down YTHDF2 as a potential therapeutic method.
YTHDF3 may boost translation by interacting with ribosomal protein and significantly raise the translation efficiency of YTHDF1/3 common target (106). YTHDF3 can enhance the stability of its target factor zinc finger E-box binding homeobox 1 (ZEB1) mRNA, which is an EMT transcription factor (107). Chang et al (108)'s study on brain metastasis of BC indicated that YTHDF3 regulates its own mRNA translation by binding to m6A residues in its 5'UTR. YTHDF3 also combines with m6A-modified mRNA to promote the expression of brain metastasis genes, such as ST6 N-acetylgalactosaminide α-2,6-sialyltransferase 5, gap junction protein α1 and EGFR. It is noteworthy that in comparison to primary BC, YTHDF3 expression was significantly increased in its brain metastases, but not in other organs such as lung, bone, liver, spleen, lymph nodes and adrenal glands.
In conclusion, YTHDF disorder is a prevalent occurrence in cancer tissues. YTHDF1 and YTHDF3 are responsible for improving the translation efficiency of m6A-modified mRNA, and they are frequently amplified in BC cells. Their high levels are closely related to poor prognosis and low survival rates. Conversely, YTHDF2 promotes mRNA degradation and also acts as a carcinogen most of the time. It is plausible that the YTH family proteins work collaboratively to execute their regulatory role in translation, but their respective roles in cancer cannot be replaced, providing potential targets for BC treatment. The functions of YTHDFs in BC are displayed in Fig. 6.
IGF2BPs in BC
IGF2BP protein is a newly discovered m6A binding protein, which selectively binds to mRNA transcripts (109). The carcinogenic effect of IGF2BPs depends on its function of improving the stability and translation efficiency of certain oncogene mRNAs, such as MYC (109,110). The translocation of IGF2BPs may lead to the anomalous accumulation of carcinogenic products, thus stimulating the malignant development of cancer tissue (109).
Qiao et al (111) found that long intergenic ncRNA 483 (LINC00483) can promote the proliferation of BC cells and is negatively associated with the survival rate of patients with BC. A high level of IGF2BP1 significantly increased the expression of LINC00483, thus inducing carcinogenesis. Shi et al (112) reported that proto-oncogene MYCN activates IGF2BP1, and subsequently, IGF2BP1 enhances the stability of the carcinogen miR210HG and mediates its carcinogenic function in BC. According to the latest research, the ubiquitin specific peptidase 10 (USP10)/IGF2BP1/carnitine palmitoyl transfer 1A (CPT1A) axis plays an important role in BC metastasis (113). They found that the de-ubiquitination enzyme USP10 reduces its cleavage by de-ubiquitination of IGF2BP1. Subsequently, IGF2BP1 binds to the m6A site on CPT1A mRNA and makes it more stable, thus promoting the growth and metastasis of BC (114,115).
In BC, the level of PD-L1 increases with the increase of IGF2BP3. Knocking down IGF2BP3 significantly inhibited the expression of PD-L1, which cooperates with tumor cells to escape immune surveillance (70). In addition, the IGF2BP3/tripartite motif containing 25 (TRIM25)/miR-3614 axis represents a new way to regulate tumor cell proliferation. TRIM25 is mainly expressed in estrogen target tissues, which can improve cell viability and promote cell proliferation. MiR-3614-3p can be used as a tumor suppressor to inhibit the growth of BC cells. IGF2BP3 can induce the expression of TRIM25 and inhibit the maturation of miR-3614, which conversely protects TRIM25 mRNA from miR-3614-mediated degradation (116).
In short, the IGF2BP gene and its downstream targets are generally amplified in BC, thereby resulting in enhanced proliferation, metastasis and poor prognosis. These results provide a foundation for evaluating IGF2BP as a potential target for BC treatment, while the specific mechanism of IGF2BP should be further studied. The functions of IGF2BPs in BC are shown in Fig. 7.
Other m6A regulators in BC
The dysfunction of METTL14, WTAP, RBM15/15B and ZC3H13 in methyltransferase are also commonly recorded in cancer databases. METTL14 has been reported as an oncogene in most studies and its expression is usually positively correlated with the expression of METTL3 and WTAP. It can improve the stability of target mRNA through HuR (RNA-binding protein) mediation, involving in the regulation of cell cycle, EMT and other tumor growth processes (90). The expression level of WTAP in BC is higher than that in normal breast tissue, and it is positively correlated with tumor size and grade (117). Certain scholars have reported that the complement C5a receptor 1 (C5AR1)+/WTAP/enolase 1 (ENO1) axis regulates the glycolytic activity of BC cells and the lncRNA DLG-associated protein 1-antisense 1/miR-299-3p/WTAP axis promotes the proliferation of drug-resistant BC cells, which is worthy of further exploration (118,119). However, the function of WTAP in tumors cannot be separated from the expression of METTL3 (120).
HNRNPs, another family of m6A readers, are also involved in regulating various types of RNA processing, including translation and splicing (121). The tumor suppressor HNRNP E1 regulates the expression of EMT-related genes. Silencing HNRNP E1 increased BC-cell migration and endowed cells with stem cell characteristics, which promoted abnormal proliferation and metastatic growth of cancer cells (122). Knocking down HNRNP A1 accelerated cell death and reduced cell invasion (123). HNRNPC is upregulated in diverse cancers and HNRNPC silencing significantly suppressed BC-cell proliferation and tumor growth (124). The functions of these regulators in BC are illustrated in Fig. 8.
M6A modification and genomic instability in BC
Genomic instability is a hallmark of cancer and refers to the increased rate at which cells acquire genomic alterations (125). Certain regulatory factor-mediated m6A modifications have been linked to genomic instability, specifically in terms of regulating the effect of m6A modification on DNA damage and repair processes (126). While this relationship has been established and verified in numerous studies pertaining to tumors, investigations into its role in BC remain scarce.
M6A modification and genomic instability
METTL3 can be specifically recruited to gene fragments damaged by ultraviolet radiation and rapidly methylated RNA; subsequently, m6A-modified RNA starts the DDR pathway to improve the cell survival rate (126,127). METTL3-mediated m6A methylation also regulates homologous recombination (HR)-mediated double-stranded DNA break (DSB) repair (128). Phosphorylated METTL3 can be localized in the DSB region so that the damaged chromatin region of the RNA is modified by m6A. The m6A-modified RNA is then recognized by YTHDC1 and forms a DNA-RNA hybrid with DSBs, which recruits repair-related proteins and promotes HR-mediated repair (129). It has been reported that a low level of METTL3 increases the sensitivity of cancer cells to the treatment of DNA damage, while upregulated METTL3 reduces the survival rate of patients with head and neck squamous cell carcinoma who have received cisplatin or radiation treatment of DNA damage (126). Knocking down METTL3-mediated and YTHDC2-mediated m6A modification led to the accumulation of DNA-RNA hybridization (R loop) and γH2AX (a DSB marker), which plays a key role in inhibiting cell growth and regulating genome stability (130).
A study revealed that METTL3-mediated m6A modification improves the stability of transcription factor activated enhancer binding protein 2C mRNA, thus increasing the abundance of DNA repair genes, which endows spermatogonioma cells with resistance to DNA damage induced by cisplatin treatment, promoting tumor cell survival (131). METTL14 arginine methylation is positively correlated with enhanced translation of DNA repair genes (132). VIRMA was also demonstrated to enhance the invasion and cisplatin resistance of teratoma cells by regulating DNA damage (133). On the contrary, FTO participates in the upregulation of repair gene resection repair cross complementation group 1 through β-catenin mRNA demethylation, equipping cervical squamous cell carcinoma with radiochemotherapy resistance (79,134). ALKBH5 can also be inhibited by small ubiquitin-like modifier, which upregulates DNA repair genes and protects cells from reactive oxygen species (ROS)-induced DNA damage (126). This indicates that m6A plays a dual role in anti-cancer therapy based on DNA damage through DDR.
M6A modification is also involved in the regulation of telomere length and genomic integrity in human cancer (135). Telomere shortening is closely associated with cancer-related genomic changes (136). Homebox-containing protein 1 (HMBOX1) is a telomere-binding protein. HMBOX1 mRNA has been identified as the real target of m6A modification in cancer cells. HMBOX1 degradation caused by upregulation of METTL3 and YTHDF2 in cancer cells leads to telomere shortening and dysfunction of p53-dependent DNA damage response pathway inactivation. This change is likely to lead to various types of telomere-related chromosome aberrations, thus enhancing the tumorigenicity and invasiveness of cancer cells. Conversely, the malignant progression of cancer cells caused by METTL3-induced genomic instability can be alleviated or even reversed by introducing HMBOX1 (135).
M6A modification and genomic instability in BC
The pathogenesis of BC primarily entails the hyperactivation and overexpression of oncogenes, coupled with deficiencies in DDR gene defects, DDR gene transcription defects and mitotic defects, among others. The defective repair of damaged DNA leads to genomic instability, which is closely related to the malignant progress and poor prognosis of BC.
It has been found that tumor genome subtypes of BC are related to tumor gene expression, which involves the methylation gain and loss processes of a large number of loci (137). The researchers suggested that extensive aberrations in methylation induce epigenomic instability, rendering tumors more prone to regulatory mutation and deterioration. They even linked different methylation scores with higher epigenetic instability and higher chromosomal instability in BC, predicting the disease stage and progress (137).
Based on the above principles and experimental evidence, it may be reasoned that METTL3-mediated modification of m6DSB repair may serve as a promising target for cancer treatment, including BC. Whether targeted inhibition of METTL3 can reduce the proliferation activity and invasiveness of BC cells by inhibiting DNA repair or improve the sensitivity of BC cells to DNA damage therapies (such as chemotherapy or radiotherapy) is also likely to become a new topic. It may also be true for other m6A methylases and demethylases.
M6A modification and therapeutic resistance in BC
One of the main reasons for reduced efficacy of non-surgical treatment for tumors is drug resistance of tumor cells. Intrinsic resistance is mainly related to gene mutations, while acquired resistance refers to a weakened response to drugs after treatment, which may be related to secondary mutations in drug targets (138). In recent years, research on the role of m6A regulators in drug resistance in cancer treatment has made significant progress, which has also been confirmed in the treatment of BC (139).
Tamoxifen chemotherapy, as a first-line endocrine therapy option for BC, is facing a major problem of drug resistance. Research has proved that long-term exposure to tamoxifen can induce an increase in METTL3 expression, further resulting in an increase in m6A of the 5'UTR of adenylate kinase 4 (AK4; a mitochondrial nucleotide kinase) mRNA. High levels of AK4 inhibit mitochondrial apoptosis and promote ROS production, activating p38, ultimately leading to increased resistance of MCF-7 cells to tamoxifen (140). High-expression HNRNPA2B1 in endocrine-resistant MCF-7 and LCC9 BC cell lines endows cancer cells with acquired endocrine resistance by activating the Ser/Thr kinase growth factor signaling pathway that regulates its downstream target (141). The increased expression of activating transcription factor 3 (ATF3) protein caused by low levels of YTHDF2 is also the reason for the development of tamoxifen-resistant MCF-7 cells (142). Therefore, selective inhibition of AK4, HNRNPA2B1 and ATF3 may serve as a potential strategy for preventing BC cells from acquiring endocrine therapy resistance.
Similarly, the abnormal expression of m6A regulatory factors can make BC cells resistant to certain chemotherapy drugs. Anthracyclines have been considered to be the most effective chemotherapeutic drugs for BC in recent years, but are facing serious drug resistance problems. Research has shown that miR-221-3p is an miRNA involved in tumor development, metastasis and drug resistance. High levels of METTL3 increase the expression of miR221-3p and negatively regulate homeodomain interacting protein kinase 2, a tumor suppressor that can be activated by doxorubicin, thereby reducing the efficacy of doxorubicin (143). The latest research shows that METTL3 and YTHDC1 promote the synthesis of EGF and DNA repair protein RAD51 recombinase (RAD51) and improve HR and cell survival during doxorubicin treatment, resulting in drug resistance of BC cells by co-regulating m6A-modified related mRNA (144). Li et al (145) found that the m6A modification of METTL3 increased the level of MALAT1 protein, recruited E2F1 and activated the transcription of downstream anterior gradient 2, protein disulphide isomerase family member, contributing to doxorubicin resistance in BC. Wu et al (146) found that ALKBH5 removes m6A modification to stabilize BRCA1 (DNA repair protein) mRNA, further enhance its DNA repair ability and increase the resistance of BC cells to doxorubicin. Wang et al (147) have shown that FTO mediates doxorubicin resistance in BC by activating signal converters such as transcription activator STAT3 in BC. ALKBH5-mediated FOXO1 m6A demethylation increases the expression of superoxide dismutase 2 and leads to a lower ROS level, thus promoting the maintenance of cancer stem cell characteristics and doxorubicin resistance in TNBC. Furthermore, a study indicated that targeted inhibition of FOXO1 both in vivo and in vitro can restore the drug sensitivity of TNBC (148). The exosomal Piwi-interacting RNA-17560 derived from senescent neutrophils enhances the stability and expression of ZEB1 transcript by upregulating FTO levels, leading to chemical resistance and EMT in tumor cells (149).
Radiation resistance refers to the adaptability of tumor cells or tissues to radiation therapy. DDR is one of the main reasons for tumor cells to develop radiation resistance (150). The transmembrane glycoprotein neuropilin 1 (NRP1) can enhance the stem cell characteristics of BC cells, making them resistant to radiation therapy. NRP1 has been shown to reduce cell apoptosis and enhance radiation resistance by downregulating Bcl-2 through m6A methyltransferase WTAP (151).
Summary and outlook
So far, the role of m6A methylation in cancer remains in the preliminary stages of research and its application in clinical targeted therapy is limited. The available studies consistently show that m6A is subject to dynamic reversible modification by three distinct types of regulators, which can effectively regulate mRNA splicing, translation, stability and decay. Through specific mRNA modification, m6A can regulate the expression of target genes and consequently impact tumor progression. However, these target genes contain oncogenes and tumor suppressor genes, and the change trend of m6A regulatory factors differs from one type of cancer tissue to another, which means that m6A plays a dual role in tumor progression.
Furthermore, the interaction between m6A methylation and tumor metabolism is complex. Tumor metabolic stress can abnormally regulate m6A methylation, while disorder of m6A methylation can in turn regulate signaling pathways related to tumor metabolism. Other studies have found that m6A is involved in regulating the metabolic reprogramming of BC (7). For instance, C5AR1-positive neutrophils are capable of secreting IL-1β and TNF-α, which activate ERK1/2 signaling to enhance WTAP stability. The upregulated WTAP subsequently elevates the expression of ENO1, thereby promoting glycolysis and ultimately facilitating the progression of BC (118). In addition, breast tumor cells develop resistance to radiation, chemotherapy and endocrine therapy drugs due to the abnormal expression of certain m6A regulatory factors and regulation of specific signaling pathways. Therefore, targeting upregulation or downregulation of certain m6A-related genes and activating or inhibiting certain m6A regulatory factors can enhance the sensitivity of tumors to radiotherapy, chemotherapy and endocrine therapy (150). It provides an innovative idea for the combination treatment of BC and the development of new drugs.
At present, joint research on m6A and BC focus on investigating the correlation and specific regulatory mechanism linking the expression of certain m6A regulatory factors with their corresponding target genes, as well as with various BC subtypes, malignancy, metabolism, growth, metastasis, immunity, drug resistance and adverse prognosis. While the experimental findings hold some informative value, their reliability and applicability necessitate further validation through larger BC sample sizes and clinical implementation. In conclusion, research regarding m6A and BC holds promise for yielding diagnostic and therapeutic breakthroughs in the treatment of BC. However, these models and methods necessitate further refinement and the mechanism by which m6A-related proteins regulate the progression of BC is required to be further explored.
The majority of the experimental findings indicate that m6A regulators are imbalanced in BC. Based on the function of m6A modification in tumor tissue and the close correlation between m6A regulatory factors and tumors, many scholars claim that targeted m6A methylchemotherapy is likely to become a promising option for tumor treatment. Consequently, exploring the optimal strategy and conducting clinical trials on the combination of m6A enzyme-related drugs and m6A targeted therapy will likely represent a novel direction for BC treatment in the future. These strategies require the determination of the specific pathways and mechanisms through which m6A impacts the progression of BC, serving as the theoretical foundation. This paper summarizes the relevant mechanisms that have been found so far, yet it should be acknowledged that numerous challenges remain to be overcome.
First, m6A regulates the progression of BC by complex mechanisms, involving a variety of regulatory factors, signaling pathways and oncogenes. Modifying any of these pathways may trigger a series of associated reactions, which should be predicted before implementing targeted therapy. Furthermore, certain m6A regulators play dual roles in BC, so it is necessary to consider tumor heterogeneity and specific maladjustment factors before designing a treatment plan.
In addition, a variety of serious problems should be considered when screening potential targeted drugs. For instance, the specific ways that m6A-related drugs affect the methylation level, whether these drugs have cytotoxicity, whether they are generally applicable to different subtypes of BC and how to deal with BC resistance should be determined. In addition, it is worth noting that different subtypes of BC exhibit varying degrees of different sensitivity to radiotherapy, chemotherapy, immunotherapy and various drugs, and they are regulated by m6A modification, which greatly affects the treatment effect.
Finally, in-depth exploration of cancer epigenomics and the advancement of high-quality nucleic acid probes facilitate the precise identification of biomarkers, which is essential for predicting potential therapeutic targets, individualized treatment and improvement of prognosis. By resolving these challenges, the prospect of m6A targeted therapy for BC will expand significantly.
Availability of data and materials
Not applicable.
Authors' contributions
YBY was a major contributor in writing the manuscript. FG and LQR contributed to the information retrieval and selection. NR and JNP contributed to editing of the figures. QPX proposed the writing ideas for this article and conducted a final review. All authors read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Acknowledgements
Not applicable.
Funding
This study was funded by the Key Medical Discipline of Hangzhou City (grant no. 2021-21); Key Medical Discipline of Zhejiang Province (grant no. 2018-2-3); Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province (grant no. 2020E10021); Medical and Health Science and Technology Program of Zhejiang Province (grant no. 2023KY933); and the Traditional Chinese Medicine Science and Technology Project of Zhejiang Province (grant no. 2023ZL565).
References
Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin. 70:7–30. 2020. View Article : Google Scholar : PubMed/NCBI | |
Patel S: Breast cancer: Lesser-known facets and hypotheses. Biomed Pharmacother. 98:499–506. 2018. View Article : Google Scholar | |
Zhu Z, Albadawy E, Saha A, Zhang J, Harowicz MR and Mazurowski MA: Deep learning for identifying radiogenomic associations in breast cancer. Comput Biol Med. 109:85–90. 2019. View Article : Google Scholar : PubMed/NCBI | |
Lord SJ, Bahlmann K, O'Connell DL, Kiely BE, Daniels B, Pearson SA, Beith J, Bulsara MK and Houssami N: De novo and recurrent metastatic breast cancer-A systematic review of population-level changes in survival since 1995. EClinicalMedicine. 44:1012822022. View Article : Google Scholar | |
Dai D, Wang H, Zhu L, Jin H and Wang X: N6-methyladenosine links RNA metabolism to cancer progression. Cell Death Dis. 9:1242018. View Article : Google Scholar : PubMed/NCBI | |
Hyun K, Jeon J, Park K and Kim J: Writing, erasing and reading histone lysine methylations. Exp Mol Med. 49:e3242017. View Article : Google Scholar : PubMed/NCBI | |
An Y and Duan H: The role of m6A RNA methylation in cancer metabolism. Mol Cancer. 21:142022. View Article : Google Scholar : PubMed/NCBI | |
Lin H, Wang Y, Wang P, Long F and Wang T: Mutual regulation between N6-methyladenosine (m6A) modification and circular RNAs in cancer: Impacts on therapeutic resistance. Mol Cancer. 21:1482022. View Article : Google Scholar : PubMed/NCBI | |
Liu Z, Zou H, Dang Q, Xu H, Liu L, Zhang Y, Lv J, Li H, Zhou Z and Han X: Biological and pharmacological roles of m6A modifications in cancer drug resistance. Mol Cancer. 21:2202022. View Article : Google Scholar | |
Deng LJ, Deng WQ, Fan SR, Chen MF, Qi M, Lyu WY, Qi Q, Tiwari AK, Chen JX, Zhang DM and Chen ZS: m6A modification: Recent advances, anticancer targeted drug discovery and beyond. Mol Cancer. 21:522022. View Article : Google Scholar : PubMed/NCBI | |
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI | |
DeSantis CE, Ma J, Gaudet MM, Newman LA, Miller KD, Goding Sauer A, Jemal A and Siegel RL: Breast cancer statistics, 2019. CA Cancer J Clin. 69:438–451. 2019. View Article : Google Scholar : PubMed/NCBI | |
Loibl S, Poortmans P, Morrow M, Denkert C and Curigliano G: Breast cancer. Lancet. 397:1750–1769. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hong R and Xu B: Breast cancer: an up-to-date review and future perspectives. Cancer Commun (Lond). 42:913–936. 2022. View Article : Google Scholar : PubMed/NCBI | |
Golshan M, Loibl S, Wong SM, Houber JB, O'Shaughnessy J, Rugo HS, Wolmark N, McKee MD, Maag D, Sullivan DM, et al: Breast conservation after neoadjuvant chemotherapy for triple-negative breast cancer: Surgical results from the brightness randomized clinical trial. JAMA Surg. 155:e1954102020. View Article : Google Scholar : PubMed/NCBI | |
Waks AG and Winer EP: Breast cancer treatment: A review. JAMA. 321:288–300. 2019. View Article : Google Scholar : PubMed/NCBI | |
Jääskeläinen A, Roininen N, Karihtala P and Jukkola A: High parity predicts poor outcomes in patients with luminal B-like (HER2 negative) early breast cancer: A prospective finnish single-center study. Front Oncol. 10:14702020. View Article : Google Scholar : PubMed/NCBI | |
Choong GM, Cullen GD and O'Sullivan CC: Evolving standards of care and new challenges in the management of HER2-positive breast cancer. CA Cancer J Clin. 70:355–374. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yu KD, Ye FG, He M, Fan L, Ma D, Mo M, Wu J, Liu GY, Di GH, Zeng XH, et al: Effect of adjuvant paclitaxel and carboplatin on survival in women with triple-negative breast cancer: A phase 3 randomized clinical trial. JAMA Oncol. 6:1390–1396. 2020. View Article : Google Scholar : PubMed/NCBI | |
Garrido-Castro AC, Lin NU and Polyak K: Insights into molecular classifications of triple-negative breast cancer: Improving patient selection for treatment. Cancer Discov. 9:176–198. 2019. View Article : Google Scholar : PubMed/NCBI | |
Gaudet MM, Gierach GL, Carter BD, Luo J, Milne RL, Weiderpass E, Giles GG, Tamimi RM, Eliassen AH, Rosner B, et al: Pooled analysis of nine cohorts reveals breast cancer risk factors by tumor molecular subtype. Cancer Res. 78:6011–6021. 2018. View Article : Google Scholar : PubMed/NCBI | |
Nur U, El Reda D, Hashim D and Weiderpass E: A prospective investigation of oral contraceptive use and breast cancer mortality: Findings from the Swedish women's lifestyle and health cohort. BMC Cancer. 19:8072019. View Article : Google Scholar : PubMed/NCBI | |
Trabert B, Sherman ME, Kannan N and Stanczyk FZ: Progesterone and breast cancer. Endocr Rev. 41:320–344. 2020. View Article : Google Scholar : | |
Reiner AS, Sisti J, John EM, Lynch CF, Brooks JD, Mellemkjær L, Boice JD, Knight JA, Concannon P, Capanu M, et al: Breast cancer family history and contralateral breast cancer risk in young women: an update from the women's environmental cancer and radiation epidemiology study. J Clin Oncol. 36:1513–1520. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ho PJ, Ho WK, Khng AJ, Yeoh YS, Tan BK, Tan EY, Lim GH, Tan SM, Tan VKM, Yip CH, et al: Overlap of high-risk individuals predicted by family history, and genetic and non-genetic breast cancer risk prediction models: Implications for risk stratification. BMC Med. 20:1502022. View Article : Google Scholar : PubMed/NCBI | |
Lu HM, Li S, Black MH, Lee S, Hoiness R, Wu S, Mu W, Huether R, Chen J, Sridhar S, et al: Association of breast and ovarian cancers with predisposition genes identified by large-scale sequencing. JAMA Oncol. 5:51–57. 2019. View Article : Google Scholar : | |
Breast Cancer Association Consortium; Dorling L, Carvalho S, Allen J, González-Neira A, Luccarini C, Wahlström C, Pooley KA, Parsons MT, Fortuno C, et al: Breast cancer risk genes-association analysis in more than 113,000 women. N Engl J Med. 384:428–439. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ru W, Zhang X, Yue B, Qi A, Shen X, Huang Y, Lan X, Lei C and Chen H: Insight into m6A methylation from occurrence to functions. Open Biol. 10:2000912020. View Article : Google Scholar | |
Li Z, Peng Y, Li J, Chen Z, Chen F, Tu J, Lin S and Wang H: N6-methyladenosine regulates glycolysis of cancer cells through PDK4. Nat Commun. 11:25782020. View Article : Google Scholar | |
Huang H, Weng H and Chen J: m6A modification in coding and non-coding RNAs: Roles and therapeutic implications in cancer. Cancer Cell. 37:270–288. 2020. View Article : Google Scholar : PubMed/NCBI | |
Choe J, Lin S, Zhang W, Liu Q, Wang L, Ramirez-Moya J, Du P, Kim W, Tang S, Sliz P, et al: mRNA circularization by METTL3-eIF3h enhances translation and promotes oncogenesis. Nature. 561:556–560. 2018. View Article : Google Scholar : PubMed/NCBI | |
Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, Cesarkas K, Jacob-Hirsch J, Amariglio N, Kupiec M, et al: Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 485:201–206. 2012. View Article : Google Scholar : PubMed/NCBI | |
Shi B, Liu WW, Yang K, Jiang GM and Wang H: The role, mechanism, and application of RNA methyltransferase METTL14 in gastrointestinal cancer. Mol Cancer. 21:1632022. View Article : Google Scholar : PubMed/NCBI | |
Zaccara S, Ries RJ and Jaffrey SR: Reading, writing and erasing mRNA methylation. Nat Rev Mol Cell Biol. 20:608–624. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chen H, Wang Y, Su H, Zhang X, Chen H and Yu J: RNA N6-methyladenine modification, cellular reprogramming, and cancer stemness. Front Cell Dev Biol. 10:9352242022. View Article : Google Scholar | |
Wang X, Feng J, Xue Y, Guan Z, Zhang D, Liu Z, Gong Z, Wang Q, Huang J, Tang C, et al: Structural basis of N(6)-adenosine methylation by the METTL3-METTL14 complex. Nature. 534:575–578. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zeng C, Huang W, Li Y and Weng H: Roles of METTL3 in cancer: mechanisms and therapeutic targeting. J Hematol Oncol. 13:1172020. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Peng C, Chen J, Chen D, Yang B, He B, Hu W, Zhang Y, Liu H, Dai L, et al: WTAP facilitates progression of hepatocellular carcinoma via m6A-HuR-dependent epigenetic silencing of ETS1. Mol Cancer. 18:1272019. View Article : Google Scholar : PubMed/NCBI | |
Su R, Dong L, Li Y, Gao M, He PC, Liu W, Wei J, Zhao Z, Gao L, Han L, et al: METTL16 exerts an m6A-independent function to facilitate translation and tumorigenesis. Nat Cell Biol. 24:205–216. 2022. View Article : Google Scholar : PubMed/NCBI | |
Hu Y, Ouyang Z, Sui X, Qi M, Li M, He Y, Cao Y, Cao Q, Lu Q, Zhou S, et al: Oocyte competence is maintained by m6A methyltransferase KIAA1429-mediated RNA metabolism during mouse follicular development. Cell Death Differ. 27:2468–2483. 2020. View Article : Google Scholar : PubMed/NCBI | |
Tan C, Xia P, Zhang H, Xu K, Liu P, Guo D and Liu Z: YY1-Targeted RBM15B promotes hepatocellular carcinoma cell proliferation and sorafenib resistance by promoting TRAM2 expression in an m6A-dependent manner. Front Oncol. 12:8730202022. View Article : Google Scholar : PubMed/NCBI | |
Wen J, Lv R, Ma H, Shen H, He C, Wang J, Jiao F, Liu H, Yang P, Tan L, et al: Zc3h13 regulates nuclear RNA m6A methylation and mouse embryonic stem cell self-renewal. Mol Cell. 69:1028–1038.e6. 2018. View Article : Google Scholar | |
Wang T, Kong S, Tao M and Ju S: The potential role of RNA N6-methyladenosine in Cancer progression. Mol Cancer. 19:882020. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Song C, Wang N, Li S, Liu Q, Sun Z, Wang K, Yu SC and Yang Q: NADP modulates RNA m6A methylation and adipogenesis via enhancing FTO activity. Nat Chem Biol. 16:1394–1402. 2020. View Article : Google Scholar : PubMed/NCBI | |
Bartosovic M, Molares HC, Gregorova P, Hrossova D, Kudla G and Vanacova S: N6-methyladenosine demethylase FTO targets pre-mRNAs and regulates alternative splicing and 3'-end processing. Nucleic Acids Res. 45:11356–11370. 2017. View Article : Google Scholar : PubMed/NCBI | |
Tang B, Yang Y, Kang M, Wang Y, Wang Y, Bi Y, He S and Shimamoto F: m6A demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Mol Cancer. 19:32020. View Article : Google Scholar | |
Jiang X, Liu B, Nie Z, Duan L, Xiong Q, Jin Z, Yang C and Chen Y: The role of m6A modification in the biological functions and diseases. Signal Transduct Target Ther. 6:742021. View Article : Google Scholar : PubMed/NCBI | |
Du H, Zhao Y, He J, Zhang Y, Xi H, Liu M, Ma J and Wu L: YTHDF2 destabilizes m(6)A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat Commun. 7:126262016. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Zhao BS, Roundtree IA, Lu Z, Han D, Ma H, Weng X, Chen K, Shi H and He C: N(6)-methyladenosine modulates messenger RNA translation efficiency. Cell. 161:1388–1399. 2015. View Article : Google Scholar : PubMed/NCBI | |
Chen Z, Zhong X, Xia M and Zhong J: The roles and mechanisms of the m6A reader protein YTHDF1 in tumor biology and human diseases. Mol Ther Nucleic Acids. 26:1270–1279. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zaccara S and Jaffrey SR: A unified model for the function of YTHDF proteins in regulating m6A-modified mRNA. Cell. 181:1582–1595.e18. 2020. View Article : Google Scholar | |
Xiao W, Adhikari S, Dahal U, Chen YS, Hao YJ, Sun BF, Sun HY, Li A, Ping XL, Lai WY, et al: Nuclear m(6)A reader YTHDC1 regulates mRNA splicing. Mol Cell. 61:507–519. 2016. View Article : Google Scholar : PubMed/NCBI | |
Roundtree IA, Luo GZ, Zhang Z, Wang X, Zhou T, Cui Y, Sha J, Huang X, Guerrero L, Xie P, et al: YTHDC1 mediates nuclear export of N6-methyladenosine methylated mRNAs. Elife. 6:e313112017. View Article : Google Scholar | |
Mao Y, Dong L, Liu XM, Guo J, Ma H, Shen B and Qian SB: m6A in mRNA coding regions promotes translation via the RNA helicase-containing YTHDC2. Nat Commun. 10:53322019. View Article : Google Scholar | |
Wu B, Su S, Patil DP, Liu H, Gan J, Jaffrey SR and Ma J: Molecular basis for the specific and multivariant recognitions of RNA substrates by human hnRNP A2/B1. Nat Commun. 9:4202018. View Article : Google Scholar : PubMed/NCBI | |
Sun CY, Cao D, Du BB, Chen CW and Liu D: The role of Insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs) as m6A readers in cancer. Int J Biol Sci. 18:2744–2758. 2022. View Article : Google Scholar : | |
Liu T, Wei Q, Jin J, Luo Q, Liu Y, Yang Y, Cheng C, Li L, Pi J, Si Y, et al: The m6A reader YTHDF1 promotes ovarian cancer progression via augmenting EIF3C translation. Nucleic Acids Res. 48:3816–3831. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Zhang Y, Du Y, Zhou M, Hu Y and Zhang S: Emerging roles of N6-methyladenosine (m6A) modification in breast cancer. Cell Biosci. 10:1362020. View Article : Google Scholar | |
Li Y, Xiao J, Bai J, Tian Y, Qu Y, Chen X, Wang Q, Li X, Zhang Y and Xu J: Molecular characterization and clinical relevance of m6A regulators across 33 cancer types. Mol Cancer. 18:1372019. View Article : Google Scholar | |
Wei M, Bai JW, Niu L, Zhang YQ, Chen HY and Zhang GJ: The complex roles and therapeutic implications of m6A modifications in breast cancer. Front Cell Dev Biol. 8:6150712021. View Article : Google Scholar | |
Han H, Yang C, Zhang S, Cheng M, Guo S, Zhu Y, Ma J, Liang Y, Wang L, Zheng S, et al: METTL3-mediated m6A mRNA modification promotes esophageal cancer initiation and progression via Notch signaling pathway. Mol Ther Nucleic Acids. 26:333–346. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chen M, Wei L, Law CT, Tsang FH, Shen J, Cheng CL, Tsang LH, Ho DW, Chiu DK, Lee JM, et al: RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 67:2254–2270. 2018. View Article : Google Scholar | |
Jin H, Ying X, Que B, Wang X, Chao Y, Zhang H, Yuan Z, Qi D, Lin S, Min W, et al: N6-methyladenosine modification of ITGA6 mRNA promotes the development and progression of bladder cancer. EBioMedicine. 47:195–207. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Bai R, Li M, Ye H, Wu C, Wang C, Li S, Tan L, Mai D, Li G, et al: Excessive miR-25-3p maturation via N6-methyladenosine stimulated by cigarette smoke promotes pancreatic cancer progression. Nat Commun. 10:18582019. View Article : Google Scholar | |
Ma S, Chen C, Ji X, Liu J, Zhou Q, Wang G, Yuan W, Kan Q and Sun Z: The interplay between m6A RNA methylation and noncoding RNA in cancer. J Hematol Oncol. 12:1212019. View Article : Google Scholar : PubMed/NCBI | |
Xie JW, Huang XB, Chen QY, Ma YB, Zhao YJ, Liu LC, Wang JB, Lin JX, Lu J, Cao LL, et al: m6A modification-mediated BATF2 acts as a tumor suppressor in gastric cancer through inhibition of ERK signaling. Mol Cancer. 19:1142020. View Article : Google Scholar | |
Cui Q, Shi H, Ye P, Li L, Qu Q, Sun G, Sun G, Lu Z, Huang Y, Yang CG, et al: m6A RNA methylation regulates the self-renewal and tumorigenesis of glioblastoma stem cells. Cell Rep. 18:2622–2634. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zheng W, Dong X, Zhao Y, Wang S, Jiang H, Zhang M, Zheng X and Gu M: Multiple functions and mechanisms underlying the role of METTL3 in human cancers. Front Oncol. 9:14032019. View Article : Google Scholar | |
Wang G, Dai Y, Li K, Cheng M, Xiong G, Wang X, Chen S, Chen Z, Chen J, Xu X, et al: Deficiency of Mettl3 in bladder cancer stem cells inhibits bladder cancer progression and angiogenesis. Front Cell Dev Biol. 9:6277062021. View Article : Google Scholar : PubMed/NCBI | |
Shi Y, Zheng C, Jin Y, Bao B, Wang D, Hou K, Feng J, Tang S, Qu X, Liu Y, et al: Reduced expression of METTL3 promotes metastasis of triple-negative breast cancer by m6A methylation-mediated COL3A1 up-regulation. Front Oncol. 10:11262020. View Article : Google Scholar : PubMed/NCBI | |
Wan W, Ao X, Chen Q, Yu Y, Ao L, Xing W, Guo W, Wu X, Pu C, Hu X, et al: METTL3/IGF2BP3 axis inhibits tumor immune surveillance by upregulating N6-methyladenosine modification of PD-L1 mRNA in breast cancer. Mol Cancer. 21:602022. View Article : Google Scholar | |
Cai X, Wang X, Cao C, Gao Y, Zhang S, Yang Z, Liu Y, Zhang X, Zhang W and Ye L: HBXIP-elevated methyltransferase METTL3 promotes the progression of breast cancer via inhibiting tumor suppressor let-7g. Cancer Lett. 415:11–19. 2018. View Article : Google Scholar | |
Ma J, Zhang J, Weng YC and Wang JC: EZH2-mediated microRNA-139-5p regulates epithelial-mesenchymal transition and lymph node metastasis of pancreatic cancer. Mol Cells. 41:868–880. 2018.PubMed/NCBI | |
Hu S, Song Y, Zhou Y, Jiao Y and Li G: METTL3 accelerates breast cancer progression via regulating EZH2 m6A modification. J Healthc Eng. 2022:57944222022. | |
Li W, Xue D, Xue M, Zhao J, Liang H, Liu Y and Sun T: Fucoidan inhibits epithelial-to-mesenchymal transition via regulation of the HIF-1α pathway in mammary cancer cells under hypoxia. Oncol Lett. 18:330–338. 2019.PubMed/NCBI | |
Zhao C, Ling X, Xia Y, Yan B and Guan Q: The m6A methyltransferase METTL3 controls epithelial-mesenchymal transition, migration and invasion of breast cancer through the MALAT1/miR-26b/HMGA2 axis. Cancer Cell Int. 21:4412021. View Article : Google Scholar : PubMed/NCBI | |
Qian JY, Gao J, Sun X, Cao MD, Shi L, Xia TS, Zhou WB, Wang S, Ding Q and Wei JF: KIAA1429 acts as an oncogenic factor in breast cancer by regulating CDK1 in an N6-methyladenosine-independent manner. Oncogene. 38:6123–6141. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Dai XY, Qian JY, Xu F, Wang ZW, Xia T, Zhou XJ, Li XX, Shi L, Wei JF and Ding Q: SMC1A regulated by KIAA1429 in m6A-independent manner promotes EMT progress in breast cancer. Mol Ther Nucleic Acids. 27:133–146. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhou S, Bai ZL, Xia D, Zhao ZJ, Zhao R, Wang YY and Zhe H: FTO regulates the chemo-radiotherapy resistance of cervical squamous cell carcinoma (CSCC) by targeting β-catenin through mRNA demethylation. Mol Carcinog. 57:590–597. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Ren D, Du Z, Wang H, Zhang H and Jin Y: m6A demethylase FTO facilitates tumor progression in lung squamous cell carcinoma by regulating MZF1 expression. Biochem Biophys Res Commun. 502:456–464. 2018. View Article : Google Scholar : PubMed/NCBI | |
Shimura T, Kandimalla R, Okugawa Y, Ohi M, Toiyama Y, He C and Goel A: Novel evidence for m6A methylation regulators as prognostic biomarkers and FTO as a potential therapeutic target in gastric cancer. Br J Cancer. 126:228–237. 2022. View Article : Google Scholar | |
Azzam SK, Alsafar H and Sajini AA: FTO m6A demethylase in obesity and cancer: implications and underlying molecular mechanisms. Int J Mol Sci. 23:38002022. View Article : Google Scholar : PubMed/NCBI | |
Zheng QK, Ma C, Ullah I, Hu K, Ma RJ, Zhang N and Sun ZG: Roles of N6-methyladenosine demethylase FTO in malignant tumors progression. Onco Targets Ther. 14:4837–4846. 2021. View Article : Google Scholar : PubMed/NCBI | |
Niu Y, Lin Z, Wan A, Chen H, Liang H, Sun L, Wang Y, Li X, Xiong XF, Wei B, et al: RNA N6-methyladenosine demethylase FTO promotes breast tumor progression through inhibiting BNIP3. Mol Cancer. 18:462019. View Article : Google Scholar : PubMed/NCBI | |
Xu Y, Ye S, Zhang N, Zheng S, Liu H, Zhou K, Wang L, Cao Y, Sun P and Wang T: The FTO/miR-181b-3p/ARL5B signaling pathway regulates cell migration and invasion in breast cancer. Cancer Commun (Lond). 40:484–500. 2020. View Article : Google Scholar : PubMed/NCBI | |
Basu A: The interplay between apoptosis and cellular senescence: Bcl-2 family proteins as targets for cancer therapy. Pharmacol Ther. 230:1079432022. View Article : Google Scholar | |
Gao X, Wang Y, Lu F, Chen X, Yang D, Cao Y, Zhang W, Chen J, Zheng L, Wang G, et al: Extracellular vesicles derived from oesophageal cancer containing P4HB promote muscle wasting via regulating PHGDH/Bcl-2/caspase-3 pathway. J Extracell Vesicles. 10:e120602021. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Wang R, Zhang L, Li J, Lou K and Shi B: The lipid metabolism gene FTO influences breast cancer cell energy metabolism via the PI3K/AKT signaling pathway. Oncol Lett. 13:4685–4690. 2017. View Article : Google Scholar : PubMed/NCBI | |
Qu J, Yan H, Hou Y, Cao W, Liu Y, Zhang E, He J and Cai Z: RNA demethylase ALKBH5 in cancer: From mechanisms to therapeutic potential. J Hematol Oncol. 15:82022. View Article : Google Scholar : PubMed/NCBI | |
Wu L, Wu D, Ning J, Liu W and Zhang D: Changes of N6-methyladenosine modulators promote breast cancer progression. BMC Cancer. 19:3262019. View Article : Google Scholar : PubMed/NCBI | |
Hu Y, Liu H, Xiao X, Yu Q, Deng R, Hua L, Wang J and Wang X: Bone marrow mesenchymal stem cell-derived exosomes inhibit triple-negative breast cancer cell stemness and metastasis via an ALKBH5-dependent mechanism. Cancers (Basel). 14:60592022. View Article : Google Scholar : PubMed/NCBI | |
Fry NJ, Law BA, Ilkayeva OR, Carraway KR and Mansfield KD: N6-methyladenosine contributes to cellular phenotype in a genetically-defined model of breast cancer progression. Oncotarget. 9:31231–31243. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhang C, Samanta D, Lu H, Bullen JW, Zhang H, Chen I, He X and Semenza GL: Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. Proc Natl Acad Sci USA. 113:E2047–E2056. 2016. | |
Zhang C, Zhi WI, Lu H, Samanta D, Chen I, Gabrielson E and Semenza GL: Hypoxia-inducible factors regulate pluripotency factor expression by ZNF217- and ALKBH5-mediated modulation of RNA methylation in breast cancer cells. Oncotarget. 7:64527–64542. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zhang S, You X, Zheng Y, Shen Y, Xiong X and Sun Y: The UBE2C/CDH1/DEPTOR axis is an oncogene and tumor suppressor cascade in lung cancer cells. J Clin Invest. 133:e1624342023. View Article : Google Scholar : | |
Wang Y, Xie Y, Niu Y, Song P, Liu Y, Burnett J, Yang Z, Sun D, Ran Y, Li Y and Sun L: Carboxypeptidase A4 negatively correlates with p53 expression and regulates the stemness of breast cancer cells. Int J Med Sci. 18:1753–1759. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chen H, Yu Y, Yang M, Huang H, Ma S, Hu J, Xi Z, Guo H, Yao G, Yang L, et al: YTHDF1 promotes breast cancer progression by facilitating FOXM1 translation in an m6A-dependent manner. Cell Biosci. 12:192022. View Article : Google Scholar : PubMed/NCBI | |
Sun Y, Dong D, Xia Y, Hao L, Wang W and Zhao C: YTHDF1 promotes breast cancer cell growth, DNA damage repair and chemoresistance. Cell Death Dis. 13:2302022. View Article : Google Scholar : PubMed/NCBI | |
Anita R, Paramasivam A, Priyadharsini JV and Chitra S: The m6A readers YTHDF1 and YTHDF3 aberrations associated with metastasis and predict poor prognosis in breast cancer patients. Am J Cancer Res. 10:2546–2554. 2020.PubMed/NCBI | |
Zhong L, Liao D, Zhang M, Zeng C, Li X, Zhang R, Ma H and Kang T: YTHDF2 suppresses cell proliferation and growth via destabilizing the EGFR mRNA in hepatocellular carcinoma. Cancer Lett. 442:252–261. 2019. View Article : Google Scholar | |
Chen YG, Chen R, Ahmad S, Verma R, Kasturi SP, Amaya L, Broughton JP, Kim J, Cadena C, Pulendran B, et al: N6-methyladenosine modification controls circular RNA immunity. Mol Cell. 76:96–109.e9. 2019. View Article : Google Scholar : PubMed/NCBI | |
Paris J, Morgan M, Campos J, Spencer GJ, Shmakova A, Ivanova I, Mapperley C, Lawson H, Wotherspoon DA, Sepulveda C, et al: Targeting the RNA m6A reader YTHDF2 selectively compromises cancer stem cells in acute myeloid leukemia. Cell Stem Cell. 25:137–148.e6. 2019. View Article : Google Scholar | |
Dixit D, Prager BC, Gimple RC, Poh HX, Wang Y, Wu Q, Qiu Z, Kidwell RL, Kim LJY, Xie Q, et al: The RNA m6A Reader YTHDF2 maintains oncogene expression and is a targetable dependency in glioblastoma stem cells. Cancer Discov. 11:480–499. 2021. View Article : Google Scholar : | |
Li J, Xie H, Ying Y, Chen H, Yan H, He L, Xu M, Xu X, Liang Z, Liu B, et al: YTHDF2 mediates the mRNA degradation of the tumor suppressors to induce AKT phosphorylation in N6-methyladenosine-dependent way in prostate cancer. Mol Cancer. 19:1522020. View Article : Google Scholar : PubMed/NCBI | |
Einstein JM, Perelis M, Chaim IA, Meena JK, Nussbacher JK, Tankka AT, Yee BA, Li H, Madrigal AA, Neill NJ, et al: Inhibition of YTHDF2 triggers proteotoxic cell death in MYC-driven breast cancer. Mol Cell. 81:3048–3064.e9. 2021. View Article : Google Scholar : PubMed/NCBI | |
Li A, Chen YS, Ping XL, Yang X, Xiao W, Yang Y, Sun HY, Zhu Q, Baidya P, Wang X, et al: Cytoplasmic m6A reader YTHDF3 promotes mRNA translation. Cell Res. 27:444–447. 2017. View Article : Google Scholar : PubMed/NCBI | |
Lin Y, Jin X, Nie Q, Chen M, Guo W, Chen L, Li Y, Chen X, Zhang W, Chen H, et al: YTHDF3 facilitates triple-negative breast cancer progression and metastasis by stabilizing ZEB1 mRNA in an m6A-dependent manner. Ann Transl Med. 10:832022. View Article : Google Scholar | |
Chang G, Shi L, Ye Y, Shi H, Zeng L, Tiwary S, Huse JT, Huo L, Ma L, Ma Y, et al: YTHDF3 induces the translation of m6A-enriched gene transcripts to promote breast cancer brain metastasis. Cancer Cell. 38:857–871.e7. 2020. View Article : Google Scholar | |
Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, Zhao BS, Mesquita A, Liu C, Yuan CL, et al: Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 20:285–295. 2018. View Article : Google Scholar : PubMed/NCBI | |
Müller S, Glaß M, Singh AK, Haase J, Bley N, Fuchs T, Lederer M, Dahl A, Huang H, Chen J, et al: IGF2BP1 promotes SRF-dependent transcription in cancer in a m6A- and miRNA-dependent manner. Nucleic Acids Res. 47:375–390. 2019. View Article : Google Scholar : | |
Qiao YS, Zhou JH, Jin BH, Wu YQ and Zhao B: LINC00483 is regulated by IGF2BP1 and participates in the progression of breast cancer. Eur Rev Med Pharmacol Sci. 25:1379–1386. 2021.PubMed/NCBI | |
Shi W, Tang Y, Lu J, Zhuang Y and Wang J: MIR210HG promotes breast cancer progression by IGF2BP1 mediated m6A modification. Cell Biosci. 12:382022. View Article : Google Scholar : PubMed/NCBI | |
Shi J, Zhang Q, Yin X, Ye J, Gao S, Chen C, Yang Y, Wu B, Fu Y, Zhang H, et al: Stabilization of IGF2BP1 by USP10 promotes breast cancer metastasis via CPT1A in an m6A-dependent manner. Int J Biol Sci. 19:449–464. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zeng F, Yao M, Wang Y, Zheng W, Liu S, Hou Z, Cheng X, Sun S, Li T, Zhao H, et al: Fatty acid β-oxidation promotes breast cancer stemness and metastasis via the miRNA-328-3p-CPT1A pathway. Cancer Gene Ther. 29:383–395. 2022. View Article : Google Scholar | |
Xiong Y, Liu Z, Li Z, Wang S, Shen N, Xin Y and Huang T: Long non-coding RNA nuclear paraspeckle assembly transcript 1 interacts with microRNA-107 to modulate breast cancer growth and metastasis by targeting carnitine palmitoyltransferase-1. Int J Oncol. 55:1125–1136. 2019.PubMed/NCBI | |
Wang Z, Tong D, Han C, Zhao Z, Wang X, Jiang T, Li Q, Liu S, Chen L, Chen Y, et al: Blockade of miR-3614 maturation by IGF2BP3 increases TRIM25 expression and promotes breast cancer cell proliferation. EBioMedicine. 41:357–369. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wang CQ, Tang CH, Wang Y, Huang BF, Hu GN, Wang Q and Shao JK: Upregulated WTAP expression appears to both promote breast cancer growth and inhibit lymph node metastasis. Sci Rep. 12:10232022. View Article : Google Scholar : PubMed/NCBI | |
Ou B, Liu Y, Yang X, Xu X, Yan Y and Zhang J: C5aR1-positive neutrophils promote breast cancer glycolysis through WTAP-dependent m6A methylation of ENO1. Cell Death Dis. 12:7372021. View Article : Google Scholar : PubMed/NCBI | |
Huang T, Cao L, Feng N, Xu B, Dong Y and Wang M: N6-methyladenosine (m6A)-mediated lncRNA DLGAP1-AS1enhances breast canceradriamycin resistance through miR-299-3p/WTAP feedback loop. Bioengineered. 12:10935–10944. 2021. View Article : Google Scholar : PubMed/NCBI | |
Fan Y, Li X, Sun H, Gao Z, Zhu Z and Yuan K: Role of WTAP in cancer: From mechanisms to the therapeutic potential. Biomolecules. 12:12242022. View Article : Google Scholar : PubMed/NCBI | |
Howley BV and Howe PH: TGF-beta signaling in cancer: Post-transcriptional regulation of EMT via hnRNP E1. Cytokine. 118:19–26. 2019. View Article : Google Scholar | |
Howley BV, Mohanty B, Dalton A, Grelet S, Karam J, Dincman T and Howe PH: The ubiquitin E3 ligase ARIH1 regulates hnRNP E1 protein stability, EMT and breast cancer progression. Oncogene. 41:1679–1690. 2022. View Article : Google Scholar : PubMed/NCBI | |
Loh TJ, Moon H, Cho S, Jang H, Liu YC, Tai H, Jung DW, Williams DR, Kim HR, Shin MG, et al: CD44 alternative splicing and hnRNP A1 expression are associated with the metastasis of breast cancer. Oncol Rep. 34:1231–1238. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wu Y, Zhao W, Liu Y, Tan X, Li X, Zou Q, Xiao Z, Xu H, Wang Y and Yang X: Function of HNRNPC in breast cancer cells by controlling the dsRNA-induced interferon response. EMBO J. 37:e990172018. View Article : Google Scholar : PubMed/NCBI | |
Duijf PHG, Nanayakkara D, Nones K, Srihari S, Kalimutho M and Khanna KK: Mechanisms of genomic instability in breast cancer. Trends Mol Med. 25:595–611. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hong J, Xu K and Lee JH: Biological roles of the RNA m6A modification and its implications in cancer. Exp Mol Med. 54:1822–1832. 2022. View Article : Google Scholar : PubMed/NCBI | |
Xiang Y, Laurent B, Hsu CH, Nachtergaele S, Lu Z, Sheng W, Xu C, Chen H, Ouyang J, Wang S, et al: RNA m6A methylation regulates the ultraviolet-induced DNA damage response. Nature. 543:573–576. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhang C, Chen L, Peng D, Jiang A, He Y, Zeng Y, Xie C, Zhou H, Luo X, Liu H, et al: METTL3 and N6-methyladenosine promote homologous recombination-mediated repair of DSBs by modulating DNA-RNA hybrid accumulation. Mol Cell. 79:425–442.e7. 2020. View Article : Google Scholar : PubMed/NCBI | |
D'Alessandro G, Whelan DR, Howard SM, Vitelli V, Renaudin X, Adamowicz M, Iannelli F, Jones-Weinert CW, Lee M, Matti V, et al: BRCA2 controls DNA:RNA hybrid level at DSBs by mediating RNase H2 recruitment. Nat Commun. 9:53762018. View Article : Google Scholar : PubMed/NCBI | |
Abakir A, Giles TC, Cristini A, Foster JM, Dai N, Starczak M, Rubio-Roldan A, Li M, Eleftheriou M, Crutchley J, et al: N6-methyladenosine regulates the stability of RNA: DNA hybrids in human cells. Nat Genet. 52:48–55. 2020. View Article : Google Scholar | |
Wei J, Yin Y, Zhou J, Chen H, Peng J, Yang J and Tang Y: METTL3 potentiates resistance to cisplatin through m6A modification of TFAP2C in seminoma. J Cell Mol Med. 24:11366–11380. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yang Z, Yang S, Cui YH, Wei J, Shah P, Park G, Cui X, He C and He YY: METTL14 facilitates global genome repair and suppresses skin tumorigenesis. Proc Natl Acad Sci USA. 118:e20259481182021. View Article : Google Scholar : PubMed/NCBI | |
Miranda-Gonçalves V, Lobo J, Guimarães-Teixeira C, Barros-Silva D, Guimarães R, Cantante M, Braga I, Maurício J, Oing C, Honecker F, et al: The component of the m6A writer complex VIRMA is implicated in aggressive tumor phenotype, DNA damage response and cisplatin resistance in germ cell tumors. J Exp Clin Cancer Res. 40:2682021. View Article : Google Scholar | |
Qu F, Tsegay PS and Liu Y: N6-methyladenosine, DNA repair, and genome stability. Front Mol Biosci. 8:6458232021. View Article : Google Scholar | |
Ji HL, Hong J, Zhang Z, de la Peña Avalos B, Proietti CJ, Deamicis AR, Guzmán GP, Lam HM, Garcia J, Roudier MP, et al: Regulation of telomere homeostasis and genomic stability in cancer by N6-adenosine methylation (m6A). Sci Adv. 7:eabg70732021. View Article : Google Scholar | |
Maciejowski J and de Lange T: Telomeres in cancer: Tumour suppression and genome instability. Nat Rev Mol Cell Biol. 18:175–186. 2017. View Article : Google Scholar : PubMed/NCBI | |
Batra RN, Lifshitz A, Vidakovic AT, Chin SF, Sati-Batra A, Sammut SJ, Provenzano E, Ali HR, Dariush A, Bruna A, et al: DNA methylation landscapes of 1538 breast cancers reveal a replication-linked clock, epigenomic instability and cis-regulation. Nat Commun. 12:54062021. View Article : Google Scholar : PubMed/NCBI | |
Lippert TH, Ruoff HJ and Volm M: Intrinsic and acquired drug resistance in malignant tumors. The main reason for therapeutic failure. Arzneimittelforschung. 58:261–264. 2008.PubMed/NCBI | |
Taketo K, Konno M, Asai A, Koseki J, Toratani M, Satoh T, Doki Y, Mori M, Ishii H and Ogawa K: The epitranscriptome m6A writer METTL3 promotes chemo- and radioresistance in pancreatic cancer cells. Int J Oncol. 52:621–629. 2018.PubMed/NCBI | |
Liu X, Gonzalez G, Dai X, Miao W, Yuan J, Huang M, Bade D, Li L, Sun Y and Wang Y: Adenylate kinase 4 modulates the resistance of breast cancer cells to tamoxifen through an m6A-based epitranscriptomic mechanism. Mol Ther. 28:2593–2604. 2020. View Article : Google Scholar : PubMed/NCBI | |
Petri BJ, Piell KM, South Whitt GC, Wilt AE and Klinge CM, Lehman NL, Clem BF, Nystoriak MA, Wysoczynski M and Klinge CM: HNRNPA2B1 regulates tamoxifen- and fulvestrant-sensitivity and hallmarks of endocrine resistance in breast cancer cells. Cancer Lett. 518:152–168. 2021. View Article : Google Scholar : PubMed/NCBI | |
Liu X, Yuan J, Zhang X, Li L, Dai X, Chen Q and Wang Y: ATF3 modulates the resistance of breast cancer cells to tamoxifen through an N6-methyladenosine-based epitranscriptomic mechanism. Chem Res Toxicol. 34:1814–1821. 2021. View Article : Google Scholar : PubMed/NCBI | |
Pan X, Hong X, Li S, Meng P and Xiao F: METTL3 promotes adriamycin resistance in MCF-7 breast cancer cells by accelerating pri-microRNA-221-3p maturation in a m6A-dependent manner. Exp Mol Med. 53:91–102. 2021. View Article : Google Scholar : PubMed/NCBI | |
Li E, Xia M, Du Y, Long F, Pan F, He L, Hu Z and Guo Z: METTL3 promotes homologous recombination repair and modulates chemotherapeutic response by regulating the EGF/Rad51 axis. bioRxiv. 2021. | |
Li S, Jiang F, Chen F, Deng Y and Pan X: Effect of m6A methyltransferase METTL3-mediated MALAT1/E2F1/AGR2 axis on adriamycin resistance in breast cancer. J Biochem Mol Toxicol. 36:e229222022. View Article : Google Scholar | |
Wu Y, Wang Z, Han L, Guo Z, Yan B, Guo L, Zhao H, Wei M, Hou N, Ye J, et al: PRMT5 regulates RNA m6A demethylation for doxorubicin sensitivity in breast cancer. Mol Ther. 30:2603–2617. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Cheng Z, Xu J, Lai M, Liu L, Zuo M and Dang L: Fat mass and obesity-associated protein (FTO) mediates signal transducer and activator of transcription 3 (STAT3)-drived resistance of breast cancer to doxorubicin. Bioengineered. 21:1874–1889. 2021. View Article : Google Scholar | |
Liu X, Li P, Huang Y, Li H, Liu X, Du Y, Lin X, Chen D, Liu H and Zhou Y: M6A demethylase ALKBH5 regulates FOXO1 mRNA stability and chemoresistance in triple-negative breast cancer. Redox Biol. 69:1029932024. View Article : Google Scholar | |
Ou B, Liu Y, Gao Z, Xu J, Yan Y, Li Y and Zhang J: Senescent neutrophils-derived exosomal piRNA-17560 promotes chemoresistance and EMT of breast cancer via FTO-mediated m6A demethylation. Cell Death Dis. 13:9052022. View Article : Google Scholar : PubMed/NCBI | |
Zhuang H, Yu B, Tao D, Xu X, Xu Y, Wang J, Jiao Y and Wang L: The role of m6A methylation in therapy resistance in cancer. Mol Cancer. 22:912023. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Zhang L, Sun XL, Lu YC, Chen S, Pei DS and Zhang LS: NRP1 contributes to stemness and potentiates radioresistance via WTAP-mediated m6A methylation of Bcl-2 mRNA in breast cancer. Apoptosis. 28:233–246. 2023. View Article : Google Scholar |