Skip to main content

Functions of methyltransferase-like 3 in breast cancer: pathogenesis, drug resistance, and therapeutic target

Abstract

Breast cancer (BC) is a highly prevalent malignancy worldwide, with complex pathogenesis and treatment challenges. Research reveals that methyltransferase-like 3 (METTL3) is widely involved in the pathogenesis of several tumors through methylation of its target RNAs, and its role and mechanisms in BC are also extensively studied. In this review, we aim to provide a comprehensive interpretation of available studies and elucidate the relationship between METTL3 and BC. This review suggests that high levels of METTL3 are associated with the pathogenesis, poor prognosis, and drug resistance of BC, suggesting METTL3 as a potential diagnostic or prognostic biomarker and therapeutic target. Collectively, this review provides a comprehensive understanding of how METTL3 functions through RNA methylation, which provides a valuable reference for future fundamental studies and clinical applications.

Background

Breast cancer (BC) is a prevalent malignancy worldwide [1]. Moreover, resistance to available therapies is a major obstacle to the effective treatment of BC [2]. BC is considered a heterogeneous disease, and its pathogenesis, which is affected by genes and external circumstances, is complex and has not yet been completely clarified [1]. Therefore, insights into the mechanisms of BC occurrence and development may help improve its diagnosis, prognosis, and treatment.

Tumor formation and progression are complex processes and involve multiple mechanisms that usually require precise regulation of gene expression across diverse cells and organs [3]. N6-methyladenosine (m6A) regulates gene expression in a post-transcriptional manner, which includes the components of “writers”, “erasers”, and “readers” [4]. Writers specifically act as the catalytic subunit to add a methyl group to the N-6position of adenosine, including the subunit of methyltransferase-like 3/14 (METTL3/14) and Wilms’ tumor 1-associating protein (WTAP) [4]. Meanwhile, this modification is removed by erasers and is further recognized by readers such as YTHDF (YTH N6-methyladenosine RNA binding protein F), YTHDC (YTH domain-containing proteins), and IGF2BP (Insulin-like growth factor 2 mRNA binding protein) [4]. Recently, RNA methylation byMETTL3 has emerged as a crucial modulator of mammalian gene expression [5]. METTL3, in contrast to the structural subunit of METTL14 and WTAP in RNA m6A modification, has the methyltransferase function to regulate RNA stability and life-cycle events, such as splicing, translation, and degradation [6]. METTL3 also regulates the biological processes of tumor formation and progression, either as an oncogene, or by regulating other oncogenes [7]. In addition, METTL3-mediated RNA m6A modification induces drug resistance in cancer via epigenetic regulation of candidate drug-resistance genes [7]. Studies have especially emphasized the risk role of METTL3 in the occurrence, progression, metastasis, poor prognosis, and drug resistance of BC [8,9,10]. However, there is a lack of reviews that comprehensively summarize and interpret these studies.

Therefore, in this review, we comprehensively analyze all original publications on METTL3 and its roles in BC, aiming to provide a better understanding of the current status of research on METTL3 in BC.

Functions and mechanisms of METTL3 in BC

METTL3 contributes to the onset and progression of BC

BC is a complex disease at the cellular and molecular levels [11]. Clinical data demonstrate that METTL3 is upregulated in BC tissues, especially in phase T3-T4, or in patients with BC that are diagnosed with tumor metastasis in the lymphatic system [12]. Furthermore, findings also demonstrate the overexpression of METTL3 in BCSCs, and high expression levels of METTL3 are usually associated with a worse prognosis in patients with BC [12]. Therefore, METTL3 might be a risk factor for BC.

METTL3 functions by targeting oncogenes

METTL3-mediated m6A modification of RNAs includes coding (mRNA) and non-coding (i.e., microRNA [miRNA], long non-coding RNA [lncRNA], circular RNA [circRNA]) RNAs.

METTL3 and mRNA modification

BC stem cells (BCSCs) possess self-renewal and differentiation abilities to produce more BC cells, which function primarily during BC initiation, as well as play a fundamental role in BC progression, drug resistance, and recurrence [13]. Xie et al. [12] demonstrated that METTL3 modifies SOX2 mRNA via m6A modification and activates BCSC stemness and malignant progression. Other studies demonstrate that by targeting EZH2 mRNA, METTL3 promotes epithelial-mesenchymal transition (EMT) and metastasis of BCSCs, thereby aggravating the malignant phenotype of BC [14, 15]. In addition, METTL3 also increases the methylation levels, accelerates proliferation rates, promotes tumor growth, and reduces apoptosis of BC cells via m6A modification on Bcl-2 mRNA [14, 15]. Xu et al. [16] reported that METTL3, together with YTHDF2, reduces large tumor suppressor kinase 1(LTSK1) mRNA stability, thereby promoting the proliferation and glycolytic metabolism in BC through the YAP/TAZ axis and Hippo pathways. Furthermore, alternative splicing (AS) enables differential inclusion of exons from a given transcript, thereby contributing to the transcriptome and proteome diversity [17]. Aberrant AS patterns play key roles in BC development. METTL3 functions in modulating BC-associated AS processes [18]. Specifically, both m6A deposition in splice site boundaries and in splicing and transcription factor transcripts, such as MYC, direct AS switches of specific BC-associated transcripts, which are associated with a poor overall survival rate in patients with BC, suggesting the use of these AS events as a novel potential prognostic biomarker [18].

Triple-negative BC (TNBC) is the most dominant and malignant pathological type of BC [19]. m6A regulators exhibit remarkable dysregulation in TNBC tissues, whereas METTL3 shows significant downregulation [20, 21]. Moreover, the level of METTL3 is found to be tightly linked to the progression and poor survival in patients with TNBC, whereas its gene mutation and amplification are not associated with these outcomes [21]. FAM83D is markedly elevated in TNBC tissues and cells, and high-level of FAM83D is related to poor prognosis in patients with TNBC [22, 23]. Yu et al. [22] reported that METTL3 mediates m6A modification on FAM838D and upregulates FAM83D expression to activate the Wnt/β-catenin pathway, which in turn promotes TNBC malignant behaviors [22]. These findings suggest that METTL3 aggravates TNBC progression by targeting the FAM83D/Wnt/β-catenin pathway (Fig. 1).

Fig. 1
figure 1

METTL3-mediated RNA m6A modification induces breast cancer. High levels of METTL3 are associated with the tumorigenesis, progression, invasion, and metastasis of breast cancer, which functions by mediating m6A modification on different RNAs, inducing the abnormal process of proliferation, migration, apoptosis, and cell cycle. Methyltransferase-like 3 (METTL3). N6-methyladenosine (m6A). Acetyl-CoA acetyltransferase 1 (ACAT1). Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1). High mobility group AT-hook 2 (HMGA2). High zinc finger protein 217 (ZNF217). Large tumor suppressor kinase 1 (LATS1). Transcriptional coactivator with PDZ-binding motif (TAZ). Host gene of circular RNA METTL3 (circMETTL3). Cyclin-dependent kinase 1 (CDK1). Cleaved form of METTL3 (METTL3a). Hepatitis B X-interacting protein (HBXIP). Epidermal growth factor receptor (EGFR). Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1). P-element-induced wimpy testis (PIWI)-interacting RNAs (piRNAs). YTH N6-methyladenosine RNA binding protein F1 (YTHDF1)

Collectively, these findings suggest that METTL3 functions as an oncogenic driver to modify mRNAs and triggers BC onset and progression.

METTL3 and non-coding RNAs

METTL3 also exerts its oncogenic function by modulating the expression of noncoding RNAs. Ren et al. [23] found that LINC00667 is an m6A-modified lncRNA, which is up-regulated upon KIAA1429 overexpression. The high expression of LINC00667 is correlated with the poor prognosis of patients with BC. LINC00667 promotes the proliferation and migration of BC cells. KIAA1429 targets the m6A modification site on LINC00667 and enhances its mRNA stability [24]. Moreover, LINC00667 positively regulates KIAA1429 via sponging miR-556-5p, forming a KIAA1429/m6A/LINC00667/miR-556-5p feedback loop [24]. Collectively, the central findings of this study suggest that KIAA1429-induced LINC00667 exerts functions as an oncogene in BC progression through an m6A-dependent feedback loop. miR-26b exhibits consistently low levels, while METTL3, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), and high mobility group AT-hook 2 (HMGA2) demonstrate elevated levels in BC [25]. METTL3 increases MALAT1 expression by modulating its m6A modification, which subsequently promotes HMGA2 expression through miR-26b sponging, ultimately leading to EMT, migration, and invasion of BC cells [25]. Notably, METTL3 drives BC tumorigenesis via the MALAT1/miR-26b/HMGA2 axis.

High zinc finger protein 217 (ZNF217) expression and low miR-135 expression levels are found in BC tissues and cells [26]. Silencing ZNF217 or elevating miR-135 inhibits BC cell malignant behaviors [26]. ZNF217 upregulates METTL3 and that in turn targets NANOG to promote BC progression [26]. Therefore, the miR-135/ZNF217/METTL3/NANOG axis promotes BC progression, emphasizing METTL3 or NANOG as a potential therapeutic target for BC treatment. Rong et al. [27] showed that METTL3 facilitates LINC00958 expression by enhancing its stability in BC tissues and cells, thus revealing a novel METTL3-involving mechanism for regulating BC tumorigenesis. LINC00958 interacts with miR-378a-3p in a competitive endogenous RNA (ceRNA) manner to promote YY1 expression [27]. Therefore, BC tumor progression is further promoted and aggravated via the METTL3-LINC00958/miR-378a-3p-YY1 axis. Fan et al. [28] discovered that METTL3 downregulates LINC00675 expression, which is indicative of a higher tumor grade, worse lymphovascular invasion, and shorter survival in patients with BC. Mechanistically, LINC00675 acts as a ceRNA by interacting with miR-513b-5p and repressing its expression [28].

Furthermore, the level of LncRNA-WFDC21P is abnormally high in TNBC, which promotes TNBC cell proliferation and metastasis, as well as is strongly linked to a poor survival rate of patients. Wei et al. [29] reported that METTL3-mediated m6A modification on WFDC21P upregulates WFDC21P expression in TNBC cells, which in turn interacts with miR-628-5p and further inhibits Smad3-related gene expression to promote cell proliferation and metastasis [29]. This study demonstrates the vital roles of METTL3-mediated m6A modification on WFDC21P and its regulation in the proliferation and metastasis of TNBC cells via the METTL3/WFDC21P/miR-628/SMAD3 axis (Fig. 1).

METTL3 functions as the target oncogene METTL3 functions in BC by modifying itself

In addition to regulating target gene expression, METTL3 itself is regulated by other factors in BC. Li et al. [30] revealed that METTL3, the host gene of circular RNA METTL3 (circMETTL3), regulates circMETTL3 expression in an m6A-dependent manner. However, circMETTL3 does not affect METTL3 expression. Mechanistically, circMETTL3 interacts with miR-31-5p in a ceRNA manner, thereby upregulating the expression of its target, cyclin-dependent kinase 1 (CDK1) [30]. Therefore, circMETTL3 facilitates BC cell proliferation, migration, and invasion via the circMETTL3/miR-31-5p/CDK1 axis. In addition to investigating the novel relationship between the circRNA and its corresponding host gene, another study indicated that the cleaved form of METTL3 (termed METTL3a), which is mediated by proteasomes in an mTOR-dependent manner, is essential for regulating the assembly of the METTL3-METTL14-WTAP complex [31]. Subsequently, the METTL3-METTL14-WTAP complex functions to promote BC progression by depositing m6A modification on RNAs, which primarily depends on its methyltransferase activity that is supported by the catalytic role of METTL3, the structural role of METTL14, as well as the adaptor role of WTAP.

In contrast, a previous study revealed a striking reduction of METTL3 levels in TNBC samples, including tissues and cell lines [32]. Functionally, METTL3 upregulates its target, circMETTL3, which consequently interacts with miR-34c-3p in a ceRNA manner [32]. Furthermore, miR-34c-3p represses METTL3 expression [32]. The negative feedback-loop regulatory mechanism of METTL3-circMETTL3/ miR-34c-3p-METTL3 promotes TNBC tumor cell proliferation, invasion, metastasis, and growth [32]. Meanwhile, a low level of METTL3 in TNBC is strongly linked to short, distant metastasis-free survival [33]. METTL3 augments migration, invasion, and adhesion of TNBC cell lines by upregulating the collagen type III alpha 1 chain, ultimately promoting the metastasis of TNBC cells [33]. In addition, Zhang et al. [34] reported an acetylation-related mechanism for TNBC progression, demonstrating that nuclear receptor subfamily 2 group F member 6 transcriptionally activates acetyl-CoA acetyltransferase 1 (ACAT1), which promotes TNBC cell migration and invasion via ACAT1-mediated METTL3 acetylation. Notably, although the level of METTL3 is downregulated, the modified-METTL3 (circMETTL3, METTL3 acetylation) is upregulated, which together affect the malignant processes in BC (Fig. 1).

Collectively, these findings suggest that METTL3 contributes to BC by regulating itself, suggesting its oncogenic role. Moreover, although findings suggest a low level of METTL3 in TNBC, it functions through its modified types with the oncogenic role. Therefore, METTL3 is a risk gene for BC.

METTL3 functions through a regulatory network

Consistently, Cai et al. [35] reported that hepatitis B X-interacting protein (HBXIP) increases METTL3 expression in BC by suppressing miRNA let-7 g. Remarkably, METTL3-mediated m6A modification in turn enhances HBXIP expression [35]. Overexpression of HBXIP not only facilitates BC cell proliferation but also impairs apoptosis [35]. In summary, a positive feedback-loop regulatory mechanism, HBXIP-let-7 g/METTL3-HBXIP, is established by HBXIP to expedite BC cell proliferation. Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1) interacts with METTL3 and thereby blocks its ubiquitin-dependent proteasomal and lysosomal degradation [36]. In clinical settings, METTL3 expression is significantly increased with tumor progression and is positively correlated with PIN1 expression in BC tissues [36]. PIN1 stabilizes METTL3, which in turn increases the m6A modification and translation of the transcriptional coactivator with PDZ-binding motif (TAZ) and the epidermal growth factor receptor (EGFR) mRNA [36]. Inhibition of MEK1/2 kinases and PIN1 destabilizes METTL3, which blocks BC cell proliferation and induces cell cycle arrest at the G0/G1 phases [36]. Moreover, silencing METTL3 reduces PIN1 overexpression-induced colony formation in MCF7 cells and enhances tumor growth in 4T1 cells in mouse models [36]. Therefore, PIN1 upregulates METTL3, and the PIN1/METTL3 axis may be an alternative therapeutic target for BC. P-element-induced wimpy testis (PIWI)-interacting RNAs (piRNAs) are novel non-coding RNAs whose abnormal expression is closely associated with multiple cancers [37]. Huang et al. [38] reported that the level of PiRNA-31,106 is high in BC tissues and cell lines (MDA-MB-231 and MCF-7), which promotes the m6A methylation levels and facilitates METTL3 expression that further promotes BC progression by upregulating the expressions of MDM2, CDK4, and cyclinD1 [38]. Therefore, PiRNA-31,106 upregulates METTL3 to promote BC progression, suggesting its potential as a therapeutic target in BC (Fig. 1).

To sum up, these findings highlight that METTL3 regulates BC progression through various mechanisms, including interacting with other molecules and influencing multiple pathways. Therefore, while METTL3 does have a direct impact on oncogenic processes, its role is also mediated through complex regulatory networks.

METTL3 functions by interacting with A-to-I RNA editing

Furthermore, A-to-I RNA editing is another prevalent type of RNA modification that controls gene expression in mammals and regulates tumorigenesis and tumor progression [39]. Specifically, ADAR1 is an A-to-I RNA-editing enzyme, both ADAR1 and METTL3 are upregulated in BC samples [40]. Li et al. [40] showed that ADAR1 interacts with METTL3 in a YTHDF1-dependent manner and further increases METTL3 expression. Mechanically, ADAR1 changes the binding site of METTL3 mRNA on miR532-5p and is recognized by YTHDF1. Subsequently, METTL3 expression is increased, which further targets ARHGAP5 to promote the proliferation, migration, and invasion of BC cells [40]. Loss of ADAR1 or METTL3 significantly inhibits BC growth in vivo [40]. Notably, the ADAR1-METTL3 axis is a novel pathway that connects the RNA regulation mechanism of A-to-I editing and METTL3-mediated m6A modification during BC progression. Therefore, the combined targeting of ADAR1 or METTL3 might be more effective for BC treatment (Fig. 1).

Collectively, these findings suggest that METTL3 is primarily associated with BC initiation and progression, which functions by regulating the oncogene target or functions as the target oncogene. Furthermore, these findings suggest that METTL3 has great clinical potential as a diagnosis biomarker.

METTL3 induces drug resistance and poor prognosis of BC

Chemotherapy is an important strategy for BC treatment [41]. Adriamycin (ADR), Tamoxifen, and Paclitaxel are currently considered the primary therapeutic drugs for patients with BC [42]. However, clinical data suggest that the majority of patients with BC experience resistance to chemotherapy [41]. Specifically, resistance to ADR and Tamoxifen is considered a troublesome challenge for curing patients with BC [43]. Currently, studies have also suggested the implication of METTL3-mediated RNA m6A modification in the development of BC chemoresistance. The global levels of mRNA m6A methylation and METTL3 are upregulated in ADR-resistant BC cell lines while silencing METTL3 partially overcomes this resistance [44]. METTL3 cooperates with IGF2BP3 to modulate the m6A modification on HYOU1 mRNA and increases HYOU1 stability, which subsequently increases ADR resistance in BC cells [44]. Therefore, the METTL3/IGF2BP3-HYOU1 axis modulates ADR sensitivity in BC cells, targeting this axis might be a strategy to improve ADR efficacy in BC treatment.

Zhou et al. [43] demonstrated that METTL3-mediated m6A modification on long noncoding RNA (lncRNA) KCNQ1OT1 promotes ADR resistance in BC by regulating the miR-103a-3p/multidrug resistance protein 1 (MDR1) axis. LncRNA KCNQ1OT1 is highly expressed in ADR-resistant BC cells, and its expression is modulated by MELLT3 via m6A modification. A further mechanistic study found that miR-103a-3p acts as a sponge for lncRNA KCNQ1OT1 and MDR1 [43]. Altogether, these findings indicate that the METTL3-lncRNAKCNQ1OT1/ miR-103a-3p-MDR1 axis is responsible for ADR resistance in BC, which suggests a novel approach to prevent ADR resistance in patients with BC. Consistently, high levels of METTL3 enhance the ADR resistance for BC cells by upregulating miR-221-3p through positive regulation of homeodomain-interacting protein kinase 2 (HIPK2) and its direct target Che-1 [45]. More detailed, METTL3 functions to promote miR-221-3p expression by decreasing m6A mRNA methylation on pri-miR-221-3p [45]. Subsequently, miR-221-3p further enhances the expression of MDR1 and BCRP, which promotes a higher production of ADR-resistant cells while inhibiting apoptosis in such cells [45]. Together, after the cooperation of these signaling molecules, the METTL3-HIPK2/Che-1-miR-221-3p-MDR1/BCRP axis triggers ADR resistance in BC cells. Li et al. [46] found that METTL3 is highly expressed in ADR-resistant BC cells and upregulates MALAT1 through m6A modification, which, in turn, recruits E2F transcription factor 1(E2F1) to activate anterior gradient 2 (AGR2) transcription and increases ADR resistance in BC cells. Collectively, METTL3 promotes ADR resistance in BC by targeting the MALAT1/E2F1/AGR2 axis. Moreover, Li et al. [47] reported that METTL3 promotes the efficiency of homologous recombination repair and inhibits ADR-induced DNA damage. METTL3 functions by enhancing epidermal growth factor (EGF) synthesis in a YTHDC1-dependent manner, and EGF subsequently inhibits RAD51 expression [47]. These findings reveal the mechanism of METTL3 in regulating ADR resistance in BC by targeting the EGF/RAD51 axis. Furthermore, Liu et al. [48] revealed that the expression of METTL3 and adenylate kinase 4 (AK4) is remarkably elevated in Tamoxifen-resistant BC cells compared with that in their parental counterparts. Further investigation found that METTL3 methylates AK4 mRNA on the 5′-untranslated region, leading to Tamoxifen resistance in patients with BC [46].

Consistently, Jing et al. [49] reported that METTL3 regulates Docetaxel resistance of TNBC cells by regulating a feedback loop. They found that METTL3 uses LINC00662 as its target, which directly interacts with miR-186-5p, and METTL3 expression is in turn regulated by miR-186-5p [49]. LINC00662 and miR-186-5p regulate the viability of Docetaxel-resistant BC cells, and miR-186-5p subsequently regulates METTL3 expression [49]. Overall, these findings suggest that suppression of METTL3 promotes Docetaxel resistance in BC through the METTL3-LINC00662/miR‑186‑5p-METTL3 signaling (Fig. 2).

Fig. 2
figure 2

METTL3 contributes to the drug resistance of breast cancer. High levels of METTL3 are associated with drug resistance and poor prognosis of breast cancer by mediating m6A modification on different RNAs. Methyltransferase-like 3 (METTL3). N6-methyladenosine (m6A). Multidrug resistance protein 1 (MDR1). Homeodomain-interacting protein kinase 2 (HIPK2). Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1). E2F transcription factor 1(E2F1). Anterior gradient 2 (AGR2). Epidermal growth factor (EGF). YTH domain containing 1 (YTHDC1). Adenylate kinase 4 (AK4)

However, METTL3 knockdown decreases the drug sensitivity of hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR + HER2) cells by targeting CDKN1A, activating the AKT pathway, and promoting EMT [50]. Moreover, METTL3 knockdown also leads to the inhibition of BAX and caspase-3/-9/-8 [50]. Therefore, METTL3 might play a tumor-suppressor role and it could be a potential biomarker for predicting the prognosis of patients with HR + HER2-BC.

Collectively, these data suggest that METTL3 contributes to various drug resistance of BC, which usually indicates a poor prognosis and recurrence of BC. Therefore, we suppose that METTL3 is a promising prognostic biomarker for BC.

Targeting METTL3 for BC therapy

Considering that a high level of METTL3 promotes BC occurrence, progression, and drug resistance. Therefore, inhibition of METTL3 is considered a potential approach for promoting chemotherapeutic effectiveness against BC. STM2457, the small-molecule inhibitor of METTL3, significantly reduces the proliferation, viability, and migration of TNBC cells, including both BRCA1/2 wild-type and BRCA1-mutated cell lines [51]. Therefore, STM2457 displays anti-tumor activity in TNBC and enhances chemotherapy response while sensitizing BRCA1/2 wildtype patients to Olaparib.

Epigenetic drugs are a novel therapeutic method for BC [52]. Cromolyn is a mast cell stabilizer emerging as an anticancer drug [53]. The study suggests that chitosan nanoparticle encapsulated Cromolyn shows a prominent anticancer effect in MCF-7 cells by reducing the cell viability percent and enhancing DNA damage [54]. Mechanistically, it functions by inhibiting DNA methyltransferase 1 (DNMT1) and METTL3, as well as reversing the hypermethylation pattern of the tumor suppressor genes RASSF1A and p16 [54]. In addition, it diminishes ERK1/2 phosphorylation, which affects DNMT1 expression [54]. Moreover, in vivo, it lessens the tumor volume and halts DNMT1 and METTL3 expression in BC mice [54]. Therefore, chitosan nanoparticle encapsulated Cromolyn acts as an epigenetic drug by inhibiting ERK1/2 phosphorylation/DNMT1/DNA methylation and impacting the RNA methylation via inhibiting METTL3 expression. Additionally, Cheng et al. [55] reported that Metformin inhibits BC progression by reducing METTL3 expression mediated by miR-483-3p. In detail, the inhibited expression of METTL3 reduces BC cell proliferation by modulating p21 expression in an m6A-dependent manner [55]. These findings suggest that Metformin functions to relieve BC by inhibiting METTL3 via p21, suggesting METTL3 is a potential therapeutic target for BC treatment. Furthermore, Wan et al. [56] reported that the inhibition of METTL3 enhances tumor immune surveillance. Mechanistically, the downregulated expression of METTL3 cooperates with IGF2BP3 to reduce the m6A modification and the expression of programmed cell death 1 ligand 1 (PDL-1), which subsequently heightens antitumor immunity through PDL-1- mediated T-cell activation, exhaustion, and infiltration [56]. These findings suggest that inhibition of METTL3 is an effective strategy for BC immunotherapy.

Conclusions

The high incidence and mortality of BC have been the major health problems worldwide for the last few decades [57]. Extensive studies have identified the mechanisms underlying BC pathogenesis and therapeutic failure, which are primarily a result of alterations in oncogenes and tumor suppressor genes, leading to protein dysregulation and carcinogen activation [58]. Therefore, studies on epigenetic modifications underlying the occurrence, progression, and metastasis of BC have attracted great attention. METTL3 has the catalytic ability to regulate the biological characteristics of RNA (i.e., stability and translation) [6]. METTL3 also accelerates the proliferation, migration, and invasion of BC cells via posttranscriptional modifications. Herein, we summarize all original publications on this topic and classify the findings into different categories to further analyze and interpret the roles, mechanisms, and relationships of METTL3 during different events in BC, including occurrence and progression, treatment failure, as well as targeting METTL3 for therapy.

This review suggests that high levels of METTL3 act as an oncogene by targeting mRNAs and noncoding RNAs during the occurrence and progression of BC, and increase chemoresistance. In addition to regulating target gene expression, METTL3 functions as a target gene for other risk factors and regulators in BC. The underlying mechanism of METTL3 functions includes inducing the abnormal process of proliferation, migration, apoptosis, and cell cycle by mediating m6A modification on different RNAs. Furthermore, data also hint to us that targeting METTL3 is a promising strategy for BC treatment. In summary, these findings suggest that METTL3 with great clinical potential as a diagnostic and prognostic biomarker, as well as a treatment target.

Although the analysis of this review suggests METTL3 is a risk gene for BC, contrast findings are also found in patients with HR + HER2-BC, which suggest a tumor suppressor role for METTL3. However, due to the only one available publications on the role of METTL3 in patients with HR + HER2-BC, the conclusion for METTL3’s role in HR + HER2-BC needs further investigation. Besides, current available methods for downregulating METTL3 include the small-molecule inhibitor STM2457, epigenetic drugs Cromolyn and Metformin, as well as PDL-1. However, to our knowledge, preclinical studies for treating patients with BC by targeting METTL3 are not yet available. Furthermore, combination therapies involving METTL3 inhibitors and other methods have not been reported as well. To sum up, these are the main limitations of the existing evidence supporting METTL3’s role in BC. Therefore, we suppose that future studies and clinical validation are strongly required to bridge such gaps to improve the understanding of METTL3’ role in BC.

Collectively, this review provides a systematic interpretation of the risk role of METTL3 in BC, which provides a convenient reference for exploring its clinical potential in the future.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

BC:

Breast cancer

METTL3:

Methyltransferase-like 3

m6A:

N6-methyladenosine

BCSCs:

BC stem cells

EMT:

Epithelial-mesenchymal transition

YTHDF:

YTH N6-methyladenosine RNA binding protein F

LTSK1:

Large tumor suppressor kinase 1

AS:

Alternative splicing

MALAT1:

Metastasis-associated lung adenocarcinoma transcript 1

HMGA2:

High mobility group AT-hook 2

ZNF217:

High zinc finger protein 217

ceRNA:

Competitive endogenous RNA

circMETTL3:

Host gene of circular RNA METTL3

CDK1:

Cyclin-dependent kinase 1

METTL3a:

Cleaved form of METTL3

HBXIP:

Hepatitis B X-interacting protein

PIN1:

Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1

TAZ:

Transcriptional coactivator with PDZ-binding motif

EGFR:

Epidermal growth factor receptor

piRNAs:

P-element-induced wimpy testis (PIWI)-interacting RNAs

TNBC:

Triple-negative BC

ACAT1:

Acetyl-CoA acetyltransferase 1

ADR:

Adriamycin

IGF2BP3:

Insulin-like growth factor 2 mRNA binding protein 3

lncRNA:

Long noncoding RNA

MDR1:

Multidrug resistance protein 1

HIPK2:

Homeodomain-interacting protein kinase 2

E2F1:

E2F transcription factor 1

AGR2:

Anterior gradient 2

EGF:

Epidermal growth factor

AK4:

Adenylate kinase 4

DNMT1:

DNA methyltransferase 1

PDL-1:

Programmed cell death 1 ligand 1

References

  1. Britt KL, Cuzick J, Phillips KA. Key steps for effective breast cancer prevention. Nat Rev Cancer. 2020;20(8):417–36.

    Article  CAS  PubMed  Google Scholar 

  2. Barzaman K, Karami J, Zarei Z, Hosseinzadeh A, Kazemi MH, Moradi-Kalbolandi S et al. Breast cancer: Biology, biomarkers, and treatments. Int Immunopharmacol. 2020;84.

  3. Fedele M, Sgarra R, Battista S, Cerchia L, Manfioletti G. The epithelial-mesenchymal transition at the crossroads between metabolism and tumor progression. Int J Mol Sci. 2022;23(2).

  4. 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.

    Article  CAS  PubMed  Google Scholar 

  5. You SY, Su XJ, Ying JJ, Li SP, Qu Y, Mu DZ. Research Progress on the role of RNA m6A modification in glial cells in the regulation of neurological diseases. Biomolecules. 2022;12(8).

  6. Su X, Qu Y, Mu D. The Regulatory Network of METTL3 in the nervous system: diagnostic biomarkers and therapeutic targets. Biomolecules. 2023;13(4):664.

    Article  CAS  PubMed  Google Scholar 

  7. Wang TY, Kong S, Tao M, Ju SQ. The potential role of RNA N6-methyladenosine in Cancer progression. Mol Cancer. 2020;19(1).

  8. Zheng F, Du F, Zhao J, Wang X, Si Y, Jin P, et al. The emerging role of RNA N6-methyladenosine methylation in breast cancer. Biomark Res. 2021;9(1):39.

    Article  PubMed  Google Scholar 

  9. Deng X, Su R, Feng X, Wei M, Chen J. Role of N6-methyladenosine modification in cancer. Curr Opin Genet Dev. 2018;48:1–7.

    Article  CAS  PubMed  Google Scholar 

  10. Xu P, Ge R. Roles and drug development of METTL3 (methyltransferase-like 3) in anti-tumor therapy. Eur J Med Chem. 2022;230:114118.

    Article  CAS  PubMed  Google Scholar 

  11. Nolan E, Lindeman GJ, Visvader JE. Deciphering breast cancer: from biology to the clinic. Cell. 2023;186(8):1708–28.

    Article  CAS  PubMed  Google Scholar 

  12. Xie J, Ba J, Zhang M, Wan Y, Jin Z, Yao Y. The m6A methyltransferase METTL3 promotes the stemness and malignant progression of breast cancer by mediating m6A modification on SOX2. J Buon. 2021;26(2):444–9.

    PubMed  Google Scholar 

  13. Zhang L, Chen W, Liu S, Chen C. Targeting breast Cancer stem cells. Int J Biol Sci. 2023;19(2):552–70.

    Article  CAS  PubMed  Google Scholar 

  14. Wang H, Xu B, Shi J. N6-methyladenosine METTL3 promotes the breast cancer progression via targeting Bcl-2. Gene. 2020;722:144076.

    Article  CAS  PubMed  Google Scholar 

  15. Hu SJ, Song Y, Zhou Y, Jiao Y, Li GP. METTL3 Accelerates Breast Cancer Progression via Regulating EZH2 m(6)A Modification. J Healthc Eng. 2022;2022.

  16. Xu Y, Song M, Hong Z, Chen W, Zhang Q, Zhou J, et al. The N6-methyladenosine METTL3 regulates tumorigenesis and glycolysis by mediating m6A methylation of the tumor suppressor LATS1 in breast cancer. J Exp Clin Cancer Res. 2023;42(1):10.

    Article  CAS  PubMed  Google Scholar 

  17. Marasco LE, Kornblihtt AR. The physiology of alternative splicing. Nat Rev Mol Cell Biol. 2023;24(4):242–54.

    Article  CAS  PubMed  Google Scholar 

  18. Achour C, Bhattarai DP, Groza P, Román ÁC, Aguilo F. METTL3 regulates breast cancer-associated alternative splicing switches. Oncogene. 2023;42(12):911–25.

    Article  CAS  PubMed  Google Scholar 

  19. Yin L, Duan JJ, Bian XW, Yu SC. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020;22(1):61.

    Article  PubMed  Google Scholar 

  20. Wang S, Zou X, Chen Y, Cho WC, Zhou X. Effect of N6-Methyladenosine regulators on Progression and Prognosis of Triple-negative breast Cancer. Front Genet. 2020;11:580036.

    Article  CAS  PubMed  Google Scholar 

  21. Wu L, Wu D, Ning J, Liu W, Zhang D. Changes of N6-methyladenosine modulators promote breast cancer progression. BMC Cancer. 2019;19(1):326.

    Article  PubMed  Google Scholar 

  22. Yu X, Li Y, Kong F, Xu Q. METTL3 regulates FAM83D m(6)a modification to accelerate tumorigenesis of triple-negative breast cancer via the Wnt/β-catenin pathway. Toxicol Vitro. 2024;95:105746.

    Article  CAS  Google Scholar 

  23. Geng Y, Liu J, Wang Z, Liu T, Peng X, Huang Y. Systematic analysis of the oncogenic role of FAM83D across cancers based on data mining. Cell Cycle (Georgetown Tex). 2023;22(8):1005–19.

    Article  CAS  PubMed  Google Scholar 

  24. Ren S, Zhang Y, Yang X, Li X, Zheng Y, Liu Y, et al. N6-methyladenine- induced LINC00667 promoted breast cancer progression through m6A/KIAA1429 positive feedback loop. Bioengineered. 2022;13(5):13462–73.

    Article  CAS  PubMed  Google Scholar 

  25. Zhao C, Ling X, Xia Y, Yan B, 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. 2021;21(1):441.

    Article  CAS  PubMed  Google Scholar 

  26. Xu LM, Zhang J, Ma Y, Yuan YJ, Yu H, Wang J, et al. MicroRNA-135 inhibits initiation of epithelial-mesenchymal transition in breast cancer by targeting ZNF217 and promoting m6A modification of NANOG. Oncogene. 2022;41(12):1742–51.

    Article  CAS  PubMed  Google Scholar 

  27. Rong D, Dong Q, Qu H, Deng X, Gao F, Li Q, et al. M(6)A-induced LINC00958 promotes breast cancer tumorigenesis via the miR-378a-3p/YY1 axis. Cell Death Discovery. 2021;7(1):27.

    Article  CAS  PubMed  Google Scholar 

  28. Fan S, Wang L. N(6)-Methyladenosine-regulated LINC00675 suppress the proliferation, migration and invasion of breast cancer cells via inhibiting miR-513b-5p. Bioengineered. 2021;12(2):10690–702.

    Article  CAS  PubMed  Google Scholar 

  29. Wei YB, Liang DM, Zhang ML, Li YJ, Sun HF, Wang Q, et al. WFDC21P promotes triple-negative breast cancer proliferation and migration through WFDC21P/miR-628/SMAD3 axis. Front Oncol. 2022;12:1032850.

    Article  CAS  PubMed  Google Scholar 

  30. Li Z, Yang HY, Dai XY, Zhang X, Huang YZ, Shi L, et al. CircMETTL3, upregulated in a m6A-dependent manner, promotes breast cancer progression. Int J Biol Sci. 2021;17(5):1178–90.

    Article  CAS  PubMed  Google Scholar 

  31. Yan C, Xiong J, Zhou Z, Li Q, Gao C, Zhang M et al. A cleaved METTL3 potentiates the METTL3-WTAP interaction and breast cancer progression. Elife. 2023;12.

  32. Ruan HG, Gu WC, Xia W, Gong Y, Zhou XL, Chen WY, et al. METTL3 is suppressed by circular RNA circMETTL3/miR-34c-3p signaling and limits the Tumor Growth and Metastasis in Triple negative breast Cancer. Front Oncol. 2021;11:778132.

    Article  CAS  PubMed  Google Scholar 

  33. Shi Y, Zheng C, Jin Y, Bao B, Wang D, Hou K, et al. Reduced expression of METTL3 promotes metastasis of Triple-negative breast Cancer by m6A methylation-mediated COL3A1 Up-Regulation. Front Oncol. 2020;10:1126.

    Article  PubMed  Google Scholar 

  34. Zhang G, Huang RY, Zhao H, Xia YK, Huang H, Qian MJ, et al. ACAT1-mediated METTL3 acetylation inhibits cell migration and invasion in triple negative breast cancer. Genes Immun. 2023;24(2):99–107.

    Article  CAS  PubMed  Google Scholar 

  35. Cai X, Wang X, Cao C, Gao Y, Zhang S, Yang Z, et al. HBXIP-elevated methyltransferase METTL3 promotes the progression of breast cancer via inhibiting tumor suppressor let-7 g. Cancer Lett. 2018;415:11–9.

    Article  CAS  PubMed  Google Scholar 

  36. Bhattarai PY, Kim G, Lim SC, Mariappan R, Ohn T, Choi HS. METTL3 stabilization by PIN1 promotes breast tumorigenesis via enhanced m(6)A-dependent translation. Oncogene. 2023;42(13):1010–23.

    Article  CAS  PubMed  Google Scholar 

  37. Lin Y, Zheng J, Lin D. PIWI-interacting RNAs in human cancer. Sem Cancer Biol. 2021;75:15–28.

    Article  CAS  Google Scholar 

  38. Huang S, Chen B, Qiu P, Yan Z, Liang Z, Luo K, et al. In vitro study of piwi interaction RNA-31106 promoting breast carcinogenesis by regulating METTL3-mediated m6A RNA methylation. Translational cancer Res. 2023;12(6):1588–601.

    Article  CAS  Google Scholar 

  39. Eisenberg E, Levanon EY, A-to-I. RNA editing - immune protector and transcriptome diversifier. Nat Rev Genet. 2018;19(8):473–90.

    Article  CAS  PubMed  Google Scholar 

  40. Li Y, Wang NX, Yin C, Jiang SS, Li JC, Yang SY. RNA editing enzyme ADAR1 regulates METTL3 in an editing dependent manner to promote breast Cancer progression via METTL3/ARHGAP5/YTHDF1 Axis. Int J Mol Sci. 2022;23(17).

  41. Marra A, Curigliano G. Adjuvant and Neoadjuvant Treatment of Triple-negative breast Cancer with Chemotherapy. Cancer J (Sudbury Mass). 2021;27(1):41–9.

    Article  CAS  Google Scholar 

  42. Fahad Ullah M. Breast Cancer: current perspectives on the Disease Status. Adv Exp Med Biol. 2019;1152:51–64.

    Article  CAS  PubMed  Google Scholar 

  43. Zhou ZY, Cao YK, Yang Y, Wang SM, Chen FY. METTL3-mediated m(6)a modification of lnc KCNQ1OT1 promotes doxorubicin resistance in breast cancer by regulating miR-103a-3p/MDR1 axis. Epigenetics-Us. 2023;18(1).

  44. Zhou S, Sheng L, Zhang L, Zhang J, Wang L. METTL3/IGF2BP3-regulated m6A modification of HYOU1 confers doxorubicin resistance in breast cancer. Biochim et Biophys acta Gen Subj. 2024;1868(3):130542.

    Article  CAS  Google Scholar 

  45. Pan X, Hong X, Li S, Meng P, 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. 2021;53(1):91–102.

    Article  CAS  PubMed  Google Scholar 

  46. Li SM, Jiang FR, Chen FY, Deng YZ, Pan XP. Effect of m6A methyltransferase METTL3-mediated MALAT1/E2F1/AGR2 axis on adriamycin resistance in breast cancer. J Biochem Mol Toxic. 2022;36(1).

  47. Li EJ, Xia MY, Du Y, Long KL, Ji F, Pan FY et al. METTL3 promotes homologous recombination repair and modulates chemotherapeutic response in breast cancer by regulating the EGF/RAD51 axis. Elife. 2022;11.

  48. Liu X, Gonzalez G, Dai X, Miao W, Yuan J, Huang M, et al. Adenylate kinase 4 modulates the resistance of breast Cancer cells to tamoxifen through an m(6)A-Based epitranscriptomic mechanism. Mol Therapy: J Am Soc Gene Therapy. 2020;28(12):2593–604.

    Article  CAS  Google Scholar 

  49. Jing L, Lan L, Mingxin Z, Zhaofeng Z. METTL3/LINC00662/miR-186-5p feedback loop regulates docetaxel resistance in triple negative breast cancer. Sci Rep. 2022;12(1):16715.

    Article  CAS  PubMed  Google Scholar 

  50. Ouyang D, Hong T, Fu M, Li Y, Zeng L, Chen Q, et al. METTL3 depletion contributes to tumour progression and drug resistance via N6 methyladenosine-dependent mechanism in HR + HER2-breast cancer. Breast Cancer Res. 2023;25(1):19.

    Article  CAS  PubMed  Google Scholar 

  51. Cesaro B, Iaiza A, Piscopo F, Tarullo M, Cesari E, Rotili D, et al. Enhancing sensitivity of triple-negative breast cancer to DNA-damaging therapy through chemical inhibition of the m6A methyltransferase METTL3. Cancer Commun (London England). 2024;44(2):282–6.

    Article  Google Scholar 

  52. Mondal P, Natesh J, Penta D, Meeran SM. Progress and promises of epigenetic drugs and epigenetic diets in cancer prevention and therapy: a clinical update. Sem Cancer Biol. 2022;83:503–22.

    Article  CAS  Google Scholar 

  53. Castells M, Butterfield J. Mast cell activation syndrome and mastocytosis: initial treatment options and long-term management. J Allergy Clin Immunol Pract. 2019;7(4):1097–106.

    Article  PubMed  Google Scholar 

  54. Motawi TK, El-Maraghy SA, Sabry D, Nady OM, Senousy MA. Cromolyn Chitosan nanoparticles reverse the DNA methylation of RASSF1A and p16 genes and mitigate DNMT1 and METTL3 expression in breast cancer cell line and tumor xenograft model in mice. Chem Biol Interact. 2022;365:110094.

    Article  CAS  PubMed  Google Scholar 

  55. Cheng L, Zhang X, Huang YZ, Zhu YL, Xu LY, Li Z, et al. Metformin exhibits antiproliferation activity in breast cancer via miR-483-3p/METTL3/m(6)A/p21 pathway. Oncogenesis. 2021;10(1):7.

    Article  CAS  PubMed  Google Scholar 

  56. Wan W, Ao X, Chen Q, Yu Y, Ao L, Xing W, et al. METTL3/IGF2BP3 axis inhibits tumor immune surveillance by upregulating N(6)-methyladenosine modification of PD-L1 mRNA in breast cancer. Mol Cancer. 2022;21(1):60.

    Article  CAS  PubMed  Google Scholar 

  57. Harbeck N, Gnant M. Breast cancer. Lancet (London England). 2017;389(10074):1134–50.

    Article  PubMed  Google Scholar 

  58. Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, et al. The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012;486(7403):400–4.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Editage [www.editage.cn] for English language editing.

Funding

This work was supported in part by the National Natural Science Foundation of China, grant numbers [82371717 and 81971428].

Author information

Authors and Affiliations

Authors

Contributions

DX and MZ contribute to the conception and original draft preparation. YQ and XS contribute to the revision, funding, and supervision of this manuscript. All authors reviewed the manuscript.

Corresponding author

Correspondence to Xiaojuan Su.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiao, D., Zhang, M., Qu, Y. et al. Functions of methyltransferase-like 3 in breast cancer: pathogenesis, drug resistance, and therapeutic target. Breast Cancer Res 26, 110 (2024). https://doi.org/10.1186/s13058-024-01869-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13058-024-01869-8

Keywords