Abstract
Epitranscriptomic modification of RNA regulates human development, health, and disease. The true diversity of the transcriptome in breast cancer including chemical modification of transcribed RNA (epitranscriptomics) is not well understood due to limitations of technology and bioinformatic analysis. N-6-methyladenosine (m6A) is the most abundant epitranscriptomic modification of mRNA and regulates splicing, stability, translation, and intracellular localization of transcripts depending on m6A association with reader RNA binding proteins. m6A methylation is catalyzed by the METTL3 complex and removed by the specific m6A demethylase ALKBH5, with FTO’s role as an ‘eraser’ uncertain. In this review, we provide and overview of epitranscriptomics related to mRNA and focus on m6A in mRNA and its detection. We summarize current knowledge on altered levels of writers, readers, and erasers of m6A and their roles in breast cancer and association with prognosis. We summarize studies identifying m6A peaks and sites in genes in breast cancer cells.
Keywords: epitranscriptome, m6A, mRNA, breast cancer
Introduction
Breast cancer is the most commonly diagnosed cancer worldwide, although countries with the highest human development index, related to life expectancy, education and wealth, have the highest incidence of breast cancer (BC) (Wilkinson and Gathani 2021). Breast tumors are characterized by cellular heterogeneity, abnormal gene expression, and chaotic signaling associated with genetic, epigenetic, and epitranscriptomic changes (Lüönd, et al. 2021; Yang, et al. 2021). Breast cancer is divided into four major clinical subtypes based on varying expression of hormone receptors (HR) estrogen receptor α (ERα, referred to as ER+) and progesterone receptor(PR) and human epidermal growth factor receptor (HER2 ERBB2-)) (Dai, et al. 2016) (Dunnwald, et al. 2007), or the lack of these receptors in triple negative breast cancer (TNBC). These molecular markers inform treatment options. Due to early diagnosis and improved treatment options, the death rate from BC has decreased significantly during the last 20 years (Siegel, et al. 2021; Siegel, et al. 2022; Sung, et al. 2021). Based on individual risk assessment, patients with HR+/HER2- primary tumors receive 5–10 years of single agent endocrine therapies (ET): an aromatase inhibitor (AI, e.g., letrozole) for postmenopausal women or tamoxifen (TAM) and ovarian suppression for premenopausal women (Burstein, et al. 2016; Burstein, et al. 2010; Burstein, et al. 2014). For metastatic ER+ BC, patients are treated with combined ET and CDK4/CDK6 inhibitors, PI3K inhibitors, PARP inhibitors, and or anti-PD-L1 immunotherapy, depending on molecular profiling (reviewed in (Loibl, et al. 2021)). Although the PAM50 risk of recurrence score (PAM50-ROS) is an example of ‘precision medicine’ and patient-customized-therapy’ with a predictive recurrence value in ET-treated ERα+ BC patients (Giorgi Rossi, et al. 2021), > 43,700 women in the U.S. will die of metastatic BC in 2022 (Siegel et al. 2022).
If a breast tumor expresses little (< 1%) or no ER/PR, but has a high expression of HER2, the patient is treated with the humanized monoclonal antibodies trastuzumab and pertuzumab directed against the extracellular domain of HER2 (ERB2) (von Minckwitz, et al. 2017) and Trastuzumab deruxtecan (T-DXd), an antibody-drug conjugate with a topoisomerase I inhibitor payload for metastatic of unresectable disease (Bardia, et al. 2022). Patients with TNBC receive chemotherapy +/− radiation therapy as first line therapy (Bianchini, et al. 2016). New FDA-approved therapeutic options used in combination with chemotherapy for patients with local recurrent unresectable or metastatic TNBC include PD-L1 inhibitors atezolizumab and pembrolizumab (Torres and Emens 2022), PARP inhibitors (olaparib or talazoparib, depending on germline BRCA gene mutations) (Cardoso, et al. 2020), and Sacituzumab govitecan (SG) that is antibody conjugate with anti-trophoblast cell surface antigen-2 (Trop-2), targeting Trop-2-expressing cancer cells, linked with cytotoxic moiety SN-38 (govitecan) with topoisomerase I inhibitor activity (Michaleas, et al. 2022). A recent pilot clinical trial using personalized immunotherapy, infiltrating lymphocytes(TILs), showed that six of 42 patients with metastatic BC developed objective cancer regression (Zacharakis, et al. 2022).
For ~ 25–40% of BC patients with ER+ breast tumors, ET-resistance develops and overall survival (OS) for patients with metastatic disease is only ~ 44.6 mos. (Polley, et al. 2021). Significant progress has been made in understanding the molecular mechanisms for ET resistance (reviewed in (Clarke, et al. 2015)) and includes mutations in ESR1 and ESR1 fusion transcripts in ~30% of metastatic tumors from patients on aromatase inhibitor ET therapy (reviewed in (Piscuoglio, et al. 2018; Toy, et al. 2017)). Increased progression-free survival (PFS) has been realized with the addition of cyclin-dependent kinase 4 & 6 (CDK4 & 6) inhibitors to ET for women with advanced BC (Cardoso et al. 2020; Sledge, et al. 2020). However, metastatic spread occurs early in tumor progression and likely precedes detection of the primary tumor (Celià-Terrassa and Kang 2016; Sanger, et al. 2011). Thus, identifying features in the primary tumor that reflect its metastatic potential and blocking progression is key to preventing recurrent disease. Whether epitranscriptomic RNA modifications in breast tumors may hold clues to metastatic potential and the mechanism(s) and targets that these modifications regulate are currently unknown. Analysis of BC data in ONCOMINE revealed changes in the transcript levels of writers, readers, and erasers of the most common RNA modification, N-6-methyladenosine (m6A) with two readers, RBM15B and ZC3H13, positively associated with ER and PR expression and with longer relapse-free survival (RFS) (Zhang, et al. 2020). However, there are conflicting data with respect to associations of these and other m6A writers, readers, and erasers in BC (Liu, et al. 2022a). There are few studies of how the position of m6A affects transcript splicing, stability, or translation in BC cells or tumors, indicting the need for closer examination of the importance of the regulation of the epitranscriptome in subtype specific BC and metastatic disease.
An Overview of Epitranscriptomics
Over 160 chemical modifications of transcribed RNA have been identified (Mathlin, et al. 2020). The proteins that add the chemical modification, recognize the specific RNA base modification, and remove that modification are termed ‘writers, readers, and erasers’. The term epitranscriptomics refers to these chemical modifications of transcribed RNA and includes modifications of adenosine (A), guanosine (G), cytidine (C), and uridine (U) (reviewed in (McCown, et al. 2020)). The Modomics database of RNA modification pathways summarizes these modifications (Boccaletto, et al. 2022). These modifications affect the structure of RNA and its interactions with proteins, including readers (Jones, et al. 2022; Kierzek, et al. 2022; Szabat, et al. 2022). Transfer RNAs (tRNAs) are the most extensively modified cellular RNAs with over 100 chemical modifications (Boccaletto and Bagiński 2021) and each tRNA containing 11–13 modifications (Delaunay and Frye 2019). The tRNA chemical modifications within the D-, T-, and anticodon- loops regulate tRNA stability, folding, and interaction with the Elongator complex for translational fidelity (Pereira, et al. 2018). 130 modifications have been identified in rRNA, including isomerization of uridine to pseudouridine (Ψ) and 2′-O-methylation of the ribose (Am) (Delaunay and Frye 2019). Mitochondrial-encoded rRNAs are also highly modified (Laptev, et al. 2020). There are thirteen known internal mRNA modifications located in the 5’ and 3’ UTRs as well as exons and introns (Anreiter, et al. 2021). m6A is the most common dynamic modification of the transcriptome and regulates the function and processing of mRNAs, long non-coding RNAs (lncRNAs), and primary microRNAs (pri-miRNAs) to precursor (pre)-miRNAs (Zaccara, et al. 2019). In mammalian mRNA, ~ 0.1%–0.4% of RNA are exposed to m6A modification, with an average of 3–5 m6A sites per transcript (Roundtree, et al. 2017a). m6A is also enriched in circRNAs, lncRNAs, and U6 snRNA (Zaccara et al. 2019). m6A is enriched in the 3’UTR and within exons, highlighting its role in regulating mRNA processing, alternative splicing, stability, and translation (Kretschmer, et al. 2018; Roundtree et al. 2017a). The ‘landscape’ of m6A modification is species- and cell-type specific (Liu, et al. 2019a; Wang and Wang 2020) and plays an important role in various cancers (reviewed in (Niu, et al. 2022))
m6A is dynamic and is added by a METTL3 ‘writer’ complex at DRACH (RRACH) (D or R = A/G/U, R = A/G, H = A/C/U) consensus motifs and removed by ‘erasers’ FTO and ALKBH5 (Knuckles and Bühler 2018). FTO and ALKBH5 are α-ketogluterate-dependent dioxygenases sensitive to D-2-hydroxygluterate-mediated inhibition connecting glucose metabolism to m6A (Su, et al. 2018). Demethylases FTO and ALKBH5 are the ‘erasers’ of m6A methylation (Zhao, et al. 2020), although since FTO is nuclear, it may only demethylate m6Am in the 5’ N-terminal cap of mRNA (Mauer, et al. 2019). The role of m6A depends on the proteins that recognize and ‘read’ this marker, including YTHDC1–3, YTHDF1–3, and HNRNPA2B1 (Lewis, et al. 2017; Licht and Jantsch 2016). An additional number of RNA binding proteins and transcription factors, including glucocorticoid receptor (GR), also bind m6A (An, et al. 2020).
The mechanisms regulating and roles of epitranscriptomic mRNA modifications other than m6A are largely unknown (Zhao et al. 2020). Common epitranscriptomic marks in mRNA are involved in translation efficiency, i.e., m6A, m6Am, m1A, m5C, Ψ, and hm5C, RNA structure and stability, export, and degradation (Table 1) (Kumar and Mohapatra 2021). The biological roles of the epitranscriptome in human biology and disease and the writers, readers, and erasers of these modifications is of great research interest. New strategies are being developed to address the technical challenges in identifying and understanding the biological importance of the epitranscriptome
Table 1:
Selected mRNA epitranscriptomics (Wiener and Schwartz 2021)
| modification | Role in mRNA | Writer | Readers | Erasers | Role in breast cancer |
|---|---|---|---|---|---|
| Ψ (Pseudouridine) | enriched in alternatively spliced regions (Martinez, et al. 2022); enhanced mRNA translation (Hoernes, et al. 2019), role in mRNA not well-understood (Borchardt, et al. 2020) | PUS1, PUS3, PUS7, PUS7L, PUS10, DKC1, TRUB1,(Anreiter et al. 2021; Chen et al. 2021a; Chen, et al. 2019b; Pereira et al. 2018) Ψ is also converted from uridine through a post-transcriptional isomerization reaction (Adachi, et al. 2019). |
Unknown for mRNA (Levi and Arava 2021) | Unknown for mRNA (Cerneckis, et al. 2022) | Role in mRNA modification is unknown (Xue, et al. 2022) |
| m5C (5-methylcytosine) | mRNA stability, translation, and export (Chen et al. 2021a; Chen, et al. 2021b; Yang, et al. 2017); enriched in CG-rich regions in the 3’UTRs of genes and immediately downstream of translation initiation sites and has conserved, tissue-specific and dynamic features across mammalian transcriptomes (Yang et al. 2017). | NSUN2 (Yang et al. 2017) | ALYREF/THOC4, the mammalian mRNA export adaptor (Yang et al. 2017) | TET2 (Chen et al. 2021b) | m5C writer NSUN2 was upregulated in breast cancer and promoted proliferation, migration, invasion and tumorigenicity (reviewed in (Li, et al. 2022c)) |
| m1A (1-methyladenosine) | affects mRNA structure and interaction with RNA binding proteins (Dominissini, et al. 2016). | TRMT6 TRMT61A TRMT61B (mt) (Li, et al. 2016b; Pereira et al. 2018) |
YTHDF1–3(Anreiter et al. 2021) | ALKBH1 (Liu, et al. 2016), ALKBH3 (Chen, et al. 2019c) |
Demethylation of m1A promotes breast cancer invasion (reviewed in (Kumari, et al. 2021)) |
| m6A (N6-Methyladenosine) | splicing, stability, translation, degradation (Dinescu, et al. 2019) | METTL3 METTL14 WTAP VIRMA RBM15 RBM15B ZC3H13 WTAP CBLL1 METTL16 (Balacco and Soller 2019; Licht and Jantsch 2016; Wei, et al. 2018; Wen, et al. 2018) |
YTHDF1–3, YTHDC1–2, HNRNPA2B1, IGFBP1–3, PRRC2A, FMR1, ELAVL1, HNRNPC, HNRNPG (Alarcón et al. 2015a; Anreiter et al. 2021; Bi, et al. 2019; Chen, et al. 2019a; Yang, et al. 2018; Zhou, et al. 2019) |
FTO, ALKBH5 (Licht and Jantsch 2016) | Writers, readers, and erasers have roles in breast cancer (Reviewed in (Deng, et al. 2018; Fang, et al. 2021)) |
METTL3 methyltransferase complex (MTC)
METTL3 is the ‘writer’ of the m6A modification in mRNA. The METTL3 complex (875 kDa) includes the METTL3-METTL14 heterodimeric complex and auxiliary proteins that enhance the m6A methyltransferase activity: WTAP (Wilms tumor suppressor-1–associated protein), CBLL1 (Cbl Proto-Oncogene Like 1, HAKAI), VIRMA (Vir Like m6A Methyltransferase Associated), RBM15 and RBM15B (RNA Binding Motif Protein 15), and ZC3H13 (Zinc Finger CCCH-Type Containing 13) (reviewed in (Covelo-Molares, et al. 2021; Zaccara et al. 2019)) (Table 2). Crystal structure studies revealed that METTL3 and METTL14 interact over a large surface area via their methyltransferase domains (MTD) to bind RNA (Zaccara et al. 2019). METTL3 catalyzes the m6A methylation by taking the methyl from the cofactor S-adenosylmethionine (SAM) and releasing S-adenosylhomocysteine (SAH) while METTL14 is catalytically inactive (Wang, et al. 2016; Zhou and Pan 2016). METTL14 stabilizes METTL3 and contributes to RRACH (R = A/G, H = A/U/C) site binding. METTL3/METTL14 was also shown to preferentially bind RNA G-quadruplex motifs (G4) that are enriched in human gene promoters, suggesting that G4s are involved in transcription regulation by the MTC (Yoshida, et al. 2022). The MTC is localized to gene promoters to deposit m6A on nascent transcripts, including enhancer RNAs (eRNAs) and promoter upstream transcripts (PROMPTs) (Xu, et al. 2022), and plays a role in RNA polymerase II pause release (Akhtar, et al. 2021).
Table 2:
METTL3 methyltransferase complex proteins and their role in breast cancer.
| Protein (other names) | Function | Breast cancer activity |
|---|---|---|
| METTL3 | m6A methyltransferase | Oncogene (Cai et al. 2018; Pan et al. 2021; Wang et al. 2020a) Tumor suppressor in TNBC (Shi et al. 2020) |
| METTL14 | Heterodimer partner for METTL3 m6A methyltransferase | Oncogenic (Sun et al. 2020). |
| WTAP | Stabilizes the METTL3-METTL14 heterodimer to nuclear speckles and interacts with splicing factors (Horiuchi, et al. 2021) | Oncogene, but inhibits metastasis (Wang et al. 2022) |
| CBLL1 (Hakai) | E3 ubiquitin ligase, but its enzymatic activity is dispensable for its function in stabilizing the METTL3 complex by interacting with WTAP and VIRMA (Zhang, et al. 2022) | Tumor suppressor and low expression associated with TAM resistance (Zheng, et al. 2021a); higher expression associated with higher RFS (Zheng, et al. 2021b) |
| VIRMA (KIAA1429) | Cofactor that guides the METTL3 complex to bind RNA | Oncogene (Qian et al. 2019); upregulated in breast tumors (Fang et al. 2021) |
| ZC3H13 (KIAA0853) | Component of the WTAP complex | Oncogene, positively associated with ER/PR expression (Zhang et al. 2020); high expression correlated with reduced OS (Mu, et al. 2022; Tai, et al. 2022) |
| RBM15/15B | RNA binding proteins that interact with METTL14 | RBM15 is high in basal-like breast tumors whereas RBM15B was higher in Luminal A and normal breast-like subtypes of breast cancer (Zhang et al. 2020). |
The N-terminal coiled-coil domain of WTAP interacts with the N-terminal leader helix of METTL3 and is required for methyltransferase activity (Zaccara et al. 2019). WTAP regulates METTL3/METTL14 localization to nuclear speckles for pre-mRNA interaction, methylation, and splicing (Ping, et al. 2014). Nuclear speckles are dynamic interchromatin granules that function as sites for RNA 5’capping, 3’end processing, splicing and m6A methylation (reviewed in (Galganski, et al. 2017; Ilık and Aktaş 2021)). Recent studies show that METTL3 undergoes phase separation in HEK-293T nuclei supporting the scaffolding of the METTL3 complex to promote m6A mRNA modification (Han, et al. 2022). CBLL1 (HAKAI) is an E3 ubiquitin ligase that interacts with WTAP and is required to writer-complex interactions (Zaccara et al. 2019). ZC3H13 also interacts with WTAP and with RBM15/15B and is required for m6A methylation (Zaccara et al. 2019).
BioID, a proximity-dependent labelling approach in which a bait protein is fused to an E. coli biotin ligase (BirA*) is used to label proteins within a 10 nm radius (Liu, et al. 2018), and coupled to liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to identify METTL3 interacting proteins in HEK-293T cells (Covelo-Molares et al. 2021). In addition to the proteins listed in Table 2, a number of other proteins involved in transcription, e.g., STAT5B, ZEB2; chromatin remodeling, i.e., BCL7C; DNA replication, i.e., CHFR and MCMBP; and protein modification, e.g., SUGT1, DNAJA2, FKBP4/5, were identified (Covelo-Molares et al. 2021). These results have not been validated or extended in other cell lines, nor in BC cells.
A novel circRNA from the METTL3 gene, circMETTL3 (623 nt originating from exon-2 and exon-3) was identified as highly expressed in in breast tumors (Li, et al. 2021b). Experiments in ZR-75–1 and SUM1315 BC cells demonstrated that siRNA targeting METTL3 inhibited cell proliferation, migration, invasion, and xenograft tumor growth in vivo (Li et al. 2021b). circMETTL3 was reported to sponge miR-31–5p, thus increasing its target CDK1 which drives BC progression (Li et al. 2021b).
MTC transcripts and proteins in breast cancer
Examination of BC data in The Cancer Genome Atlas (TCGA) showed decreased transcript levels of METTL3, METTL14, and WTAP (1.575-, 1.200- and 1.883-fold) in breast tumors compared to normal controls (Wu, et al. 2019). Similar findings for METTL3, METTL14, and WTAP using the ONCOMINE BC data were reported; additionally, ZC3H13 transcript was also decreased, but RBM15B and VIRMA transcripts were increased (Zhang et al. 2020). qRT-PCR examination of 36 breast tumor samples (no histopathological data were included) identified significant reductions in the expression of METTL3, METTL14, and WTAP (Wu et al. 2019). These breast tumors had reduced global m6A levels in total RNA (primarily rRNA and tRNA with ~ 5% mRNA), although the inter-sample variability was high, with some tumor samples showing elevations in gene transcript and in m6A (Wu et al. 2019).
Other studies reported increased METTL3 in BC cells and tumors. Increased METTL3 mRNA expression in breast tumors correlated with increased expression of Late Endosomal/Lysosomal Adaptor, MAPK and MTOR Activator 5 (LAMTOR5, HBXIP) in BC cell lines and patient breast tumor arrays (TMA) (Cai, et al. 2018). This study demonstrated that HBXIP increased let-7g which directly targeted and reduced METTL3 in MCF-7 luminal A (ER+/PR+, HER2-) BC cells and that METTL3-induced m6A methylation of HBXIP increased HBXIP protein expression (Cai et al. 2018). METTL3 transcript levels were higher in GEPIA database, in breast tumors, in BC cell lines (including MCF-7 and MDA-MB-231 TNBC cells) compared to MCF-10A (immortalized ‘normal’ breast epithelial) cells (Wang, et al. 2020a). Knockdown of METTL3 by shRNA (~ 60% decrease in protein) in MCF-7 and MDA-MB-231 cells inhibited cell proliferation and reduced BCL2,an apoptosis regulator (Wang et al. 2020a). High METTL3 transcript was associated with lower RFS in the Kaplan-Meier plotter (Gyorffy, et al. 2010) used in the study (Wang et al. 2020a). Higher METTL3 protein correlated with higher levels of m6A (dot blot) in BC cell lines (MCF-7, T47D (luminal A), MDA-MB-231, SKBR3 (ER-/PR-/HER2(ERBB2)+, and BT474 (ER+/PR+/HER2+)) and knockdown of METTL3 (shRNA) increased sensitivity to growth inhibition by chemotherapeutics in vitro: ADR, 5-FU, cisplatin, paclitaxel, and carboplatin (Li, et al. 2022b). METTL3 promoted double-stranded DNA (dsDNA) break repair and homologous recombination repair (HHR) after DNA damage induced by etoposide (ETO) and adriamycin in MCF-7 and MDA-MB-231 cell lines (Li et al. 2022b). The mechanism involved METTL3 m6A methylation of epidermal growth factor (EGF) which increased EGF translation via interaction with m6A reader YTHDC1. In turn, EGF increased the expression of RAD51, a DNA repair enzyme, RAD51 recombinase (Li et al. 2022b). Inhibiting DNA repair is an important target in BC to enhance the efficacy of chemotherapy, e.g., the use of PARP1 inhibitors (McCann, et al. 2019). METTL3 expression was upregulated in BC stem cells (BCSC) and increased expression of AF4/FMR2 family member 4 (AFF4) which binds the MYC and SOX2 gene promoters to regulate ‘stemness’ (Gao, et al. 2020)
METTL14 mRNA and protein expression was increased in breast tumors and some BC cell lines (Sun, et al. 2020). LINC00942 (LNC942) is upregulated in BC was shown to promote METTL14-mediated m6A methylation by interacting with METTL14 and upregulating the transcription of specific target genes, CXCR4 and CYP1B1, that play roles in EMT and BC tumorigenesis (Sun et al. 2020). Upregulation of oncogenic lncRNA UCA1 in T47D BC cells inhibited METTL14 transcription by recruiting DNMT1, DNMT3A/B to increase METTL14 promoter DNA methylation. Consequently the decrease in METLL14 reduced m6A in pri-miR-375 which increased pre-miR-375 and mature miR-375 that reduced SOX12 translation, resulting in increased proliferation and invasion, and reducing apoptosis (Zhao, et al. 2022).
Aurora kinase A (AURKA) positively regulated METTL14 protein expression in BC cells in a protein kinase-independent manner by inhibiting its ubiquitination-mediated degradation to increase m6A content in the DROSHA mRNA transcript resulting in increased DROSHA mRNA stability and binding of IGF2BP2 in MDA-MB-231 BCSC (Peng, et al. 2021).
Nuclear WTAP protein expression was higher in breast tumor specimen (n = 347) compared to noncancerous breast tissue (n = 23) with higher staining associated with tumor size > 2cm, higher tumor grade (II or III), and in ER- tumors compared to TNBC which had the highest WTAP staining (Wang, et al. 2022). We reported that WTAP transcript abundance was higher in LCC9 endocrine-resistant BC cells compared to MCF-7 parental cells (Klinge, et al. 2019b). Whether ER inhibits WTAP expression in a ligand-dependent manner is unknown. HER2 status had no correlation with WTAP staining (Wang et al. 2022). Interestingly, higher WTAP staining was observed in LN metastatic negative compared to LN metastatic positive breast tumors. The authors suggested this result to indicate a role for WTAP as a suppressor of metastasis; however, no metastatic tumors from distant site were examined (Wang et al. 2022). There was no association WTAP protein expression and breast cancer patient survival (Wang et al. 2022).
Hypoxia increases the expression of METTL14 in MDA-MB-231 cells (Panneerdoss, et al. 2018). Knockdown of METTL14 inhibited the growth, migration, and invasion of MDA-MB-231, MDA-MB-468, and BT-549 TNBC cells in vitro and MDA-MB-231 tumor xenografts in vitro by ~ 30% (Panneerdoss et al. 2018). The authors found that METTL14 and the m6A eraser ALKBH5 constitute a positive feedback loop with the RNA binding protein HuR (ELAV1) to regulate the stability of target genes, i.e., cyclin D1, SMAD3, cyclin E1, TGFβ1, VEGFA, HMGA2, and PDGF, in MDA-MB-231 cells (Panneerdoss et al. 2018).
METTL3 methylation of m6A on pri-miRNAs (Alarcón, et al. 2015b) and RNA-dependent interaction of the m6A reader HNRNPA2B1 with DGCR8, a component of the DROSHA complex, stimulate processing of selected pri-miRNA-m6A to precursor miRNA (pre-miRNA) (Alarcón, et al. 2015a). METTL3 protein expression was increased in adriamycin-resistant MCF-7 breast cancer cells (MCF-7/ADR) and enhanced pri-miR-221–3p m6A methylation and increased mature miR-221–3p expression which decreased Homeodomain Interacting Protein Kinase 2 (HIPK2) protein expression in MCF-7/ADR cells (Pan, et al. 2021).
VIRMA (KIAA1429) is the largest (202 kDa) component of the METTL3 complex and acts as a scaffold and bridges the METTL3/METTL14/WTAP complex with RNA and is upregulated in BC at the transcript and protein levels (Qian, et al. 2019). Knockdown of VIRMA (KIAA1429) in MCF-7 and MDA-MB-231 BC cells inhibited proliferation, migration, invasion, and colony formation (Qian et al. 2019). VIRMA was shown to have the most copy number variations (CNV) among m6A readers, writers, and erasers (Zhao, et al. 2021). Further studies are needed to validate this report.
RNA binding proteins as m6A readers
While studies have identified altered m6A on transcripts in human disease, their potential mechanism of action and involvement in human pathologies is not well understood. A focus of m6A research has been characterizing the protein complexes responsible for identifying the reversible and dynamic m6A mark in mRNA and investigating the downstream effects resulting from m6A modification (reviewed in (Zaccara and Jaffrey 2020; Zaccara et al. 2019)). At the time of this review, 20 RNA-binding proteins (RBPs) have been identified to bind to m6A-methylated regions of the mRNA transcript and regulate transcript outcome for protein expression (Du, et al. 2016; Wang, et al. 2014; Wang, et al. 2015; Xiao, et al. 2016). m6A readers with known relevance to breast cancer, their cellular location, and molecular functions are summarized in Table 3. These m6A readers have been shown to play a role in embryonic stem cell fate (Batista, et al. 2014; Lasman, et al. 2020) energy homeostasis (Edupuganti, et al. 2017), human circadian rhythm (Robinson, et al. 2019), and various cancers (Jing, et al. 2021; von Hagen, et al. 2021). Evidence suggesting that the dysregulation of m6A readers plays a pivotal role breast cancer progression will be described below.
Table 3. RNA-binding proteins identified as m6A readers.
Each m6A reader, location, function, and role in breast cancer are listed.
| Enzymes | Location | Cellular function | Additional roles in breast cancer |
|---|---|---|---|
| YTHDF1 | Cytoplasm | RNA translation efficiency (Wang et al. 2015) | YTHDF1 mediated translation of Spred2, a gene that encodes for SPRED2, a negative regulator of ERK signaling, in murine bone marrow-derived macrophages (BMDMs) (Yin, et al. 2021). Elongation and translation of KRT7, which is higher in BT-549 cells, is mediated by the YTHDF1/eEF-1 complex (Chen et al. 2021a) |
| YTHDF2 | Cytoplasm | RNA degradation (Du et al. 2016; Wang et al. 2014) | YTHDF2 is upregulated in TCGA-BRCA analysis of TNBC tumor tissue (Wang, et al. 2020b). |
| YTHDF3 | Cytoplasm | RNA degradation or translation efficiency (Li, et al. 2017; Shi et al. 2017) | Knockdown of YTHDF3 in MDA-MB-231 and BT-549 cells decreased the expression of EMT network protein ZEB1 (Lin et al. 2022). |
| YTHDC1 | Nucleus | Alternative splicing (Patil et al. 2016; Roundtree, et al. 2017b) | YTHDC1 is downregulated in TCGA-BRCA analysis of TNBC tumor tissue (Wang et al. 2020b). |
| YTHDC2 | Cytoplasm | RNA translation efficiency (Hsu et al. 2017; Mao, et al. 2019) | YTHDC2 is downregulated in TCGA-BRCA analysis of TNBC tumor tissue (Wang et al. 2020b) |
| IGF2BP1 (IMP1, CRD-BP) | Cytoplasm | RNA stability (Fakhraldeen, et al. 2015; Stohr, et al. 2012; Zhang et al. 2021a) | IGF2BP1 interacted with lncRNA KB-1980E6.3 to stabilize MYC in breast cancer stem cells (Zhu, et al. 2021). Decreased expression of IGF2BP1 by miR-873–5p in BT-549 cells reduces cMYC expression (Tang, et al. 2020). Phosphorylation of tyrosine 260 on a truncated form of IGF1BP2 (ΔN-IMP1), which is expressed in ER+ cell lines MCF-7 and ZR-75–1, is required for MYC mRNA stabilization (Abdullah, et al. 2018) |
| IGF2BP2 | Cytoplasm | RNA stability (Dai, et al. 2015; Pu, et al. 2020; Simon, et al. 2014) | Overexpression of IGF2BP2 (p62/IMP2) in MDA-MB-231 cells increased MYC expression and cell migration, while reducing cell adhesion (Li, et al. 2015) |
| IGF2BP3 | Cytoplasm | RNA stability (Yang et al. 2020) | IGF2BP3 interacts with circFOXK2 and miR-370 to promote cell migration and invasion in BT-549 cells (Zhang, et al. 2021b). IGF2BP3 depletion repressed TRIM25 expression and reduced MCF-7 and MDA-MB-231 proliferation (Wang, et al. 2019). |
| HNRNPA1 | Nucleus | Alternative splicing (Loh, et al. 2015) | HNRNPA1 is higher in breast, colorectal, and prostate cancer cells and associated with increased expression of type I and type II methyltransferases (PRMTs) (Li et al. 2021a). Alternative splicing of CD44 was mediated by HNRNPA1 in MDA-MB-231 cells and was associated with metastatic progression (Loh et al. 2015). |
| HNRNPA2B1 | Nucleus | Alternative splicing and miRNA processing (Alarcón et al. 2015a) | Based in TCGA breast cancer datasets, HNRNPAB is increased with elevated histological grade and higher in MDA-MB-468 TNBC cells compared to MCF-7 cells (An et al. 2021). Plays a role in endocrine-therapy resistance in an MCF-7 cell model (Klinge et al. 2019a; Petri et al. 2021). |
| HNRNPC | Nucleus | m6A switch recognition, Alternative splicing (Liu et al. 2015) | HNRNPC is upregulated in TCGA-BRCA analysis of TNBC tumor tissue (Wang et al. 2020b). |
| FMR1 | Nucleus/Cytoplasm | RNA trafficking, translation regulation (Darnell et al. 2011) | Genotype analysis of BRCA patients identified an association between FMR1 expression and the BRCA1/2 mutation (Tea, et al. 2013). |
| LRPPRC | Nucleus/Mitochondria | mRNA maturation (Cui, et al. 2019) | LRPPRC suppression by miR-1 in stem cells isolated from MCF-7 cells induced mitochondrial damage (Zhang, et al. 2019). |
| PRRC2A | Nucleus/Cytoplasm | RNA stabilization (Chen et al. 2019a) | PRRC2A is upregulated in breast cancer compared to normal samples in TCGA breast cancer data (Yang et al. 2021) |
| SND1 | Nucleus/Cytoplasm | Alternative splicing, RNA editing, and miRNA-mediated mRNA degradation (Paukku, et al. 2008; Scadden 2005) | SND1 interacted with Linc00668 and induced SMAD2, SMAD3, and SMAD4 transcription in MDA-MB-231 cells. Depletion of SND1 in MDA-MB-231 and MCF-7 cells reduced migratory ability and invasiveness (Qian, et al. 2020; Yu et al. 2015). Higher SND1 (IHC tumor vs. normal) was associated with advanced stages and larger breast tumor size (Gu, et al. 2018). |
| ELAV1 (HuR) | Nucleus/Cytoplasm | RNA stability (Ling, et al. 2020) | ELAV1 stabilized and promoted transcription of ZEB1 in MDA-MB-436 and MDA-MB-453 cells, promoting migratory ability and invasiveness (Luo, et al. 2020). |
| EIF3 | Cytoplasm | RNA translation initiation | EIF3m knockdown in MDA-MB-231 and MDA-MB-468 cells decreased cell proliferation, migration, and invasion (Han et al. 2020). EIF3D knockdown inhibits activation of Wnt/β-catenin signaling in MDA-MB-231 and MCF-7 cells (Fan and Guo 2015). EIF3e knockdown reduces levels of PARP1 in MDA-MB-231 cells inducing senescence (Morris et al. 2021). |
YTH Domain m6A-binding proteins
The earliest identified m6A readers contain a broadly conserved YT521-B homology (YTH) domain with tryptophan residues forming a hydrophobic pocket that specifically recognizes and encapsulates m6A-containing RNA (Luo and Tong 2014). Five YTH domain-containing proteins have been identified in the mammalian genome; YTHDC1, YTHDC2, and three YTHDF paralogs - YTHDF1, YTHDF2, and YHDF3 (reviewed in (Zaccara and Jaffrey 2020)). In 2012, the binding affinity of YTHDC1, YTHDF2, and YTHDF3 to m6A was validated using affinity chromatography followed by mass spectrometry (MS) (Dominissini, et al. 2012). The direct binding of these reader proteins as well as YTHDC2 have been validated using individual-nucleotide-resolution UV crosslinking and immunoprecipitation (iCLIP) (Patil, et al. 2016). YTHDF family members are primarily cytoplasmic and contain the YTH domain located at the C-terminus as well as a larger domain enriched with Q, N, and P residues (Meyer and Jaffrey 2017). Studies suggest that m6A-containing mRNAs have a reduced half-life compared to mRNAs without m6A (Batista et al. 2014; Geula, et al. 2015); however, each YTHDF paralog has different functions related to both mRNA stability and translation efficiency. Binding of YTHDF1 to m6A in the 3’ untranslated region (UTR) recruits eukaryotic translation initiation factor 3 (eIF3) to facilitate 5’ cap and polyA tail addition, as well as the forming of the cyclic structure to promote translation (Wei, et al. 2022). In contrast, binding of YTHDF2 to m6A sites in the 3’UTR leads to shortening of the polyA tail length and localizes m6A-modified mRNA transcripts to processing bodies in the cytoplasm, promoting mRNA degradation (Wang et al. 2014; Wei et al. 2022). A study in human astrocytes exploring the role of YTHDF2 in mRNA degradation showed that YTHDF3 influences proliferation and migration of glioma cells by suppressing UBX domain protein 1 (UBXN1), thus inducing NF-kB activation (Chai, et al. 2021). The role of YTHDF3 is less clear, although it appears to share many targets with YTHDF1 and YTHDF2, suggesting dynamic and cooperative activity between the YTHDF proteins regulating translation and decay of common mRNAs (Shi, et al. 2017).
YTHDC1 is predominantly nuclear (Patil et al. 2016). An investigation of YTHDC1 nuclear interacting proteins in HeLa cells revealed the enrichment of splicing enhancer binding SR proteins, SRSF1 (ASF/SF2), SRSF3 (SRp20), SRSF7 (9G8), SRSF9 (SRp30c), and SRSF10 (SRp38), identifying a role for YTHDC1 in alternative splicing (Xiao et al. 2016). The function of cytoplasmic YTHDC2 is less defined, but it includes a DEAD-box RNA helicase domain that may be targeted to specific mRNAs by m6A (Patil et al. 2016). A direct role for YTHDC2 for translation efficiency was discovered using luciferase-based tethered reporter assays in HeLa cells (Hsu, et al. 2017).
IGF2BP family m6A-binding proteins
Insulin-like growth factor-2 (IGF2) mRNA–binding proteins (IGF2BPs) constitute a conserved family of single-stranded RBPs that specifically recognize m6A-RNA and influence transcript outcomes (Bell, et al. 2013; Du, et al. 2021; Korn, et al. 2021). The role of IGF2BPs in regulating transcript outcomes has been studied since 2002, well before IGF2PBs were discovered to be m6A readers, and several synonyms for IGF2BP exist in the literature, including, but not limited to, IMP, CRD-BP, VICKZ and ZBP (Bell et al. 2013). IGF2BP1/2/3 are predominantly cytoplasmic and modulate cell function during development and cancer (Samuels, et al. 2020; Stöhr, et al. 2012; Zhou, et al. 2022). These three IGF2BP proteins have a 56% aa sequence identity with two RNA-recognition motifs (RRMs) at their N-termini and hnRNP-K homology (KH) domains at their C-termini (Bell et al. 2013). This sequence homology suggests that the IGF2BP proteins share biochemical functions. IGF2BPs form ribonucleoprotein (RNP) granules in the cytoplasm and encapsulate their target mRNAs in protein-RNA complexes (mRNPs), preventing premature decay (Kobel, et al. 2007; Lemm and Ross 2002; Vikesaa, et al. 2006).
RNA-pulldown experiments using methylated single-stranded RNA bait followed by mass spectrometry, confirmed that IGF2BPs selectively bind m6A-RNA independent of RNA secondary structure and depletion of IGF2BPs globally downregulated target gene expression (Huang, et al. 2018). In human endometrial cancer cells, IGF2BP1 binds to the m6A mark on PEG10 mRNA and recruits polyadenylate-binding protein 1 (PABC1) to stabilize the transcript, promoting PEG10 protein expression (Zhang, et al. 2021a). IGF2BP2 stabilized tuberous sclerosis complex 1 (TSC1) and peroxisome proliferator activated receptor-γ (PPARγ) contributing to mouse M2 macrophage formation (Wang, et al. 2021). In human colon cancer cells, IGF2BP3 stabilized and prevented degradation of CCND1 and VEGFA, increasing proliferation and angiogenesis (Yang, et al. 2020). Collectively, these studies established a role for IGF2BPs in mRNA stability and show that IGFBP1/2/3 bind to transcripts harboring the m6A mark and prevent mRNA degradation in human disease, including breast cancer as will be described below.
HNRNP m6A-binding proteins
The role of heterogeneous ribonucleoproteins (HNRNPs) in alternative splicing, mRNA packaging, and transport contributing to proteomic diversity in eukaryotes is well studied (Geuens, et al. 2016; Kedzierska and Piekielko-Witkowska 2017). HNRNPs were first identified and characterized in HeLa cells as A, B, and C groups comprising HNRNP particles that form on all nascent transcripts (Beyer, et al. 1977) and act as key proteins in mRNA biogenesis (Choi, et al. 1986; Osheim, et al. 1985). The major classes of HNRNPs each contain conserved domains that facilitate RNA-binding: the RRM, the quasi-RRM, an arginine-glycine-glycine (RGG) box, and a KH domain (Gorlach, et al. 1992; Siomi and Dreyfuss 1995; Swanson and Dreyfuss 1988). HNRNPs A/B are divided into four subgroups: HNRNP A1, A2/B1, A3, and A0, though the more highly expressed A1 and A2/B1 are the most studied for the role they play in mRNA translation (Park, et al. 2015), splicing (Mayeda and Krainer 1992), and trafficking (Shan, et al. 2000). Only in the past 10 years, have HNRNP proteins been associated with m6A-RNA mechanisms. In 2012, Tavazoie and others investigated how HNRNP proteins stabilize mammalian mRNA (Goodarzi, et al. 2012), and in 2015 they established a clear connection between HNRNPA2/B1 and m6A-RNA by showing that HNRNPA2/B1 recognizes m6A in mRNA in MDA-MB-231 cells to mediate alternative splicing (Alarcón et al. 2015a).
HNRNPA2/B1 was reported to bind to m6A on primary microRNA (pri-miRNAs) transcripts in MDA-MB-231 cells and interacts with the DROSHA complex to induce pri-miR to premature (pre)-miR processing (Alarcón et al. 2015b). We reported that HNRNPA2B1 was increased in endocrine-resistant ER+ breast cancer cells, played a role in sensitivity to tamoxifen and fulvestrant, and regulated miRNA expression (Klinge et al. 2019b; Petri, et al. 2021). This will be described in more detail below, HNRNPC contains only one RBD and forms self-oligomers for RNA binding (Swanson, et al. 1987). In 2015, HNRNPC was reported to bind to a secondary RNA structure that occurs when m6A remodels RNA to form truncated hairpins. This m6A-remodelled structure, also known as an m6A-switch, creates accessible U-tracts which significantly improves HNRNPC binding affinity and facilitates RNA splicing (Liu, et al. 2015). HNRNPG is another HNRNP that takes advantage of the m6A-switch mechanism by preferentially binding to target transcripts remodeled by m6A. Depletion of HNRNPG in HEK-293T cells altered the expression and splicing of hundreds of transcripts altered by METTL3 or METTL14 knockdown (Liu, et al. 2017a). Together, these reports validate the connection between m6A-remodelling and HNRNP m6A-readers in regulating mRNA metabolism.
Other m6A-binding proteins
Additional RBPs have been identified and characterized as m6A readers. High-efficiency diazirine-based RNA photo-crosslinking of m6A-mRNA in HeLa cells combined with LC-MS/MS proteomics identified novel m6A-RNA-protein interactions, including the leucine rich pentatricopeptide repeat containing (LRPPRC) and fragile X-messenger ribonucleoprotein 1 (FMR1) proteins as m6A-binders (Arguello, et al. 2017). Gene set enrichment analysis from GEO datasets of lung adenocarcinoma patients found that downregulation of LRPPRC was correlated with immune response activation, suggesting that LRPPRC may regulate the tumor environment (Li, et al. 2021b). FMR1 encodes FMRP, which contains an RGG domain that recognizes the m6A-modified RNA (Myrick, et al. 2015). FMRP associates with CDS of transcripts, stalling ribosomes and regulating translation at the elongation stage (Darnell, et al. 2011). Another RBP that recognizes m6A-RNA is staphylococcal nuclease-like (SN-like) domain-containing protein 1 (SND1), a member of the Tudor family of proteins that binds to methylated DNA. SND1 was first identified in Schizosaccharomyces pombe, but is conserved from yeast to humans (Chen, et al. 2011). Although, the exact mechanism of the m6A/SND1 axis is not understood, a pan-cancer analysis of TGCA datasets shows a correlation between increased SND1 expression and poor OS and RFS in many cancers, including breast cancer (Cui, et al. 2020). Other RBPs with roles in translation have been identified as possible m6A readers. For example, eukaryotic initiation factor 3 (eIF3), a component of the 43S preinitiation complex, interacts with YTHDF1 or YTHDF3 at m6A-modified RNA in the 5’-end of transcripts (Shi et al. 2017; Tarun, et al. 1997). However, binding of eIF3 to m6A in the 5’UTR of transcripts does not require YTHDF1, suggesting that eIF3 directly binds m6A (Meyer, et al. 2015). The identification of RBPs that recognize m6A and their roles in transcriptional-translational regulation require further investigation.
m6A readers in breast cancer
Dysregulation of m6A modification and the expression of m6A readers is associated with cancer progression (Bai, et al. 2019; Liu, et al. 2020b). Analysis of data from The Cancer Genome Atlas (TCGA) for YTHDF1 and YTHDF3 showed increased transcript expression of these readers in breast tumors compared to normal tissue that was correlated with reduced OS of breast cancer patients (Anita, et al. 2020). In a separate study, gene set enrichment analysis (GSEA) of microarray expression datasets from the TCGA database showed distinct enrichment of the WNT signaling pathway in patient samples with high YTHDF1 expression and knockdown of YTHDF1 in MCF-7 cells reduced the expression of WNT pathway proteins (Hu, et al. 2021). YTHDF1 and YTHDF3 were associated with lower OS and higher expression rates of genes related to c-Myc signaling and T cell differentiation in TCGA breast cancer data (Li, et al. 2022a). Despite evidence that YTH domain proteins play an important role in mRNA translation and stability in cancer, few studies have evaluated their specific role(s) in breast cancer (Table 3). The REPIC (RNA EPItransriptome Collection) database only lists one m6A-Seq/Me-RIP-seq data set from breast cancer cells (Liu, et al. 2020a). YTHDF1, with METTL14, promoted double stranded break (DSB) DNA damage repair and YTHDF1 knockdown increased sensitivity to adriamycin, cisplatin, and olaparib in MDA-MB-231 cells (Sun, et al. 2022). FOXM1, which promotes breast cancer metastasis, is an YTHDF1 target and binding of YTHDF1 to FOXM1 mRNA accelerated translation of FOXM1 in MCF-7 cells (Chen, et al. 2022). Binding of YTHDF1 to PKM2 mRNA stabilized the transcript to increase translation efficiency, which increased glycolytic activity in MBA-MD-231 and MCF-7 cells (Yao, et al. 2022).
Depletion of YTHDF2 increased apoptosis in MDA-MB-231 by increasing mitochondrial stress, proteotoxicity, endoplasmic reticulum stress (ERS), and the unfolded protein response (UPR) (Einstein, et al. 2021b). Interestingly, eCLIP for YTHDF identified unique YTHDF target genes in MDA-MB-231, MCF-7, and SKBR3 cells that overlapped with m6A sites identified by m6A-RIP-seq (Einstein et al. 2021b). shYTHDF2 upregulated transcript levels of genes promoting cell migration, wound healing, EMT, and metastasis in MDA-MB-231 cells (Einstein et al. 2021b). In summary, YTHDF2 was demonstrated to bind m6A peak-containing transcripts for degradation in MDA-MB-231 cells, thus ‘safeguarding’ the cells from ERS and proteotoxicity (Einstein et al. 2021b). By binding m6A, YTHDF2 stabilized ATF3 transcripts, thus increasing ATF3 protein, a transcription factor that was identified as a potential driver of TAM-resistance in TAM-resistant MCF-7 cells (Liu, et al. 2021). A study evaluating TNBC tissue samples identified an association between YTHDF3 expression, lymph node metastasis, and higher histological grade (Lin, et al. 2022). YTHDF3 was reported to increase the binding of eIF3 to ST6GALNAC5, GJA1, EGFR, and VEGFA mRNAs in MDA-MB-231 cells, suggesting YTHDF3-stimulated translation of key genes that drive breast cancer brain metastasis (Chang, et al. 2020). YTHDC1 overexpression in MDA-MB-231 cells promoted DNA replication and DNA damage repair and induced resistance to the adriamycin (Zhou, et al. 2021). Taken together, these studies suggest that the YTH domain family of m6A readers plays a dynamic role in altering the expression of genes in pathways associated with breast cancer progression and metastasis.
MYC is an important oncogenic transcription factor in TNBC (Fallah, et al. 2017) and MYC mRNA has been reported as a downstream target of IGF2BP proteins (Huang et al. 2018) as well as YTHDF1 (Zheng, et al. 2022). Binding of IGF2BPs by FGF13-ASI, a long non-coding RNA (lncRNA) that decreases glycolysis and stemness in MDA-MB-468 cells and prevents MYC mRNA stabilization by IGF2BPs (Ma, et al. 2019). IGF2BPs also interact with circCD44 to stabilize MYC mRNA in BT549 cells (Li, et al. 2021a). Further evidence for IGF2BP regulation of MYC mRNA in breast cancer is listed in Table 3.
IGF2BP1 bound directly to m6A sites on MIR210HG, an oncogenic lncRNA that promotes proliferation and EMT in MCF-7 cells (Shi, et al. 2022b). Similar associations between IGF2BP1 and other lncRNAs with roles in breast tumor progression have been established, e.g., upregulation of LINC00483, an oncogenic lncRNA associated with advanced tumor stage (Qiao, et al. 2021). IGF2BP2 stabilized DROSHA mRNA for translation in MDA-MB-231 cells and DROSHA induced the expression of genes promoting sphere formation and cell stemness, i.e. STC1, GRB10, and SLCo4A1 (Peng et al. 2021). METTL3 and IGF2BP3-mediated m6A modification that increased PD-L1, a cell surface immune checkpoint inhibitor, in MDA-MB-231 and HCC38 cells (Wan, et al. 2022). These studies characterizing IGF2BPs and their interaction with m6A in target transcripts suggest lncRNA-epitranscriptomic regulation of breast cancer progression and drug-resistance.
The role of HNRNPs in breast cancer progression and drug-resistance is being actively investigated (An, et al. 2021; Klinge, et al. 2019a; Li, et al. 2021a; Petri et al. 2021). There are 16 HNRNP family members named alphabetically A-U, classified by their RNA binding domains and have diverse structural features and functions (reviewed in (Geuens et al. 2016)). HNRNPA2B1 expression is higher in breast tumors compared to normal breast tissue (Liu, et al. 2019b; Ma, et al. 2020) and regulates stability, alternative splicing, DNA repair, and telomere maintenance (Liu and Shi 2021). HNRNPA2B1 plays a role in the processing of selected pri-miRNAs to pre-miRNAs in HEK-293T cells (Alarcón et al. 2015b). HNRNPA2B1 stimulated miRNA processing in MDA-MB-231 cells by recognizing m6A on pri-miRNAs and interacting with the DROSHA complex protein DGCR8, to increase pri-miRNA processing to pre-miRNAs (Alarcón et al. 2015b). However, some studies suggest that HRNPA2B1 may also interact with negative regulators of miRNA biogenesis and suppress miRNA processing (Suster and Feng 2021). Analysis of the miRome in MCF-7 cells with overexpressed HNRNPA2B1 found up- and down-regulation of hundreds of miRNAs and identified dysregulated pathways associated with breast cancer progression, metastatic spread, and TAM-resistance (Klinge et al. 2019a). Knockdown of HNRNPA2B1 inhibited proliferation in MCF-7 and MDA-MB-231 breast cancer cells by downregulating signal transducer and activator of transcription 3 (STAT3) and extra-cellular-signal-regulated kinase 1/2 (ERK1/2) (Hu, et al. 2017). In addition, overexpression of HNRNPA2B1 increased endocrine resistance, cell motility, and stem cell properties in MCF-7 cells whereas knockdown of HNRNPA2B1 restored TAM and fulvestrant sensitivity to endocrine resistant LCC9 and LY2 breast cancer cells (Petri et al. 2021). Investigating other HNRNPs has revealed novel m6A regulatory mechanisms in breast cancer. For example, HNRNPC expression promoted cell proliferation by inducing MAPK signaling in MDA-MB-231 and MCF-7 cells (Lv, et al. 2021). HNRNPK promotes invasion and metastasis in MDA-MB-231 and MDA-MB-468 cells by interacting with PROX1 and stimulating the WNT pathway (Zhu, et al. 2022).
Emerging roles have been identified for other m6A-associated RBPs in breast cancer progression. FMR1 and LRPPC are over-expressed in breast cancer tissue data from TCGA compared to normal tissue (Zhang et al. 2020). Additionally, GTEx data revealed a higher expression of LRPPC in breast tissue (Zhang et al. 2020). SND1 is upregulated in breast primary invasive ductal carcinomas and induced RhoA degradation, thus disrupting F-actin cytoskeletal organization, by promoting expression of Smurf1, an E3 ubiquitin ligase (Yu, et al. 2015). The role of eIF3 subunits in breast cancer tumorigenesis has been well-documented (Fan and Guo 2015; Han, et al. 2020; Morris, et al. 2021; Zheng, et al. 2016) and there is clear evidence for an m6A-eIF3 interaction (Meyer et al. 2015). Further studies investigating RBPs involved in breast cancer progression, drug-resistance, and metastasis are necessary to define their role in m6A-mediated RNA metabolism in breast cancer.
m6A Erasers in Breast Cancer
ALKBH5 and FTO are members of a conserved family of AlkB non-hem Fe(II)/2-oxoglutarate (2OG) dependent dioxygenases with nine ALKBH homologues with diverse substrates and specificities (reviewed in (Rajecka, et al. 2019)). Knockout phenotypes for ALKBH5 and FTO are divergent: infertility and impaired spermatogenesis versus growth retardation, lean phenotype and impaired adipogenesis, respectively (reviewed in (Rajecka et al. 2019)) ALKBH5 and FTO were described as the two mammalian non-heme Fe2+ and α-ketoglutarate (αKG)–dependent dioxygenases that act as m6A mRNA demethylases that catalyze the oxidation of m6A in a two-step reaction that require O2 and generates CO2, succinate, and Fe(III) and ultimately yields adenosine and formaldehyde (reviewed in (Meyer and Jaffrey 2014; Purslow, et al. 2021)).
ALKBH5
There is evidence that ALKBH5 is the specific m6A-demethylase whereas FTO is the m6Am demethylase (Mauer and Jaffrey 2018; Mauer, et al. 2017; Mauer et al. 2019). Hypoxia (1% O2) increased ALKBH expression in MCF-7 luminal A and in MDA-MB-231, SUM159, and MDA-MB-435 TNBC cell lines in a HIF-1α- and HIF-2α- dependent manner and reduced m6A methylation in total RNA (Zhang, et al. 2016). Further experiments in MCF-7 and MDA-MB-231 cells revealed that ALKBH5 demethylated m6A in NANOG mRNA which increased total NANOG mRNA transcript levels and enriched BCSC populations in these cell lines (Zhang et al. 2016). NANOG is a pluripotency factor that promotes breast cancer tumorigenesis and metastasis (Lu, et al. 2014). Knockdown of ALKBH5 in MDA-MB-231 cells reduced mammary tumor growth in vivo (Zhang et al. 2016). Immortalization of primary human mammary epithelial cells (HMECs) by stable expression of hTERT, P53DD, Cyclin D1, CDK4R24C, or C-MYCT58A increased FTO and ALKBH5 while decreasing METTL3 protein expression (Fry, et al. 2018). siALKBH5 increased overall m6A levels and led to reduced long-term viability, migration, and invasion of MDA-MB-231 cancer cells (Panneerdoss et al. 2018). Doxorubicin treatment increased m6A levels in breast tumor patient samples but overexpression of PRMT5 (Protein arginine methyltransferase 5) expression in MDA-MB-231 breast cancer cells decreased m6A levels and decreased m6A methylation of BRCA1 to increase BRCA1 stability by increasing ALKBH5 nuclear translocation through an interaction with ALKBH7 (Wu, et al. 2022). The drug tadalafil was recently demonstrated to inhibit PRMT5 methyltransferase activity and reduce ALKBH5 nuclear localization (Wu et al. 2022). Tadalafil increased total RNA m6A methylation and specific m6A methylation on the 3’-UTR of BRCA1 and decreased BRCA1 mRNA stability and protein expression, enhancing the doxorubicin-induced MDA-MB-231 cell apoptosis (Wu et al. 2022). These findings suggest that blocking nuclear localization of ALKBH5 may enhance chemotherapeutic responses in breast cancer.
FTO
FTO was originally identified in genome-wide association studies (GWAS) as having a modest, but significant, contribution to childhood and adult obesity (Blakemore and Froguel 2008). FTO was reported to share motifs with Fe(II)- and 2-oxoglutarate-dependent oxygenases and showed high mRNA expression in the brain, notably in the hypothalamic nuclei and in the arcuate nucleus that regulates satiety and feeding behaviors (Church, et al. 2009). FTO was identified as an m6A demethylase in vitro and in HeLa and HEK-293T cell studies (Jia, et al. 2011). Case-control analysis of 354 breast tumors and 364 controls revealed that three single nucleotide polymorphisms (SNPs) in intron 1 of FTO were associated with breast cancer risk (Kaklamani, et al. 2011). Another study found an association with an intron 1 SNP in FTO (rs9939609) with breast cancer risk in a large study (62,328 breast cancer cases and 83,817 controls) of women of European ancestry (Zhao, et al. 2016). In women of African ancestry, FTO SNP rs17817449 was associated with both overall breast cancer and ER+ disease (Feng, et al. 2017). Reduced FTO transcript expression was associated with TAM-resistance and early distant relapse time in an analysis of two datasets (GSE26971, GSE58644) (Zhou, et al. 2018).
FTO localizes to nuclear speckles in HeLa cells (Jia et al. 2011) and MCF-7 cells and was shown to affect RNA modifications including the 3-methyluridine/uridine and pseudouridine/uridine ratios in total brain RNA (Berulava, et al. 2013). Transgenic mice with a CRISPR/Cas9 engineered mutation modeling the human FTO-R316Q mutation (mouse FTO-R313A) resulting in a catalytically inactive FTO protein showed reduced bone mineral density and content, reduced body length and mass and reduced viability (Sachse, et al. 2018). Bovine milk consumption was suggested to be an epigenetic regulator of FTO transcription via milk exosomes containing bovine miR-29s that downregulate human cell DNA methyltransferases (DNMTs), thus reducing CpG island DNA methylation in the FTO gene and increasing FTO mRNA and protein (Melnik 2015). Persistent consumption of cow’s milk is associated with hepatocellular cancer, luminal A breast cancer, prostate cancer, and Parkinson’s disease by milk exosomal regulation of mTORC1 signaling (Melnik 2021).
Conclusions as to whether FTO is increased or decreased in breast tumors compared to normal breast depend on the type of study (transcripts versus protein) and publication (Table 4 and below). FTO transcript expression was higher in MCF-7 compared to MDA-MB-231 and HCC1937 TNBC cell lines (Liu, et al. 2017b). In silico examination of FTO transcript expression (GSE9014) found FTO upregulated in breast tumors and protein was higher in IHC staining of three breast tumors vs. normal breast, correspondingly, an m6A dot blot showed lower m6A levels in breast tumors compared to normal breast (Niu, et al. 2019). Likewise examination of Oncomine data identified increased FTO and ALKBH5 in breast tumors (Wu et al. 2019). However, neither FTO nor ALKBH5 transcript expression was associated with OS in the TCGA breast cancer dataset (Chang et al. 2020). Analysis of breast cancer and normal breast tissues in the RNA sequencing expression data in GEPIA found lower FTO but no difference in ALKBH5 in breast tumors (Shi, et al. 2020). However, the distance-metastasis-free survival (DMFS) of the FTO high-expression TNBC patients was shorter than that of the FTO low-expression group, indicating that FTO is a risk factor for DMFS in TNBC.
Table 4:
m6A Erasers ALKBH5 and FTO in breast cancer
| Protein (other names) | Function | Breast cancer |
|---|---|---|
| ALKBH5 (AlkB Homolog 5, RNA Demethylase) | Dioxygenase that specifically demethylates m6A (Zheng, et al. 2013). ALKBH5 is mainly nuclear and is specifically the m6A demethylase (Mauer et al. 2017). |
• IHC staining showed increased ALKBH5 in breast cancer tissues compared to normal breast tissue (Liu et al. 2019b). • ER+ or PR+ status was associated with high mRNA levels of ALKBH5 (Wu et al. 2019). • Transcript expression was down-regulated in breast cancer compared to adjacent normal breast (Zhang et al. 2020). • No difference in transcript expression between breast tumors and normal breast tissue (Shi et al. 2020; Zhao et al. 2021). • Copy number loss in breast tumors (Yang et al. 2021). No difference in copy number variations or somatic mutations of ALKBH5 in breast cancer (Zhao et al. 2021). |
| FTO (fat mass and obesity associated gene (FTO) Alpha-Ketoglutarate Dependent Dioxygenase) | RNA demethylase that mediates oxidative demethylation of different RNA species, such as mRNAs, tRNAs and snRNAs, and acts as a regulator of fat mass, adipogenesis and energy homeostasis (Jia et al. 2011). FTO targets m6Am (Mauer and Jaffrey 2018; Mauer et al. 2017; Mauer et al. 2019). FTO also demethylates 3-meT in ssDNA and 3-meU in ssRNA (reviewed in (Chen and Du 2019)).FTO demethylates m6A, m6Am, and m1A in the nucleus and cytoplasm (Wei et al. 2018). | • IHC showed FTO staining was both nuclear and cytoplasmic and higher in breast cancer than adjacent normal tissue and the percent of FTO –positive expression was higher in TNBC and HER2+ than ER+/PR+ breast cancer (Tan, et al. 2015). • Another study found reduced FTO IHC staining in breast carcinoma tissues compared to normal breast tissue (Liu et al. 2019b). • Lower transcript expression in breast cancer tissues compared to normal tissue (Zhang et al. 2020; Zhao et al. 2021). |
High FTO transcript abundance was associated with ER-/PR-/HER2+ and higher grade (II and III) breast tumors and with lower OS (Niu et al. 2019; Xu, et al. 2020). SKBR3 and MDA-MB-453 ER-/PR-/HER2+ breast cancer cells had higher FTO than MCF-10A, MCF-7, T47D, BT474, MDA-MB-231, or BT549 breast cancer cells (Xu et al. 2020). Lentiviral-mediated stable knockdown of FTO in MCF-7 and MDA-MB-231 cells inhibited cell proliferation, colony formation, and reduced cell apoptosis (Niu et al. 2019). Likewise, shFTO inhibited 4T1 mouse mammary tumor growth in vivo (Niu et al. 2019). RNA-seq in shFTO-MDA-MB-231 cells identified BNIP3, an apoptosis inhibitor, as upregulated by FTO knockdown in an m6A-modification-dependent manner (Niu et al. 2019). Thus, the authors concluded that increased FTO reduced m6A in the BNIP3 mRNA resulting in lower BNIP3 protein and increased breast cancer cell proliferation (Niu et al. 2019). Similarly, knockdown of FTO by siRNA in SKBR3 and MDA-MB-453 cells inhibited cell migration and invasion, although not cell proliferation. Overexpression of FTO in SKBR3 and MDA-MB-453 cells stimulated cell migration and invasion and decreased total m6A abundance (Xu et al. 2020). RNA-seq in FTO-knockdown SKBR3 cells identified miR-181b-3p as upregulated and ARL5B (ADP Ribosylation Factor Like GTPase 5B) as a direct target of miR-181b-3p, although a direct role for FTO removal of m6A in downregulating miR-181b-3p, thus increasing ARL5B in breast cancer, was not proven (Xu et al. 2020).
FTO was reduced while METTL3 and m6A levels were increased in “Lung metastasis breast cancer cells” MDA-MB-231LMF3 and BT-549LMF3 which are TNBC cell lines established by three rounds of tail vein injection and “lung metastasized cell” re-injection (Chen, et al. 2021a). However, chemical inhibitors of FTO (CS1 and CS2 at 0.1 and 1 µM) inhibited leukemia stem/initiating cell self-renewal and MDA-MB-231 cell proliferation in vitro and as tumor xenografts in vivo (Su, et al. 2020). While this finding suggests that inhibition of FTO may inhibit breast cancer metastasis, the study of the lung metastatic MDA-MB-231 cells suggest caution. More research is needed on FTO’s mechanism and role in metastatic disease.
m6A in mRNA and its detection
m6A modification of mRNA regulates RNA splicing, stability, translation, and intracellular localization and plays a role in many human diseases including cancers (breast, lung, hepatocellular, renal, prostate, gastric, bladder, and colorectal) and other diseases, e.g., osteoporosis and type II diabetes (reviewed in (Destefanis, et al. 2021; Lan, et al. 2021)). The number of m6A in transcripts is variable, some mRNAs can contain 20 m6A sites, generally found in genes that regulate development and cell fate (Zaccara et al. 2019).
The most common methods to detect m6A in mRNA were recently reviewed (Moshitch-Moshkovitz, et al. 2022; Zaccara et al. 2019). These include m6A-RNA immunoprecipitation (RIP)-seq (called meRIP-seq, m6A-RNA-seq, or m6A-seq), in which an m6A antibody is used to identify bound transcripts by Illumina Seq; m6A- cross-linking and immunoprecipitation (CLIP)-seq which employs UV cross-linking to identify m6A sites at higher specificity compared to m6A-seq; miCLIP (modified CLIP) that relies on inducing specific mutations (C to T conversions) during reverse transcription (Roberts, et al. 2021), LC-MS (Thüring, et al. 2016) and deamination adjacent to RNA modification targets (DART-seq) that uses a fusion protein consisting of the m6A-binding YTH domain tethered to the cytidine deaminase APOBEC1 to direct C-to-U editing at cytidine residues that invariably follow m6A sites (Meyer 2019; Tegowski, et al. 2022). Direct RNA sequencing by Oxford nanopore-based single molecule sequencing, which maintains and directly detects nucleic acid modifications as a single strand of RNA passes through a pore (Kono and Arakawa 2019; Parker, et al. 2020), without the need for added reagents or processing, i.e. antibodies specific for m6A for RNA immunoprecipitation (RIP)-seq or cDNA generation which introduce biases and only localizes the m6A mark within ~ 100 nt, rather than the precise position of the m6A (Pratanwanich, et al. 2021). A computational method (xPore) was demonstrated to identify m6A positions at single base resolution, within the DRACH/RRACH motif (Jenjaroenpun, et al. 2020), from nanopore reads (Pratanwanich et al. 2021).The nascent RNA m6A methylome was also identified by methylation inscribed Nascent Transcripts Sequencing (MINT-Seq) to capture nascent RNAs RNA metabolic labeling used in TT-Seq combined with m6A-RIP-seq (Lee, et al. 2021). An m6A-Atlas of the m6A epitranscriptome includes m6A sites in human, mouse, rat and other species, as well as 46 human cell lines (Tang, et al. 2021). Using the m6A-Atlas, m6A, m1A, m5C, m6Am, m7G, and Ψ modifications were identified in 2,294 gene transcripts involved in pharmacokinetic and pharmacodynamics, e.g., drug metabolizing enzymes and transporters, receptors, and targets (Liu, et al. 2022b). Based on their analysis, the authors concluded that m6A is a biomarker for anti-tumor drugs in TNBC (Liu et al. 2022b).
Roles for m6A mRNA modification in cellular functions
As indicated above, the presence and position of m6A in mRNA transcripts regulates stability, splicing, and intracellular location by the association of reader proteins with the m6A mark. Recently, YTHDC1 and unknown m6A readers recognized m6A and recruit KDM3B and other demethylases of H3K9me2, to the chromatin regions around the transcription start site of CYP2B6 in Huh-7 and HepaRG hepatoma cells, resulting in increased transcription (Isono, et al. 2022). This suggests that protein interactions between chromatin modifiers and m6A readers are a potential mechanism for transcript in initiation as well as transcript stability and splicing. m6A is involved in the maintenance of the “stemness properties” of cancer stem cells and the interaction between CSC and the tumor immune microenvironment (TIME) (reviewed in (Zhang, et al. 2021d)). m6A modification of MYC mRNA regulates transcript stability by binding YTHDF1 to increase translation (Zheng et al. 2022). Interestingly, m6A modification on mRNA accumulates within 2 min. at sites of DNA damage in U2OS cells generated by global UVC irradiation, allowing recruitment of DNA polymerase kappa (Pol κ) (Xiang, et al. 2017). Tonicity-responsive enhancer binding protein (TonEBP) was demonstrated to recruit METTL3 to R-loops, three-stranded DNA-RNA hybrids with ssDNA, by to methylate m6A and stimulates homologous recombination (HR) repair in YTHDC1 dependent manner at DNA double-stranded breaks (Zhang et al. 2020). There are few studies of how transcript-specific m6A affects gene expression or pathways in breast cancer. A recent study reported increased expression of ALKBH5 in HER2+ SKBR3 and BT474 breast cancer cell lines that are trastuzumab- and lapatinib-resistant and in breast tumors from patients who responded poorly to these treatments (Liu, et al. 2022a). m6A-RIP seq identified m6A peaks in the SKBR3 and BT474 and their HER2-therapy resistant derivatives and correlated increased ALKBH5 with increased m6A demethylation and expression of EGFR, FOXO1, and GLUT4 in the resistant cells. RIP-qPCR identified YTHDF2 –m6A interaction stabilized the GLUT4 mRNA transcript. m6A demethylation of GLUT4 by the increase in ALKBH5 in the resistant cell lines resulted in increased glycolysis and Ritonavir, a FDA-approved HIV protease inhibitor that also inhibits GLUT4, restored sensitivity to trastuzumab in the resistant cells (Liu et al. 2022a).
Identification of m6A sites in genes in breast cancer cells
There are relatively few studies identifying m6A sites in genes in breast cancer cell lines and few cell lines have been studied (Table 5). Most studies have used some form of m6A-RIP-seq to identify m6A peaks and a few have knocked down METTL14 or METTL3 as a control to verify m6A identification. In MDA-MB-231 TNBC cells, 15,981 and 17,312 m6A peaks from 6,796 and 7,194 m6A-containing transcripts in control and METTL14-silenced MDA-MB-231 cells, respectively (Panneerdoss et al. 2018). METTL14 target genes including TGFβ1, SMAD3, cyclin E1, cyclin D1, MMP9, VEGFA, and HMGA2 were found to be hyper m6A methylated and showed decreased expression in METTL14 KD cells, suggesting that increased m6A methylation may increase the stability of these transcripts in MDA-MB-231 cells (Panneerdoss et al. 2018). Another study in MDA-MB-231 cells knocked down either METTL3 or the reader IGF2BP2 to identify specific m6A peaks by m6A-RIP-seq and reported that most m6A peaks were identified in the CDS (Wan et al. 2022). Epitranscriptomic microarray and meRIP-seq identified 30 sequences from 24 genes that harbored both hypo-methylation and down-regulated m6A peaks after shMETTL3 in MDA-MB-231 cells including PD-L1 (CD274). IGF2BP2 reduced the CD274 transcript and PD-L1 protein, suggesting that m6A deposition and binding by IGFBP2 enhance transcript stability, and m6A-RIP-qPCR identified binding of IGF2BP3 to CD274 mRNA in an m6A-dependent manner (Wan et al. 2022). In MCF-7 cells, knockdown of METTL3 and METTL14 increased ERα and YTHDC1 protein levels (Lee et al. 2021), which can have secondary effects on measured outcomes. m6A-enchanced cross-linking and immunoprecipitation (m6A-eCLIP) was developed to specifically identify m6A sites in MCF-7 and MDA-MB-231 cells, although the authors did not describe the m6A-containing genes nor compare the two cell lines (Roberts et al. 2021). Secondary lung metastatic MDA-MB-231-LM2 tumor cells showed more m6A peaks (14,873 peaks) and m6A-peak modified genes (4,512 genes) that were unique from the parental MDA-MB-231 cell line; however, MYC-induction of HMECs did not alter m6A peaks (Einstein, et al. 2021a). Unique and common m6A peaks were identified when comparing MDA-MB-231 vs. MCF-7 or SKBR3 cells and between MCF-7 and SKBR3 cells; however, the genes containing different numbers of m6A peaks in the different cell lines were not discussed (Einstein et al. 2021a). Direct RNA seq using nanopore and xPore identified m6A sites in transcripts from HEK-293T cells and compared those in transcripts from cancer cell lines: MCF-7, K562 (leukemia), HCT116 (colon cancer), A549 (non-small cell lung cancer), and HepG2 (hepatocellular) (Pratanwanich et al. 2021). The number of m6A sites was lower in MCF-7 cells compared to the other cell lines; however, because the paper was focused on the computational analysis, the authors did not discuss which transcripts showed reduced m6A, nor interpret their data relative to breast tumor samples.
Table 5:
Detection of m6A peaks in genes in breast cancer cells.
| Method of m6A-RIP-seq | Breast cancer dell line(s) | Other treatments | Comments, reference and data availability |
|---|---|---|---|
| Magna MeRIP™ m6A kit (Millipore) Illumina | MDA-MB-231 | siMETTL14 | GEO database GSE81164 (Panneerdoss et al. 2018) |
| ONT Nanopore direct RNA-seq with xPore computational analysis | MCF-7 with two replicates | The data are available through the ENA (PRJEB40872) (Pratanwanich et al. 2021). | |
| m6A-enchanced cross-linking and immunoprecipitation (m6A-eCLIP) | MCF-7 and MDA-MB-231 Performed 3 biological replicates/cell line |
Observed divergent numbers of m6A peaks in the three replicates: GEO database GSE147440 (Roberts et al. 2021). | |
| Magna MeRIP™ m6A kit (Millipore) Illumina HiSeq 2000 system | BT-549 and BT-549LMF3 cells | m6A-seq analysis identified 47, 539 and 38, 383 m6A peaks from 6,284 to 5,373 m6A-modified transcripts in BT-549 and BT-549LMF3 cells, respectively (Chen et al. 2021a). KRT7 and KRT7-AS showed increased expression, m6A deposition, and increased transcript stability by binding IGF2BP1/HuR and increased interaction of YTHDF1/eEF-1 with KRT7 mRNA to increase elongation in BT-549LMF3 cells (Chen et al. 2021a) | |
| M6A-RIP-seq (Mouse mAb anti-m6A (Synaptic Systems; 202 011)-Illumina HiSeq4000 | MYC-induced human mammary epithelial cells (HMECs); MDA-MB-231, MDA-MB-231-LM2, MCF-7, SKBR3 cells in biological duplicates | Unique and common m6A peaks were identified when comparing MDA-MB-231 vs. MCF-7 or SKBR3 cells and between MCF-7 and SKBR3 cells (Einstein et al. 2021a). GEO: GSE137258 | |
| Magna MeRIP™ m6A kit (Millipore) Illumina HiSeq X Ten platform | MDA-MB-231 | +/− shMETTL3 or IGF2BP2 | No information about data availability (Wan et al. 2022). . |
Using MINT-seq, ~ 3–5 times more m6A peaks were identified in the nascent transcripts of MCF-7 cells compared to MeRIP-seq (Lee et al. 2021). The authors reported pervasive m6A signals on nascent RNAs in MCF-7 cells, including 2,207 enhancer RNAs (eRNAs) and 1,201 upstream antisense RNA (uaRNAs) (eRNAs and uaRNAs are non-coding RNAs whose expression is correlated with the activity of functional enhancers by modifying chromatin accessibility at promoters, stabilizing enhancer-promoter interactions, transcription factor-DNA interactions, and RNA Pol II stability (reviewed in (Cardiello, et al. 2020; Kim, et al. 2015; Li, et al. 2016a)). Approximately 18.8% of putative active eRNAs contained one or more m6A peaks (Lee et al. 2021). The m6A-marked transcripts had higher abundance, suggesting the m6A increased eRNA stability. Estradiol (E2, 100 nM for 24 h) did not significantly alter m6A peak levels in the 226 eRNAs induced by E2 in MCF-7 cells (Lee et al. 2021). The authors concluded that m6A modification is “largely hard wired to nascent sequences based on sequences, e.g., the RRACH motif” (Lee et al. 2021), as originally proposed (Darnell, et al. 2018). The RRACH motif f (R = A, G; H = A, C, U) (Jenjaroenpun et al. 2020) is recognized by the METTL3 complex (Knuckles and Bühler 2018; Licht and Jantsch 2016).
Conclusions and future directions
Abundant data supports the role of epitranscriptomic modification of mRNA and the readers, writers, and erasers of m6A in various diseases and cancers, including breast cancer. The number of publications on m6A in breast cancer is doubling each year (Figure 2) with 64 papers in 2022 at the time of submission of this review. Whether m6A is “hard wired” by the RRACH motif (Darnell et al. 2018) or modified depending on regulation of the METTL3 complex, other RNA binding proteins, RNA structure, intranuclear localization, e.g., condensates, and the epigenome remain to be examined in a cell- and tissue-type manner in breast cancer. How the “expososome”, including the microbiome, environmental chemical exposures, and endocrine- and metabolism- disrupting chemicals (Koual, et al. 2020; Koval, et al. 2022), affect these events to regulate the epitranscriptome in breast cancer is unknown. The advent of direct RNA sequencing and single cell RNA sequencing promises to yield new understanding of epitranscriptomics not only of m6A but less abundant modifications in breast cancer progression, endocrine therapy resistance, and metastasis. For example, identifying specific sites of m6A that impact on transcript stability and reader binding will be important in determining the role of m6A on endocrine-resistance in breast cancer as seen for HER2 resistance and GLUT4 expression (Liu et al. 2022a). Future studies to determine the mechanism by which gene- and position- specific m6A sites affects transcript stability and protein abundance, may employ inducible Crispr/dCAs13b-METTL3 and Crispr/dCas13b-ALKBH5 programmed to respectively methylate and demethylate specific m6A sites on a transcript (Li, et al. 2020; Shi, et al. 2022a; Zhang, et al. 2021c). The regulation and selectivity, role of specific cell types within a breast tumor and the tumor microenvironment, nuclear-mitochondrial or retrograde signaling of epitranscriptomic regulation of mRNA transcript processing and pathway regulation remains to be elucidated.
Figure 2: Number of publications on m6A AND breast cancer in PubMed by year.

Figure 1: Readers, writers, and erasers of m6A in mRNA regulate transcript fate.

Shown is the METTL3 m6A methyltransferase (writer) complex based on (Bawankar, et al. 2021). Readers are shown by their predominant subcellular location and roles in mRNA events, e.g., splicing, pri-miR-to-pre-miR-processing, nuclear export, mRNA stabilization or decay, translation initiation, elongation, or silencing. ALKBH5 is a specific m6A demethylase whereas there is controversy as to FTO’s specificity for m6A demethylation as described in the text. Created with BioRender.
Funding
National Institutes of Health P30ES030283, R21ES031510–01, ES031510–01S1. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. This work was supported in part by a grant from the Jewish Heritage Fund for Excellence Research Enhancement Grant Program at the University of Louisville School of Medicine.
Footnotes
Declaration of interest: The authors declare no conflict of interest.
References
- Abdullah C, Korkaya H, Iizuka S & Courtneidge SA 2018. SRC Increases MYC mRNA Expression in Estrogen Receptor-Positive Breast Cancer via mRNA Stabilization and Inhibition of p53 Function. Molecular and Cellular Biology 38 e00463–00417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adachi H, De Zoysa MD & Yu Y-T 2019 Post-transcriptional pseudouridylation in mRNA as well as in some major types of noncoding RNAs. Biochimica et biophysica acta. Gene regulatory mechanisms 1862 230–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akhtar J, Renaud Y, Albrecht S, Ghavi-Helm Y, Roignant J-Y, Silies M & Junion G 2021. m6A RNA methylation regulates promoter- proximal pausing of RNA polymerase II. Molecular Cell 81 3356–3367.e3356. [DOI] [PubMed] [Google Scholar]
- Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S & Tavazoie SF 2015a. HNRNPA2B1 Is a Mediator of m6A-Dependent Nuclear RNA Processing Events. Cell 162 1299–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alarcón CR, Lee H, Goodarzi H, Halberg N & Tavazoie SF 2015b. N6-methyladenosine marks primary microRNAs for processing. Nature 519 482–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- An J, Luo Z, An W, Cao D, Ma J & Liu Z 2021. Identification of spliceosome components pivotal to breast cancer survival. RNA Biol 18 833–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- An S, Huang W, Huang X, Cun Y, Cheng W, Sun X, Ren Z, Chen Y, Chen W & Wang J 2020. Integrative network analysis identifies cell-specific trans regulators of m6A. Nucleic Acids Research 48 1715–1729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anita R, Paramasivam A, Priyadharsini JV & Chitra S 2020. 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. [PMC free article] [PubMed] [Google Scholar]
- Anreiter I, Mir Q, Simpson JT, Janga SC & Soller M 2021. New Twists in Detecting mRNA Modification Dynamics. Trends in Biotechnology 39 72–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arguello AE, DeLiberto AN & Kleiner RE 2017. RNA Chemical Proteomics Reveals the N(6)-Methyladenosine (m(6)A)-Regulated Protein-RNA Interactome. J Am Chem Soc 139 17249–17252. [DOI] [PubMed] [Google Scholar]
- Bai Y, Yang C, Wu R, Huang L, Song S, Li W, Yan P, Lin C, Li D & Zhang Y 2019. YTHDF1 Regulates Tumorigenicity and Cancer Stem Cell-Like Activity in Human Colorectal Carcinoma. Front Oncol 9 332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balacco DL & Soller M 2019. The m6A Writer: Rise of a Machine for Growing Tasks. Biochemistry 58 363–378. [DOI] [PubMed] [Google Scholar]
- Bardia A, Harnden K, Mauro L, Pennisi A, Armitage M & Soliman H 2022. Clinical Practices and Institutional Protocols on Prophylaxis, Monitoring, and Management of Selected Adverse Events Associated with Trastuzumab Deruxtecan. The Oncologist oyac107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Batista PJ, Molinie B, Wang J, Qu K, Zhang J, Li L, Bouley DM, Lujan E, Haddad B, Daneshvar K, et al. 2014. m(6)A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell 15 707–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bawankar P, Lence T, Paolantoni C, Haussmann IU, Kazlauskiene M, Jacob D, Heidelberger JB, Richter FM, Nallasivan MP, Morin V, et al. 2021. Hakai is required for stabilization of core components of the m6A mRNA methylation machinery. Nature Communications 12 3778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bell JL, Wachter K, Muhleck B, Pazaitis N, Kohn M, Lederer M & Huttelmaier S 2013. Insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs): post-transcriptional drivers of cancer progression? Cellular and Molecular Life Sciences 70 2657–2675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berulava T, Ziehe M, Klein-Hitpass L, Mladenov E, Thomale J, Rüther U & Horsthemke B 2013. FTO levels affect RNA modification and the transcriptome. European journal of human genetics : EJHG 21 317–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beyer AL, Christensen ME, Walker BW & LeStourgeon WM 1977. Identification and characterization of the packaging proteins of core 40S hnRNP particles. Cell 11 127–138. [DOI] [PubMed] [Google Scholar]
- Bi Z, Liu Y, Zhao Y, Yao Y, Wu R, Liu Q, Wang Y & Wang X 2019. A dynamic reversible RNA N6-methyladenosine modification: current status and perspectives. Journal of Cellular Physiology 234 7948–7956. [DOI] [PubMed] [Google Scholar]
- Bianchini G, Balko JM, Mayer IA, Sanders ME & Gianni L 2016. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol 13 674–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blakemore AIF & Froguel P 2008. Is Obesity Our Genetic Legacy? Journal of Clinical Endocrinology and Metabolism 93 s51–56. [DOI] [PubMed] [Google Scholar]
- Boccaletto P & Bagiński B 2021. MODOMICS: An Operational Guide to the Use of the RNA Modification Pathways Database. Methods in Molecular Biology 2284 481–505. [DOI] [PubMed] [Google Scholar]
- Boccaletto P, Stefaniak F, Ray A, Cappannini A, Mukherjee S, Purta E, Kurkowska M, Shirvanizadeh N, Destefanis E, Groza P, et al. 2022. MODOMICS: a database of RNA modification pathways. 2021 update. Nucleic Acids Research 50 D231–D235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Borchardt EK, Martinez NM & Gilbert WV 2020. Regulation and Function of RNA Pseudouridylation in Human Cells. Annual Review of Genetics 54 309–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burstein HJ, Lacchetti C, Anderson H, Buchholz TA, Davidson NE, Gelmon KE, Giordano SH, Hudis CA, Solky AJ, Stearns V, et al. 2016. Adjuvant Endocrine Therapy for Women With Hormone Receptor–Positive Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline Update on Ovarian Suppression. Journal of Clinical Oncology 34 1689–1701. [DOI] [PubMed] [Google Scholar]
- Burstein HJ, Prestrud AA, Seidenfeld J, Anderson H, Buchholz TA, Davidson NE, Gelmon KE, Giordano SH, Hudis CA, Malin J, et al. 2010 American Society of Clinical Oncology Clinical Practice Guideline: Update on Adjuvant Endocrine Therapy for Women With Hormone Receptor-Positive Breast Cancer. Journal of Clinical Oncology JCO.2009.2026.3756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burstein HJ, Temin S, Anderson H, Buchholz TA, Davidson NE, Gelmon KE, Giordano SH, Hudis CA, Rowden D, Solky AJ, et al. 2014. Adjuvant Endocrine Therapy for Women With Hormone Receptor–Positive Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline Focused Update. Journal of Clinical Oncology 32 3959–3966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai X, Wang X, Cao C, Gao Y, Zhang S, Yang Z, Liu Y, Zhang X, Zhang W & Ye L 2018. HBXIP-elevated methyltransferase METTL3 promotes the progression of breast cancer via inhibiting tumor suppressor let-7g. Cancer Letters 415 11–19. [DOI] [PubMed] [Google Scholar]
- Cardiello JF, Sanchez GJ, Allen MA & Dowell RD 2020. Lessons from eRNAs: understanding transcriptional regulation through the lens of nascent RNAs. Transcription 11 3–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cardoso F, Paluch-Shimon S, Senkus E, Curigliano G, Aapro MS, André F, Barrios CH, Bergh J, Bhattacharyya GS, Biganzoli L, et al. 2020. 5th ESO-ESMO international consensus guidelines for advanced breast cancer (ABC 5). Annals of Oncology 31 1623–1649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Celià-Terrassa T & Kang Y 2016. Distinctive properties of metastasis-initiating cells. Genes & Development 30 892–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cerneckis J, Cui Q, He C, Yi C & Shi Y 2022. Decoding pseudouridine: an emerging target for therapeutic development. Trends in Pharmacological Sciences 43 522–535. [DOI] [PubMed] [Google Scholar]
- Chai RC, Chang YZ, Chang X, Pang B, An SY, Zhang KN, Chang YH, Jiang T & Wang YZ 2021. YTHDF2 facilitates UBXN1 mRNA decay by recognizing METTL3-mediated m(6)A modification to activate NF-kappaB and promote the malignant progression of glioma. J Hematol Oncol 14 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang G, Shi L, Ye Y, Shi H, Zeng L, Tiwary S, Huse JT, Huo L, Ma L, Ma Y, et al. 2020. YTHDF3 Induces the Translation of m6A-Enriched Gene Transcripts to Promote Breast Cancer Brain Metastasis. Cancer Cell 38 857–871.e857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen C, Nott TJ, Jin J & Pawson T 2011. Deciphering arginine methylation: Tudor tells the tale. Nat Rev Mol Cell Biol 12 629–642. [DOI] [PubMed] [Google Scholar]
- Chen F, Chen Z, Guan T, Zhou Y, Ge L, Zhang H, Wu Y, Jiang G-M, He W, Li J, et al. 2021a. N6-methyladenosine regulates mRNA stability and translation efficiency of KRT7 to promote breast cancer lung metastasis. Cancer Research 81 2847–2860. [DOI] [PubMed] [Google Scholar]
- Chen H, Yu Y, Yang M, Huang H, Ma S, Hu J, Xi Z, Guo H, Yao G, Yang L, et al. 2022. YTHDF1 promotes breast cancer progression by facilitating FOXM1 translation in an m6A-dependent manner. Cell Biosci 12 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen J & Du B 2019. Novel positioning from obesity to cancer: FTO, an m6A RNA demethylase, regulates tumour progression. Journal of Cancer Research and Clinical Oncology 145 19–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen J, Fang X, Zhong P, Song Z & Hu X 2019a. N6-methyladenosine modifications: interactions with novel RNA-binding proteins and roles in signal transduction. RNA Biol 16 991–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen L, Wang P, Bahal R, Manautou JE & Zhong X-b 2019b. Ontogenic mRNA expression of RNA modification writers, erasers, and readers in mouse liver. PLoS ONE 14 e0227102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y-S, Yang W-L, Zhao Y-L & Yang Y-G 2021b. Dynamic transcriptomic m5C and its regulatory role in RNA processing. WIREs RNA 12 e1639. [DOI] [PubMed] [Google Scholar]
- Chen Z, Qi M, Shen B, Luo G, Wu Y, Li J, Lu Z, Zheng Z, Dai Q & Wang H 2019c. Transfer RNA demethylase ALKBH3 promotes cancer progression via induction of tRNA-derived small RNAs. Nucleic Acids Research 47 2533–2545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi YD, Grabowski PJ, Sharp PA & Dreyfuss G 1986. Heterogeneous nuclear ribonucleoproteins: role in RNA splicing. Science 231 1534–1539. [DOI] [PubMed] [Google Scholar]
- Church C, Lee S, Bagg EAL, McTaggart JS, Deacon R, Gerken T, Lee A, Moir L, Mecinović J, Quwailid MM, et al. 2009. A Mouse Model for the Metabolic Effects of the Human Fat Mass and Obesity Associated FTO Gene. PLoS Genetics 5 e1000599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clarke R, Tyson JJ & Dixon JM 2015. Endocrine resistance in breast cancer – An overview and update. Molecular and Cellular Endocrinology 418, Part 3 220–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Covelo-Molares H, Obrdlik A, Poštulková I, Dohnálková M, Gregorová P, Ganji R, Potěšil D, Gawriyski L, Varjosalo M & Vaňáčová Š 2021. The comprehensive interactomes of human adenosine RNA methyltransferases and demethylases reveal distinct functional and regulatory features. Nucleic Acids Research 49 10895–10910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cui J, Wang L, Ren X, Zhang Y & Zhang H 2019. LRPPRC: A Multifunctional Protein Involved in Energy Metabolism and Human Disease. Front Physiol 10 595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cui X, Zhang X, Liu M, Zhao C, Zhang N, Ren Y, Su C, Zhang W, Sun X, He J, et al. 2020. A pan-cancer analysis of the oncogenic role of staphylococcal nuclease domain-containing protein 1 (SND1) in human tumors. Genomics 112 3958–3967. [DOI] [PubMed] [Google Scholar]
- Dai N, Zhao L, Wrighting D, Kramer D, Majithia A, Wang Y, Cracan V, Borges-Rivera D, Mootha VK, Nahrendorf M, et al. 2015. IGF2BP2/IMP2-Deficient mice resist obesity through enhanced translation of Ucp1 mRNA and Other mRNAs encoding mitochondrial proteins. Cell Metab 21 609–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dai X, Xiang L, Li T & Bai Z 2016. Cancer Hallmarks, Biomarkers and Breast Cancer Molecular Subtypes. J Cancer 7 1281–1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darnell JC, Van Driesche SJ, Zhang C, Hung KY, Mele A, Fraser CE, Stone EF, Chen C, Fak JJ, Chi SW, et al. 2011. FMRP stalls ribosomal translocation on mRNAs linked to synaptic function and autism. Cell 146 247–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darnell RB, Ke S & Darnell JE 2018. Pre-mRNA processing includes N6 methylation of adenosine residues that are retained in mRNA exons and the fallacy of “RNA epigenetics”. RNA 24 262–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Delaunay S & Frye M 2019. RNA modifications regulating cell fate in cancer. Nature Cell Biology 21 552–559. [DOI] [PubMed] [Google Scholar]
- Deng X, Su R, Feng X, Wei M & Chen J 2018. Role of N6-methyladenosine modification in cancer. Current Opinion in Genetics & Development 48 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Destefanis E, Avşar G, Groza P, Romitelli A, Torrini S, Pir P, Conticello SG, Aguilo F & Dassi E 2021. A mark of disease: how mRNA modifications shape genetic and acquired pathologies. RNA 27 367–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dinescu S, Ignat S, Lazar DA, Constantin C, Neagu M & Costache M 2019. Epitranscriptomic Signatures in lncRNAs and Their Possible Roles in Cancer. Genes (Basel) 10 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. 2012. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485 201–206. [DOI] [PubMed] [Google Scholar]
- Dominissini D, Nachtergaele S, Moshitch-Moshkovitz S, Peer E, Kol N, Ben-Haim MS, Dai Q, Di Segni A, Salmon-Divon M, Clark WC, et al. 2016. The dynamic N1-methyladenosine methylome in eukaryotic messenger RNA. Nature 530 441–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du H, Zhao Y, He J, Zhang Y, Xi H, Liu M, Ma J & Wu L 2016. YTHDF2 destabilizes m(6)A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat Commun 7 12626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du QY, Zhu ZM & Pei DS 2021. The biological function of IGF2BPs and their role in tumorigenesis. Invest New Drugs 39 1682–1693. [DOI] [PubMed] [Google Scholar]
- Dunnwald LK, Rossing MA & Li CI 2007. Hormone receptor status, tumor characteristics, and prognosis: a prospective cohort of breast cancer patients. Breast cancer research : BCR 9 R6–R6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edupuganti RR, Geiger S, Lindeboom RGH, Shi H, Hsu PJ, Lu Z, Wang SY, Baltissen MPA, Jansen P, Rossa M, et al. 2017. N(6)-methyladenosine (m(6)A) recruits and repels proteins to regulate mRNA homeostasis. Nat Struct Mol Biol 24 870–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Einstein JM, Perelis M, Chaim IA, Meena JK, Nussbacher JK, Tankka AT, Yee BA, Li H, Madrigal AA, Neill NJ, et al. 2021a. Inhibition of YTHDF2 triggers proteotoxic cell death in MYC-driven breast cancer. Molecular Cell 81 3048–3064.e3049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Einstein JM, Perelis M, Chaim IA, Meena JK, Nussbacher JK, Tankka AT, Yee BA, Li H, Madrigal AA, Neill NJ, et al. 2021b. Inhibition of YTHDF2 triggers proteotoxic cell death in MYC-driven breast cancer. Mol Cell 81 3048–3064 e3049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fakhraldeen SA, Clark RJ, Roopra A, Chin EN, Huang W, Castorino J, Wisinski KB, Kim T, Spiegelman VS & Alexander CM 2015. Two Isoforms of the RNA Binding Protein, Coding Region Determinant-binding Protein (CRD-BP/IGF2BP1), Are Expressed in Breast Epithelium and Support Clonogenic Growth of Breast Tumor Cells. J Biol Chem 290 13386–13400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fallah Y, Brundage J, Allegakoen P & Shajahan-Haq AN 2017. MYC-Driven Pathways in Breast Cancer Subtypes. Biomolecules 7 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fan Y & Guo Y 2015. Knockdown of eIF3D inhibits breast cancer cell proliferation and invasion through suppressing the Wnt/beta-catenin signaling pathway. Int J Clin Exp Pathol 8 10420–10427. [PMC free article] [PubMed] [Google Scholar]
- Fang R, Ye L & Shi H 2021. Understanding the roles of N(6)-methyladenosine writers, readers and erasers in breast cancer. Neoplasia (New York, N.Y.) 23 551–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng Y, Rhie SK, Huo D, Ruiz-Narvaez EA, Haddad SA, Ambrosone CB, John EM, Bernstein L, Zheng W, Hu JJ, et al. 2017. Characterizing Genetic Susceptibility to Breast Cancer in Women of African Ancestry. Cancer Epidemiology, Biomarkers & Prevention 26 1016–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fry NJ, Law BA, Ilkayeva OR, Carraway KR, Holley CL & Mansfield KD 2018. N(6)-methyladenosine contributes to cellular phenotype in a genetically-defined model of breast cancer progression. Oncotarget 9 31231–31243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galganski L, Urbanek MO & Krzyzosiak WJ 2017. Nuclear speckles: molecular organization, biological function and role in disease. Nucleic Acids Research 45 10350–10368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao Q, Zheng J, Ni Z, Sun P, Yang C, Cheng M, Wu M, Zhang X, Yuan L, Zhang Y, et al. 2020. The m(6)A Methylation-Regulated AFF4 Promotes Self-Renewal of Bladder Cancer Stem Cells. Stem Cells Int 2020 8849218–8849218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geuens T, Bouhy D & Timmerman V 2016. The hnRNP family: insights into their role in health and disease. Human Genetics 135 851–867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geula S, Moshitch-Moshkovitz S, Dominissini D, Mansour AA, Kol N, Salmon-Divon M, Hershkovitz V, Peer E, Mor N, Manor YS, et al. 2015. Stem cells. m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation. Science 347 1002–1006. [DOI] [PubMed] [Google Scholar]
- Giorgi Rossi P, Lebeau A, Canelo-Aybar C, Saz-Parkinson Z, Quinn C, Langendam M, McGarrigle H, Warman S, Rigau D, Alonso-Coello P, et al. 2021. Recommendations from the European Commission Initiative on Breast Cancer for multigene testing to guide the use of adjuvant chemotherapy in patients with early breast cancer, hormone receptor positive, HER-2 negative. British Journal of Cancer 124 1503–1512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goodarzi H, Najafabadi HS, Oikonomou P, Greco TM, Fish L, Salavati R, Cristea IM & Tavazoie S 2012. Systematic discovery of structural elements governing stability of mammalian messenger RNAs. Nature 485 264–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorlach M, Wittekind M, Beckman RA, Mueller L & Dreyfuss G 1992. Interaction of the RNA-binding domain of the hnRNP C proteins with RNA. EMBO J 11 3289–3295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gu X, Xue J, Ai L, Sun L, Zhu X, Wang Y & Liu C 2018. SND1 expression in breast cancer tumors is associated with poor prognosis. Ann N Y Acad Sci 1433 53–60. [DOI] [PubMed] [Google Scholar]
- Gyorffy B, Lanczky A, Eklund AC, Denkert C, Budczies J, Li Q & Szallasi Z 2010. An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Research and Treatment 123 725–731. [DOI] [PubMed] [Google Scholar]
- Han D, Longhini AP, Zhang X, Hoang V, Wilson MZ & Kosik KS 2022. Dynamic assembly of the mRNA m6A methyltransferase complex is regulated by METTL3 phase separation. PLoS Biology 20 e3001535–e3001535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han W, Zhang C, Shi CT, Gao XJ, Zhou MH, Shao QX, Shen XJ, Wu CJ, Cao F, Hu YW, et al. 2020. Roles of eIF3m in the tumorigenesis of triple negative breast cancer. Cancer Cell International 20 141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoernes TP, Heimdörfer D, Köstner D, Faserl K, Nußbaumer F, Plangger R, Kreutz C, Lindner H & Erlacher MD 2019. Eukaryotic Translation Elongation is Modulated by Single Natural Nucleotide Derivatives in the Coding Sequences of mRNAs. Genes (Basel) 10 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horiuchi K, Kawamura T & Hamakubo T 2021. Wilms’ tumor 1-associating protein complex regulates alternative splicing and polyadenylation at potential G-quadruplex-forming splice site sequences. The Journal Of Biological Chemistry 297 101248–101248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsu PJ, Zhu Y, Ma H, Guo Y, Shi X, Liu Y, Qi M, Lu Z, Shi H, Wang J, et al. 2017. Ythdc2 is an N(6)-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res 27 1115–1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu Y, Pan Q, Wang M, Ai X, Yan Y, Tian Y, Jing Y, Tang P & Jiang J 2021. m(6)A RNA Methylation Regulator YTHDF1 Correlated With Immune Microenvironment Predicts Clinical Outcomes and Therapeutic Efficacy in Breast Cancer. Front Med (Lausanne) 8 667543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu Y, Sun Z, Deng J, Hu B, Yan W, Wei H & Jiang J 2017. Splicing factor hnRNPA2B1 contributes to tumorigenic potential of breast cancer cells through STAT3 and ERK1/2 signaling pathway. Tumour Biology 39 1010428317694318. [DOI] [PubMed] [Google Scholar]
- Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, Zhao BS, Mesquita A, Liu C, Yuan CL, et al. 2018. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol 20 285–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ilık İA & Aktaş T 2021. Nuclear speckles: dynamic hubs of gene expression regulation. The FEBS Journal n/a [DOI] [PubMed] [Google Scholar]
- Isono M, Nakano M, Fukami T & Nakajima M 2022. Adenosine N6-methylation upregulates the expression of human CYP2B6 by altering the chromatin status. Biochemical Pharmacology 205 115247. [DOI] [PubMed] [Google Scholar]
- Jenjaroenpun P, Wongsurawat T, Wadley TD, Wassenaar Trudy M, Liu J, Dai Q, Wanchai V, Akel NS, Jamshidi-Parsian A, Franco AT, et al. 2020. Decoding the epitranscriptional landscape from native RNA sequences. Nucleic Acids Research 49 e7–e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, Yi C, Lindahl T, Pan T, Yang Y-G, et al. 2011. N6-Methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nature Chemical Biology 7 885–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jing JJ, Zhao X, Li H, Sun LP & Yuan Y 2021. Expression profiles and prognostic roles of m6A writers, erasers and readers in gastric cancer. Future Oncol 17 2605–2620. [DOI] [PubMed] [Google Scholar]
- Jones AN, Tikhaia E, Mourão A & Sattler M 2022. Structural effects of m6A modification of the Xist A-repeat AUCG tetraloop and its recognition by YTHDC1. Nucleic Acids Research 50 2350–2362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaklamani V, Yi N, Sadim M, Siziopikou K, Zhang K, Xu Y, Tofilon S, Agarwal S, Pasche B & Mantzoros C 2011. The role of the fat mass and obesity associated gene (FTO) in breast cancer risk. BMC Med Genet 12 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kedzierska H & Piekielko-Witkowska A 2017. Splicing factors of SR and hnRNP families as regulators of apoptosis in cancer. Cancer Lett 396 53–65. [DOI] [PubMed] [Google Scholar]
- Kierzek E, Zhang X, Watson RM, Kennedy SD, Szabat M, Kierzek R & Mathews DH 2022. Secondary structure prediction for RNA sequences including N6-methyladenosine. Nature Communications 13 1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim T-K, Hemberg M & Gray JM 2015. Enhancer RNAs: A Class of Long Noncoding RNAs Synthesized at Enhancers. Cold Spring Harbor Perspectives in Biology 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klinge CM, Piell KM, Tooley CS & Rouchka EC 2019a. HNRNPA2/B1 is upregulated in endocrine-resistant LCC9 breast cancer cells and alters the miRNA transcriptome when overexpressed in MCF-7 cells. Sci Rep 9 9430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klinge CM, Piell KM, Tooley CS & Rouchka EC 2019b. HNRNPA2/B1 is upregulated in endocrine-resistant LCC9 breast cancer cells and alters the miRNA transcriptome when overexpressed in MCF-7 cells. Sci Rep 9 9430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knuckles P & Bühler M 2018. Adenosine methylation as a molecular imprint defining the fate of RNA. FEBS Letters 592 2845–2859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kobel M, Weidensdorfer D, Reinke C, Lederer M, Schmitt WD, Zeng K, Thomssen C, Hauptmann S & Huttelmaier S 2007. Expression of the RNA-binding protein IMP1 correlates with poor prognosis in ovarian carcinoma. Oncogene 26 7584–7589. [DOI] [PubMed] [Google Scholar]
- Kono N & Arakawa K 2019. Nanopore sequencing: Review of potential applications in functional genomics. Development, Growth & Differentiation 61 316–326. [DOI] [PubMed] [Google Scholar]
- Korn SM, Ulshofer CJ, Schneider T & Schlundt A 2021. Structures and target RNA preferences of the RNA-binding protein family of IGF2BPs: An overview. Structure 29 787–803. [DOI] [PubMed] [Google Scholar]
- Koual M, Tomkiewicz C, Cano-Sancho G, Antignac J-P, Bats A-S & Coumoul X 2020. Environmental chemicals, breast cancer progression and drug resistance. Environmental health : a global access science source 19 117–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koval LE, Dionisio KL, Friedman KP, Isaacs KK & Rager JE 2022. Environmental mixtures and breast cancer: identifying co-exposure patterns between understudied vs breast cancer-associated chemicals using chemical inventory informatics. Journal of Exposure Science & Environmental Epidemiology [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kretschmer J, Rao H, Hackert P, Sloan KE, Höbartner C & Bohnsack MT 2018. The m6A reader protein YTHDC2 interacts with the small ribosomal subunit and the 5′–3′ exoribonuclease XRN1. RNA 24 1339–1350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S & Mohapatra T 2021. Deciphering Epitranscriptome: Modification of mRNA Bases Provides a New Perspective for Post-transcriptional Regulation of Gene Expression. Front Cell Dev Biol 9 628415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumari K, Groza P & Aguilo F 2021. Regulatory roles of RNA modifications in breast cancer. NAR cancer 3 zcab036–zcab036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lan Y, Liu B & Guo H 2021. The role of M6A modification in the regulation of tumor-related lncRNAs. Molecular Therapy - Nucleic Acids 24 768–779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laptev I, Dontsova O & Sergiev P 2020. Epitranscriptomics of Mammalian Mitochondrial Ribosomal RNA. Cells 9 2181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lasman L, Hanna JH & Novershtern N 2020. Role of m(6)A in Embryonic Stem Cell Differentiation and in Gametogenesis. Epigenomes 4 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee J-H, Wang R, Xiong F, Krakowiak J, Liao Z, Nguyen PT, Moroz-Omori EV, Shao J, Zhu X, Bolt MJ, et al. 2021. Enhancer RNA m6A methylation facilitates transcriptional condensate formation and gene activation. Molecular Cell 81 3368–3385.e3369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leger A, Amaral PP, Pandolfini L, Capitanchik C, Capraro F, Miano V, Migliori V, Toolan-Kerr P, Sideri T, Enright AJ, et al. 2021. RNA modifications detection by comparative Nanopore direct RNA sequencing. Nature Communications 12 7198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lemm I & Ross J 2002. Regulation of c-myc mRNA decay by translational pausing in a coding region instability determinant. Mol Cell Biol 22 3959–3969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levi O & Arava YS 2021. Pseudouridine-mediated translation control of mRNA by methionine aminoacyl tRNA synthetase. Nucleic Acids Research 49 432–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lewis CJT, Pan T & Kalsotra A 2017. RNA modifications and structures cooperate to guide RNA-protein interactions. Nat Rev Mol Cell Biol 18 202–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li A, Chen YS, Ping XL, Yang X, Xiao W, Yang Y, Sun HY, Zhu Q, Baidya P, Wang X, et al. 2017. Cytoplasmic m(6)A reader YTHDF3 promotes mRNA translation. Cell Res 27 444–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li C, Zhang C, Zhang G, Chen B, Li X, Li K, Ren C, Wen L & Liao N 2022a. YTHDF1 amplification is correlated with worse outcome and lower immune cell infiltrations in breast cancer. Cancer Biomark issue in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li E, Xia M, Du Y, Long K, Ji F, Pan F, He L, Hu Z & Guo Z 2022b. METTL3 promotes homologous recombination repair and modulates chemotherapeutic response in breast cancer by regulating the EGF/RAD51 axis. Elife 11 e75231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, Chen Z, Chen F, Xie G, Ling Y, Peng Y, Lin Y, Luo N, Chiang C-M & Wang H 2020. Targeted mRNA demethylation using an engineered dCas13b-ALKBH5 fusion protein. Nucleic Acids Research 48 5684–5694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, Gao X, Zhang Z, Lai Y, Lin X, Lin B, Ma M, Liang X, Li X, Lv W, et al. 2021a. CircCD44 plays oncogenic roles in triple-negative breast cancer by modulating the miR-502–5p/KRAS and IGF2BP2/Myc axes. Mol Cancer 20 138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li M, Tao Z, Zhao Y, Li L, Zheng J, Li Z & Chen X 2022c. 5-methylcytosine RNA methyltransferases and their potential roles in cancer. Journal of Translational Medicine 20 214–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W, Notani D & Rosenfeld MG 2016a. Enhancers as non-coding RNA transcription units: recent insights and future perspectives. Nat Rev Genet 17 207–223. [DOI] [PubMed] [Google Scholar]
- Li WJ, He YH, Yang JJ, Hu GS, Lin YA, Ran T, Peng BL, Xie BL, Huang MF, Gao X, et al. 2021a. Profiling PRMT methylome reveals roles of hnRNPA1 arginine methylation in RNA splicing and cell growth. Nat Commun 12 1946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Xiong X, Wang K, Wang L, Shu X, Ma S & Yi C 2016b. Transcriptome-wide mapping reveals reversible and dynamic N1-methyladenosine methylome. Nature Chemical Biology 12 311–316. [DOI] [PubMed] [Google Scholar]
- Li Y, Francia G & Zhang JY 2015. p62/IMP2 stimulates cell migration and reduces cell adhesion in breast cancer. Oncotarget 6 32656–32668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Gu J, Xu F, Zhu Q, Chen Y, Ge D & Lu C 2021b. Molecular characterization, biological function, tumor microenvironment association and clinical significance of m6A regulators in lung adenocarcinoma. Brief Bioinform 22. [DOI] [PubMed] [Google Scholar]
- Li Z, Yang H-Y, Dai X-Y, Zhang X, Huang Y-Z, Shi L, Wei J-F & Ding Q 2021b. CircMETTL3, upregulated in a m6A-dependent manner, promotes breast cancer progression. Int J Biol Sci 17 1178–1190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Licht K & Jantsch MF 2016. Rapid and dynamic transcriptome regulation by RNA editing and RNA modifications. Journal of Cell Biology 213 15–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin Y, Jin X, Nie Q, Chen M, Guo W, Chen L, Li Y, Chen X, Zhang W, Chen H, et al. 2022. YTHDF3 facilitates triple-negative breast cancer progression and metastasis by stabilizing ZEB1 mRNA in an m(6)A-dependent manner. Ann Transl Med 10 83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ling Z, Chen L & Zhao J 2020. m6A-dependent up-regulation of DRG1 by METTL3 and ELAVL1 promotes growth, migration, and colony formation in osteosarcoma. Bioscience Reports 40 BSR20200282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu F, Clark W, Luo G, Wang X, Fu Y, Wei J, Wang X, Hao Z, Dai Q, Zheng G, et al. 2016. ALKBH1-Mediated tRNA Demethylation Regulates Translation. Cell 167 816–828.e816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H, Lyu H, Jiang G, Chen D, Ruan S, Liu S, Zhou L, Yang M, Zeng S, He Z, et al. 2022a. ALKBH5-mediated m6A demethylation of GLUT4 mRNA promotes glycolysis and resistance to HER2-targeted therapy in breast cancer. Cancer Research CAN- 22–0800. [DOI] [PubMed] [Google Scholar]
- Liu Je, Li K, Cai J, Zhang M, Zhang X, Xiong X, Meng H, Xu X, Huang Z, Peng J, et al. 2019a. Landscape and Regulation of m6A and m6Am Methylome across Human and Mouse Tissues. Molecular Cell 77 426–440. [DOI] [PubMed] [Google Scholar]
- Liu K, Ouyang Q-Y, Zhan Y, Yin H, Liu B-X, Tan L-M, Liu R, Wu W & Yin J-Y 2022b. Pharmacoepitranscriptomic landscape revealing m6A modification could be a drug-effect biomarker for cancer treatment. Molecular Therapy - Nucleic Acids 28 464–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu L, Liu X, Dong Z, Li J, Yu Y, Chen X, Ren F, Cui G & Sun R 2019b. N6-methyladenosine-related Genomic Targets are Altered in Breast Cancer Tissue and Associated with Poor Survival. J Cancer 10 5447–5459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu N, Dai Q, Zheng G, He C, Parisien M & Pan T 2015. N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature 518 560–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu N, Zhou KI, Parisien M, Dai Q, Diatchenko L & Pan T 2017a. N6-methyladenosine alters RNA structure to regulate binding of a low-complexity protein. Nucleic Acids Res 45 6051–6063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu S, Zhu A, He C & Chen M 2020a. REPIC: a database for exploring the N(6)-methyladenosine methylome. Genome Biology 21 100–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu T, Wei Q, Jin J, Luo Q, Liu Y, Yang Y, Cheng C, Li L, Pi J, Si Y, et al. 2020b. The m6A reader YTHDF1 promotes ovarian cancer progression via augmenting EIF3C translation. Nucleic Acids Res 48 3816–3831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X, Salokas K, Tamene F, Jiu Y, Weldatsadik RG, Öhman T & Varjosalo M 2018. An AP-MS- and BioID-compatible MAC-tag enables comprehensive mapping of protein interactions and subcellular localizations. Nature Communications 9 1188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X, Yuan J, Zhang X, Li L, Dai X, Chen Q & Wang Y 2021. ATF3 Modulates the Resistance of Breast Cancer Cells to Tamoxifen through an N(6)-Methyladenosine-Based Epitranscriptomic Mechanism. Chem Res Toxicol 34 1814–1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y & Shi SL 2021. The roles of hnRNP A2/B1 in RNA biology and disease. Wiley Interdiscip Rev RNA 12 e1612. [DOI] [PubMed] [Google Scholar]
- Liu Y, Wang R, Zhang L, Li J, Lou K & Shi B 2017b. The lipid metabolism gene FTO influences breast cancer cell energy metabolism via the PI3K/AKT signaling pathway. Oncol Lett 13 4685–4690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Zhu T, Jiang Y, Bu J, Zhu X & Gu X 2022a. The Key Role of RNA Modification in Breast Cancer. Front Cell Dev Biol 10 885133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loh TJ, Moon H, Cho S, Jang H, Liu YC, Tai H, Jung DW, Williams DR, Kim HR, Shin MG, et al. 2015. CD44 alternative splicing and hnRNP A1 expression are associated with the metastasis of breast cancer. Oncology Reports 34 1231–1238. [DOI] [PubMed] [Google Scholar]
- Loibl S, Poortmans P, Morrow M, Denkert C & Curigliano G 2021. Breast cancer. The Lancet 397 1750–1769. [DOI] [PubMed] [Google Scholar]
- Lorenz DA, Sathe S, Einstein JM & Yeo GW 2020. Direct RNA sequencing enables m6A detection in endogenous transcript isoforms at base-specific resolution. RNA 26 19–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu X, Mazur SJ, Lin T, Appella E & Xu Y 2014. The pluripotency factor nanog promotes breast cancer tumorigenesis and metastasis. Oncogene 33 2655–2664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo N, Zhang K, Li X & Hu Y 2020. ZEB1 induced-upregulation of long noncoding RNA ZEB1-AS1 facilitates the progression of triple negative breast cancer by binding with ELAVL1 to maintain the stability of ZEB1 mRNA. J Cell Biochem 121 4176–4187. [DOI] [PubMed] [Google Scholar]
- Luo S & Tong L 2014. Molecular basis for the recognition of methylated adenines in RNA by the eukaryotic YTH domain. Proc Natl Acad Sci U S A 111 13834–13839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lüönd F, Tiede S & Christofori G 2021. Breast cancer as an example of tumour heterogeneity and tumour cell plasticity during malignant progression. British Journal of Cancer 125 164–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lv W, Tan Y, Xiong M, Zhao C, Wang Y, Wu M, Wu Y & Zhang Q 2021. Analysis and validation of m6A regulatory network: a novel circBACH2/has-miR-944/HNRNPC axis in breast cancer progression. J Transl Med 19 527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma F, Liu X, Zhou S, Li W, Liu C, Chadwick M & Qian C 2019. Long non-coding RNA FGF13-AS1 inhibits glycolysis and stemness properties of breast cancer cells through FGF13-AS1/IGF2BPs/Myc feedback loop. Cancer Lett 450 63–75. [DOI] [PubMed] [Google Scholar]
- Ma Y, Yang L & Li R 2020. HnRNPA2/B1 Is a Novel Prognostic Biomarker for Breast Cancer Patients. Genet Test Mol Biomarkers 24 701–707. [DOI] [PubMed] [Google Scholar]
- Mao Y, Dong L, Liu XM, Guo J, Ma H, Shen B & Qian SB 2019. m(6)A in mRNA coding regions promotes translation via the RNA helicase-containing YTHDC2. Nat Commun 10 5332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinez NM, Su A, Burns MC, Nussbacher JK, Schaening C, Sathe S, Yeo GW & Gilbert WV 2022. Pseudouridine synthases modify human pre-mRNA co-transcriptionally and affect pre-mRNA processing. Molecular Cell 82 645–659.e649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mathlin J, Le Pera L & Colombo T 2020. A Census and Categorization Method of Epitranscriptomic Marks. International Journal of Molecular Sciences 21 4684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mauer J & Jaffrey SR 2018. FTO, m(6) A(m) , and the hypothesis of reversible epitranscriptomic mRNA modifications. FEBS Letters 592 2012–2022. [DOI] [PubMed] [Google Scholar]
- Mauer J, Luo X, Blanjoie A, Jiao X, Grozhik AV, Patil DP, Linder B, Pickering BF, Vasseur J-J, Chen Q, et al. 2017. Reversible methylation of m6Am in the 5′ cap controls mRNA stability. Nature 541 371–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mauer J, Sindelar M, Despic V, Guez T, Hawley BR, Vasseur J-J, Rentmeister A, Gross SS, Pellizzoni L, Debart F, et al. 2019. FTO controls reversible m6Am RNA methylation during snRNA biogenesis. Nature Chemical Biology 15 340–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mayeda A & Krainer AR 1992. Regulation of alternative pre-mRNA splicing by hnRNP A1 and splicing factor SF2. Cell 68 365–375. [DOI] [PubMed] [Google Scholar]
- McCann KE, Hurvitz SA & McAndrew N 2019. Advances in Targeted Therapies for Triple-Negative Breast Cancer. Drugs 79 1217–1230. [DOI] [PubMed] [Google Scholar]
- McCown PJ, Ruszkowska A, Kunkler CN, Breger K, Hulewicz JP, Wang MC, Springer NA & Brown JA 2020. Naturally occurring modified ribonucleosides. WIREs RNA 11 e1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melnik BC 2015. Milk: an epigenetic amplifier of FTO-mediated transcription? Implications for Western diseases. J Transl Med 13 385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melnik BC 2021. Lifetime Impact of Cow’s Milk on Overactivation of mTORC1: From Fetal to Childhood Overgrowth, Acne, Diabetes, Cancers, and Neurodegeneration. Biomolecules 11 404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer KD 2019. DART-seq: an antibody-free method for global m6A detection. Nature Methods 16 1275–1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer KD & Jaffrey SR 2014. The dynamic epitranscriptome: N6-methyladenosine and gene expression control. Nat Rev Mol Cell Biol 15 313–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer KD & Jaffrey SR 2017. Rethinking m(6)A Readers, Writers, and Erasers. Annu Rev Cell Dev Biol 33 319–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer KD, Patil DP, Zhou J, Zinoviev A, Skabkin MA, Elemento O, Pestova TV, Qian SB & Jaffrey SR 2015. 5’ UTR m(6)A Promotes Cap-Independent Translation. Cell 163 999–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Michaleas S, Moreno Oliver A, Mueller-Berghaus J, Sarac SB, van der Elst ME, Müller-Egert S, Zander H, Enzmann H & Pignatti F 2022. The European Medicines Agency review of sacituzumab govitecan for the treatment of triple-negative breast cancer. ESMO Open 7 100497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris C, Durand S & Jalinot P 2021. Decreased expression of the translation factor eIF3e induces senescence in breast cancer cells via suppression of PARP1 and activation of mTORC1. Oncotarget 12 649–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moshitch-Moshkovitz S, Dominissini D & Rechavi G 2022. The epitranscriptome toolbox. Cell 185 764–776. [DOI] [PubMed] [Google Scholar]
- Mu L-X, Shao Y-C, Wei L, Chen F-F & Zhang J-W 2022. RNA N6-Methyladenosine Regulators Contribute to Tumor Immune Microenvironment and Have Clinical Prognostic Impact in Breast Cancer. Frontiers in Genetics 12 650499–650499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Myrick LK, Hashimoto H, Cheng X & Warren ST 2015. Human FMRP contains an integral tandem Agenet (Tudor) and KH motif in the amino terminal domain. Hum Mol Genet 24 1733–1740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niu X, Yang Y, Ren Y, Zhou S, Mao Q & Wang Y 2022. Crosstalk between m6A regulators and mRNA during cancer progression. Oncogene 41 4407–4419. [DOI] [PubMed] [Google Scholar]
- Niu Y, Lin Z, Wan A, Chen H, Liang H, Sun L, Wang Y, Li X, Xiong X-f, Wei B, et al. 2019. RNA N6-methyladenosine demethylase FTO promotes breast tumor progression through inhibiting BNIP3. Molecular Cancer 18 46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Osheim YN, Miller OL Jr & Beyer AL 1985. RNP particles at splice junction sequences on Drosophila chorion transcripts. Cell 43 143–151. [DOI] [PubMed] [Google Scholar]
- Pan X, Hong X, Li S, Meng P & Xiao F 2021. METTL3 promotes adriamycin resistance in MCF-7 breast cancer cells by accelerating pri-microRNA-221–3p maturation in a m6A-dependent manner. Experimental and Molecular Medicine 53 91–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Panneerdoss S, Eedunuri VK, Yadav P, Timilsina S, Rajamanickam S, Viswanadhapalli S, Abdelfattah N, Onyeagucha BC, Cui X, Lai Z, et al. 2018. Cross-talk among writers, readers, and erasers of m(6)A regulates cancer growth and progression. Sci Adv 4 eaar8263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park SJ, Lee H, Jo DS, Jo YK, Shin JH, Kim HB, Seo HM, Rubinsztein DC, Koh JY, Lee EK, et al. 2015. Heterogeneous nuclear ribonucleoprotein A1 post-transcriptionally regulates Drp1 expression in neuroblastoma cells. Biochim Biophys Acta 1849 1423–1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parker MT, Knop K, Sherwood AV, Schurch NJ, Mackinnon K, Gould PD, Hall AJ, Barton GJ & Simpson GG 2020. Nanopore direct RNA sequencing maps the complexity of Arabidopsis mRNA processing and m(6)A modification. Elife 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patil DP, Chen C-K, Pickering BF, Chow A, Jackson C, Guttman M & Jaffrey SR 2016. m6A RNA methylation promotes XIST-mediated transcriptional repression. Nature 537 369–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paukku K, Kalkkinen N, Silvennoinen O, Kontula KK & Lehtonen JY 2008. p100 increases AT1R expression through interaction with AT1R 3’-UTR. Nucleic Acids Res 36 4474–4487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peng F, Xu J, Cui B, Liang Q, Zeng S, He B, Zou H, Li M, Zhao H, Meng Y, et al. 2021. Oncogenic AURKA-enhanced N6-methyladenosine modification increases DROSHA mRNA stability to transactivate STC1 in breast cancer stem-like cells. Cell Research 31 345–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pereira M, Francisco S, Varanda AS, Santos M, Santos MAS & Soares AR 2018. Impact of tRNA Modifications and tRNA-Modifying Enzymes on Proteostasis and Human Disease. International Journal of Molecular Sciences 19 3738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petri BJ, Piell KM, South Whitt GC, Wilt AE, Poulton CC, Lehman NL, Clem BF, Nystoriak MA, Wysoczynski M & Klinge CM 2021. HNRNPA2B1 regulates tamoxifen- and fulvestrant-sensitivity and hallmarks of endocrine resistance in breast cancer cells. Cancer Lett 518 152–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ping X-L, Sun B-F, Wang L, Xiao W, Yang X, Wang W-J, Adhikari S, Shi Y, Lv Y, Chen Y-S, et al. 2014. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Research 24 177–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piscuoglio S, Ng CKY, Weigelt B, Chandarlapaty S & Reis-Filho JS 2018. ESR1 and endocrine therapy resistance: more than just mutations. Annals of Oncology 29 787–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polley M-YC, Dickler MN, Sinnwell J, Tenner K, de la Haba J, Loibl S, Goetz MP, Bergh J, Roberston J, Couch F, et al. 2021. A clinical calculator to predict disease outcomes in women with hormone receptor-positive advanced breast cancer treated with first-line endocrine therapy. Breast Cancer Research and Treatment 189 15–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pratanwanich PN, Yao F, Chen Y, Koh CWQ, Wan YK, Hendra C, Poon P, Goh YT, Yap PML, Chooi JY, et al. 2021. Identification of differential RNA modifications from nanopore direct RNA sequencing with xPore. Nature Biotechnology 39 1394–1402. [DOI] [PubMed] [Google Scholar]
- Pu J, Wang J, Qin Z, Wang A, Zhang Y, Wu X, Wu Y, Li W, Xu Z, Lu Y, et al. 2020. IGF2BP2 Promotes Liver Cancer Growth Through an m6A-FEN1-Dependent Mechanism. Front Oncol 10 578816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Purslow JA, Nguyen TT, Khatiwada B, Singh A & Venditti V 2021. N (6)-methyladenosine binding induces a metal-centered rearrangement that activates the human RNA demethylase Alkbh5. Sci Adv 7 eabi8215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qian J-Y, Gao J, Sun X, Cao M-D, Shi L, Xia T-S, Zhou W-B, Wang S, Ding Q & Wei J-F 2019. KIAA1429 acts as an oncogenic factor in breast cancer by regulating CDK1 in an N6-methyladenosine-independent manner. Oncogene 38 6123–6141. [DOI] [PubMed] [Google Scholar]
- Qian W, Zhu Y, Wu M, Guo Q, Wu Z, Lobie PE & Zhu T 2020. Linc00668 Promotes Invasion and Stem Cell-Like Properties of Breast Cancer Cells by Interaction With SND1. Front Oncol 10 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiao YS, Zhou JH, Jin BH, Wu YQ & Zhao B 2021. LINC00483 is regulated by IGF2BP1 and participates in the progression of breast cancer. Eur Rev Med Pharmacol Sci 25 1379–1386. [DOI] [PubMed] [Google Scholar]
- Rajecka V, Skalicky T & Vanacova S 2019. The role of RNA adenosine demethylases in the control of gene expression. Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms 1862 343–355. [DOI] [PubMed] [Google Scholar]
- Roberts JT, Porman AM & Johnson AM 2021. Identification of m6A residues at single-nucleotide resolution using eCLIP and an accessible custom analysis pipeline. RNA 27 527–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson M, Shah P, Cui YH & He YY 2019. The Role of Dynamic m(6) A RNA Methylation in Photobiology. Photochem Photobiol 95 95–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roundtree IA, Evans ME, Pan T & He C 2017a. Dynamic RNA Modifications in Gene Expression Regulation. Cell 169 1187–1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roundtree IA, Luo G-Z, Zhang Z, Wang X, Zhou T, Cui Y, Sha J, Huang X, Guerrero L, Xie P, et al. 2017b. YTHDC1 mediates nuclear export of N(6)-methyladenosine methylated mRNAs. Elife 6 e31311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sachse G, Church C, Stewart M, Cater H, Teboul L, Cox RD & Ashcroft FM 2018. FTO demethylase activity is essential for normal bone growth and bone mineralization in mice. Biochimica et biophysica acta. Molecular basis of disease 1864 843–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samuels TJ, Järvelin AI, Ish-Horowicz D & Davis I 2020. Imp/IGF2BP levels modulate individual neural stem cell growth and division through myc mRNA stability. Elife 9 e51529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanger N, Effenberger KE, Riethdorf S, Van Haasteren V, Gauwerky J, Wiegratz I, Strebhardt K, Kaufmann M & Pantel K 2011. Disseminated tumor cells in the bone marrow of patients with ductal carcinoma in situ. International Journal of Cancer 129 2522–2526. [DOI] [PubMed] [Google Scholar]
- Scadden AD 2005. The RISC subunit Tudor-SN binds to hyper-edited double-stranded RNA and promotes its cleavage. Nat Struct Mol Biol 12 489–496. [DOI] [PubMed] [Google Scholar]
- Shan J, Moran-Jones K, Munro TP, Kidd GJ, Winzor DJ, Hoek KS & Smith R 2000. Binding of an RNA trafficking response element to heterogeneous nuclear ribonucleoproteins A1 and A2. J Biol Chem 275 38286–38295. [DOI] [PubMed] [Google Scholar]
- Shi H, Wang X, Lu Z, Zhao BS, Ma H, Hsu PJ, Liu C & He C 2017. YTHDF3 facilitates translation and decay of N(6)-methyladenosine-modified RNA. Cell Res 27 315–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi H, Xu Y, Tian N, Yang M & Liang F-S 2022a. Inducible and reversible RNA N6-methyladenosine editing. Nature Communications 13 1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi W, Tang Y, Lu J, Zhuang Y & Wang J 2022b. MIR210HG promotes breast cancer progression by IGF2BP1 mediated m6A modification. Cell Biosci 12 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi Y, Zheng C, Jin Y, Bao B, Wang D, Hou K, Feng J, Tang S, Qu X, Liu Y, et al. 2020. Reduced Expression of METTL3 Promotes Metastasis of Triple-Negative Breast Cancer by m6A Methylation-Mediated COL3A1 Up-Regulation. Front Oncol 10 1126–1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siegel RL, Miller KD, Fuchs HE & Jemal A 2021. Cancer Statistics, 2021. CA: A Cancer Journal for Clinicians 71 7–33. [DOI] [PubMed] [Google Scholar]
- Siegel RL, Miller KD, Fuchs HE & Jemal A 2022. Cancer statistics, 2022. CA: A Cancer Journal for Clinicians 72 7–33. [DOI] [PubMed] [Google Scholar]
- Simon Y, Kessler SM, Bohle RM, Haybaeck J & Kiemer AK 2014. The insulin-like growth factor 2 (IGF2) mRNA-binding protein p62/IGF2BP2–2 as a promoter of NAFLD and HCC? Gut 63 861–863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siomi H & Dreyfuss G 1995. A nuclear localization domain in the hnRNP A1 protein. J Cell Biol 129 551–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sledge GW Jr, Toi M, Neven P, Sohn J, Inoue K, Pivot X, Burdaeva O, Okera M, Masuda N, Kaufman PA, et al. 2020. The Effect of Abemaciclib Plus Fulvestrant on Overall Survival in Hormone Receptor–Positive, ERBB2-Negative Breast Cancer That Progressed on Endocrine Therapy—MONARCH 2: A Randomized Clinical Trial. JAMA Oncology 6 116–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stohr N, Kohn M, Lederer M, Glass M, Reinke C, Singer RH & Huttelmaier S 2012. IGF2BP1 promotes cell migration by regulating MK5 and PTEN signaling. Genes Dev 26 176–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stöhr N, Köhn M, Lederer M, Glass M, Reinke C, Singer RH & Hüttelmaier S 2012. IGF2BP1 promotes cell migration by regulating MK5 and PTEN signaling. Genes & Development 26 176–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su R, Dong L, Li C, Nachtergaele S, Wunderlich M, Qing Y, Deng X, Wang Y, Weng X, Hu C, et al. 2018. R-2HG Exhibits Anti-tumor Activity by Targeting FTO/m6A/MYC/CEBPA Signaling. Cell 172 90–105.e123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su R, Dong L, Li Y, Gao M, Han L, Wunderlich M, Deng X, Li H, Huang Y, Gao L, et al. 2020. Targeting FTO Suppresses Cancer Stem Cell Maintenance and Immune Evasion. Cancer Cell 38 79–96.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun T, Wu Z, Wang X, Wang Y, Hu X, Qin W, Lu S, Xu D, Wu Y, Chen Q, et al. 2020. LNC942 promoting METTL14-mediated m6A methylation in breast cancer cell proliferation and progression. Oncogene 39 5358–5372. [DOI] [PubMed] [Google Scholar]
- Sun Y, Dong D, Xia Y, Hao L, Wang W & Zhao C 2022. YTHDF1 promotes breast cancer cell growth, DNA damage repair and chemoresistance. Cell Death Dis 13 230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A & Bray F 2021. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 71 209–249. [DOI] [PubMed] [Google Scholar]
- Suster I & Feng Y 2021. Multifaceted Regulation of MicroRNA Biogenesis: Essential Roles and Functional Integration in Neuronal and Glial Development. International Journal of Molecular Sciences 22 6765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swanson MS & Dreyfuss G 1988. RNA binding specificity of hnRNP proteins: a subset bind to the 3’ end of introns. EMBO J 7 3519–3529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swanson MS, Nakagawa TY, LeVan K & Dreyfuss G 1987. Primary structure of human nuclear ribonucleoprotein particle C proteins: conservation of sequence and domain structures in heterogeneous nuclear RNA, mRNA, and pre-rRNA-binding proteins. Mol Cell Biol 7 1731–1739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szabat M, Prochota M, Kierzek R, Kierzek E & Mathews DH 2022. A Test and Refinement of Folding Free Energy Nearest Neighbor Parameters for RNA Including N6-Methyladenosine. Journal of Molecular Biology 167632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tai J, Wang L, Guo H, Yan Z & Liu J 2022. Prognostic implications of N6-methyladenosine RNA regulators in breast cancer. Sci Rep 12 1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan A, Dang Y, Chen G & Mo Z 2015. Overexpression of the fat mass and obesity associated gene (FTO) in breast cancer and its clinical implications. Int J Clin Exp Pathol 8 13405–13410. [PMC free article] [PubMed] [Google Scholar]
- Tang L, Chen Y, Tang X, Wei D, Xu X & Yan F 2020. Long Noncoding RNA DCST1-AS1 Promotes Cell Proliferation and Metastasis in Triple-negative Breast Cancer by Forming a Positive Regulatory Loop with miR-873–5p and MYC. J Cancer 11 311–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang Y, Chen K, Song B, Ma J, Wu X, Xu Q, Wei Z, Su J, Liu G, Rong R, et al. 2021. m6A-Atlas: a comprehensive knowledgebase for unraveling the N6-methyladenosine (m6A) epitranscriptome. Nucleic Acids Research 49 D134–D143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarun SZ Jr, Wells SE, Deardorff JA & Sachs AB 1997. Translation initiation factor eIF4G mediates in vitro poly(A) tail-dependent translation. Proc Natl Acad Sci U S A 94 9046–9051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tea MK, Weghofer A, Wagner K & Singer CF 2013. Association of BRCA1/2 mutations with FMR1 genotypes: effects on menarcheal and menopausal age. Maturitas 75 148–151. [DOI] [PubMed] [Google Scholar]
- Tegowski M, Flamand MN & Meyer KD 2022. scDART-seq reveals distinct m(6)A signatures and mRNA methylation heterogeneity in single cells. Molecular Cell 82 868–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thüring K, Schmid K, Keller P & Helm M 2016. Analysis of RNA modifications by liquid chromatography–tandem mass spectrometry. Methods 107 48–56. [DOI] [PubMed] [Google Scholar]
- Torres ETR & Emens LA 2022. Emerging combination immunotherapy strategies for breast cancer: dual immune checkpoint modulation, antibody–drug conjugates and bispecific antibodies. Breast Cancer Research and Treatment 191 291–302. [DOI] [PubMed] [Google Scholar]
- Toy W, Weir H, Razavi P, Lawson M, Goeppert AU, Mazzola AM, Smith A, Wilson J, Morrow C, Wong WL, et al. 2017. Activating ESR1 Mutations Differentially Affect the Efficacy of ER Antagonists. Cancer Discov 7 277–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vikesaa J, Hansen TV, Jonson L, Borup R, Wewer UM, Christiansen J & Nielsen FC 2006. RNA-binding IMPs promote cell adhesion and invadopodia formation. EMBO J 25 1456–1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- von Hagen F, Gundert L, Strick A, Klumper N, Schmidt D, Kristiansen G, Tolkach Y, Toma M, Ritter M & Ellinger J 2021. N(6) -Methyladenosine (m(6) A) readers are dysregulated in renal cell carcinoma. Mol Carcinog 60 354–362. [DOI] [PubMed] [Google Scholar]
- von Minckwitz G, Procter M, de Azambuja E, Zardavas D, Benyunes M, Viale G, Suter T, Arahmani A, Rouchet N, Clark E, et al. 2017. Adjuvant Pertuzumab and Trastuzumab in Early HER2-Positive Breast Cancer. New England Journal of Medicine 377 122–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wan W, Ao X, Chen Q, Yu Y, Ao L, Xing W, Guo W, Wu X, Pu C, Hu X, et al. 2022. METTL3/IGF2BP3 axis inhibits tumor immune surveillance by upregulating N(6)-methyladenosine modification of PD-L1 mRNA in breast cancer. Molecular Cancer 21 60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C-Q, Tang C-H, Wang Y, Huang B-F, Hu G-N, Wang Q & Shao J-K 2022. Upregulated WTAP expression appears to both promote breast cancer growth and inhibit lymph node metastasis. Sci Rep 12 1023–1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Xu B & Shi J 2020a. N6-methyladenosine METTL3 promotes the breast cancer progression via targeting Bcl-2. Gene 722 144076. [DOI] [PubMed] [Google Scholar]
- Wang J & Wang L 2020. Deep analysis of RNA N(6)-adenosine methylation (m(6)A) patterns in human cells. NAR genomics and bioinformatics 2 lqaa007–lqaa007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang S, Zou X, Chen Y, Cho WC & Zhou X 2020b. Effect of N6-Methyladenosine Regulators on Progression and Prognosis of Triple-Negative Breast Cancer. Front Genet 11 580036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, Feng J, Xue Y, Guan Z, Zhang D, Liu Z, Gong Z, Wang Q, Huang J, Tang C, et al. 2016. Structural basis of N6-adenosine methylation by the METTL3–METTL14 complex. Nature 534 575–578. [DOI] [PubMed] [Google Scholar]
- Wang X, Ji Y, Feng P, Liu R, Li G, Zheng J, Xue Y, Wei Y, Ji C, Chen D, et al. 2021. The m6A Reader IGF2BP2 Regulates Macrophage Phenotypic Activation and Inflammatory Diseases by Stabilizing TSC1 and PPARgamma. Adv Sci (Weinh) 8 2100209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, Lu Z, Gomez A, Hon GC, Yue Y, Han D, Fu Y, Parisien M, Dai Q, Jia G, et al. 2014. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature 505 117–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, Zhao BS, Roundtree IA, Lu Z, Han D, Ma H, Weng X, Chen K, Shi H & He C 2015. N(6)-methyladenosine Modulates Messenger RNA Translation Efficiency. Cell 161 1388–1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z, Tong D, Han C, Zhao Z, Wang X, Jiang T, Li Q, Liu S, Chen L, Chen Y, et al. 2019. Blockade of miR-3614 maturation by IGF2BP3 increases TRIM25 expression and promotes breast cancer cell proliferation. EBioMedicine 41 357–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei J, Liu F, Lu Z, Fei Q, Ai Y, He PC, Shi H, Cui X, Su R, Klungland A, et al. 2018. Differential m6A, m6Am, and m1A Demethylation Mediated by FTO in the Cell Nucleus and Cytoplasm. Molecular Cell 71 973–985.e975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei K, Gao Y, Wang B & Qu YX 2022. Methylation recognition protein YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) regulates the proliferation, migration and invasion of osteosarcoma by regulating m6A level of CCR4-NOT transcription complex subunit 7 (CNOT7). Bioengineered 13 5236–5250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen J, Lv R, Ma H, Shen H, He C, Wang J, Jiao F, Liu H, Yang P, Tan L, et al. 2018. Zc3h13 Regulates Nuclear RNA m6A Methylation and Mouse Embryonic Stem Cell Self-Renewal. Molecular Cell 69 1028–1038.e1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiener D & Schwartz S 2021. The epitranscriptome beyond m6A. Nature Reviews Genetics 22 119–131. [DOI] [PubMed] [Google Scholar]
- Wilkinson L & Gathani T 2021. Understanding breast cancer as a global health concern. The British Journal of Radiology 95 20211033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu L, Wu D, Ning J, Liu W & Zhang D 2019. Changes of N6-methyladenosine modulators promote breast cancer progression. BMC Cancer 19 326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu Y, Wang Z, Han L, Guo Z, Yan B, Guo L, Zhao H, Wei M, Hou N, Ye J, et al. 2022. PRMT5 regulates RNA m6A demethylation for doxorubicin sensitivity in breast cancer. Molecular Therapy [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiang Y, Laurent B, Hsu C-H, Nachtergaele S, Lu Z, Sheng W, Xu C, Chen H, Ouyang J, Wang S, et al. 2017. RNA m(6)A methylation regulates the ultraviolet-induced DNA damage response. Nature 543 573–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiao W, Adhikari S, Dahal U, Chen YS, Hao YJ, Sun BF, Sun HY, Li A, Ping XL, Lai WY, et al. 2016. Nuclear m(6)A Reader YTHDC1 Regulates mRNA Splicing. Molecular Cell 61 507–519. [DOI] [PubMed] [Google Scholar]
- Xu W, He C, Kaye EG, Li J, Mu M, Nelson GM, Dong L, Wang J, Wu F, Shi YG, et al. 2022. Dynamic control of chromatin-associated m6A methylation regulates nascent RNA synthesis. Molecular Cell 82 1156–1168.e1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu Y, Ye S, Zhang N, Zheng S, Liu H, Zhou K, Wang L, Cao Y, Sun P & Wang T 2020. The FTO/miR-181b-3p/ARL5B signaling pathway regulates cell migration and invasion in breast cancer. Cancer Communications 40 484–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xue C, Chu Q, Zheng Q, Jiang S, Bao Z, Su Y, Lu J & Li L 2022. Role of main RNA modifications in cancer: N6-methyladenosine, 5-methylcytosine, and pseudouridine. Signal Transduction And Targeted Therapy 7 142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang L, Wu S, Ma C, Song S, Jin F, Niu Y & Tong W-M 2021. RNA m(6)A Methylation Regulators Subclassify Luminal Subtype in Breast Cancer. Front Oncol 10 611191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang X, Yang Y, Sun B-F, Chen Y-S, Xu J-W, Lai W-Y, Li A, Wang X, Bhattarai DP, Xiao W, et al. 2017. 5-methylcytosine promotes mRNA export — NSUN2 as the methyltransferase and ALYREF as an m5C reader. Cell Research 27 606–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Y, Hsu PJ, Chen Y-S & Yang Y-G 2018. Dynamic transcriptomic m6A decoration: writers, erasers, readers and functions in RNA metabolism. Cell Research 28 616–624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Z, Wang T, Wu D, Min Z, Tan J & Yu B 2020. RNA N6-methyladenosine reader IGF2BP3 regulates cell cycle and angiogenesis in colon cancer. J Exp Clin Cancer Res 39 203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao X, Li W, Li L, Li M, Zhao Y, Fang, Zeng X & Luo Z 2022. YTHDF1 upregulation mediates hypoxia-dependent breast cancer growth and metastasis through regulating PKM2 to affect glycolysis. Cell Death Dis 13 258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin H, Zhang X, Yang P, Zhang X, Peng Y, Li D, Yu Y, Wu Y, Wang Y, Zhang J, et al. 2021. RNA m6A methylation orchestrates cancer growth and metastasis via macrophage reprogramming. Nat Commun 12 1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshida A, Oyoshi T, Suda A, Futaki S & Imanishi M 2022. Recognition of G-quadruplex RNA by a crucial RNA methyltransferase component, METTL14. Nucleic Acids Research 50 449–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu L, Liu X, Cui K, Di Y, Xin L, Sun X, Zhang W, Yang X, Wei M, Yao Z, et al. 2015. SND1 Acts Downstream of TGFbeta1 and Upstream of Smurf1 to Promote Breast Cancer Metastasis. Cancer Res 75 1275–1286. [DOI] [PubMed] [Google Scholar]
- Zaccara S & Jaffrey SR 2020. A Unified Model for the Function of YTHDF Proteins in Regulating m6A-Modified mRNA. Cell 181 1582–1595.e1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zaccara S, Ries RJ & Jaffrey SR 2019. Reading, writing and erasing mRNA methylation. Nature Reviews Molecular Cell Biology 20 608–624. [DOI] [PubMed] [Google Scholar]
- Zacharakis N, Huq LM, Seitter SJ, Kim SP, Gartner JJ, Sindiri S, Hill VK, Li YF, Paria BC, Ray S, et al. 2022. Breast Cancers Are Immunogenic: Immunologic Analyses and a Phase II Pilot Clinical Trial Using Mutation-Reactive Autologous Lymphocytes. Journal of Clinical Oncology JCO.21.02170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang B, Gu Y & Jiang G 2020. Expression and Prognostic Characteristics of m6 A RNA Methylation Regulators in Breast Cancer. Frontiers in Genetics 11 604597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C, Samanta D, Lu H, Bullen JW, Zhang H, Chen I, He X & Semenza GL 2016. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. Proceedings of the National Academy of Sciences 113 E2047–E2056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L, Wan Y, Zhang Z, Jiang Y, Gu Z, Ma X, Nie S, Yang J, Lang J, Cheng W, et al. 2021a. IGF2BP1 overexpression stabilizes PEG10 mRNA in an m6A-dependent manner and promotes endometrial cancer progression. Theranostics 11 1100–1114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang M, Bodi Z, Mackinnon K, Zhong S, Archer N, Mongan NP, Simpson GG & Fray RG 2022. Two zinc finger proteins with functions in m6A writing interact with HAKAI. Nature Communications 13 1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang S, Liu C & Zhang X 2019. Mitochondrial Damage Mediated by miR-1 Overexpression in Cancer Stem Cells. Mol Ther Nucleic Acids 18 938–953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang W, Liu H, Jiang J, Yang Y, Wang W & Jia Z 2021b. CircRNA circFOXK2 facilitates oncogenesis in breast cancer via IGF2BP3/miR-370 axis. Aging (Albany NY) 13 18978–18992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang W, Qian Y & Jia G 2021c. The detection and functions of RNA modification m6A based on m6A writers and erasers. Journal of Biological Chemistry 297 100973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Z, Zhang C, Luo Y, Zhang G, Wu P, Sun N & He J 2021d. RNA N6-methyladenosine modification in the lethal teamwork of cancer stem cells and the tumor immune microenvironment: Current landscape and therapeutic potential. Clinical and Translational Medicine 11 e525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao C, Ling X, Xia Y, Yan B & Guan Q 2022. LncRNA UCA1 promotes SOX12 expression in breast cancer by regulating m6A modification of miR-375 by METTL14 through DNA methylation. Cancer Gene Therapy 29 1043–1055. [DOI] [PubMed] [Google Scholar]
- Zhao G, An J, Pu Q, Geng W, Song H, Zhao Q & Gao H 2021. Gene Signatures and Cancer-Immune Phenotypes Based on m6A Regulators in Breast Cancer. Front Oncol 11 756412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao Z, Meng J, Su R, Zhang J, Chen J, Ma X & Xia Q 2020. Epitranscriptomics in liver disease: Basic concepts and therapeutic potential. Journal of Hepatology 73 664–679. [DOI] [PubMed] [Google Scholar]
- Zhao Z, Wen W, Michailidou K, Bolla MK, Wang Q, Zhang B, Long J, Shu X-O, Schmidt MK, Milne RL, et al. 2016. Association of genetic susceptibility variants for type 2 diabetes with breast cancer risk in women of European ancestry. Cancer causes & control : CCC 27 679–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng F, Du F, Qian H, Zhao J, Wang X, Yue J, Hu N, Si Y, Xu B & Yuan P 2021a. Expression and clinical prognostic value of m6A RNA methylation modification in breast cancer. Biomarker Research 9 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng F, Kelly Marcus R, Ramms Dana J, Heintschel Marissa L, Tao K, Tutuncuoglu B, Lee John J, Ono K, Foussard H, Chen M, et al. 2021b. Interpretation of cancer mutations using a multiscale map of protein systems. Science 374 eabf3067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng G, Dahl JA, Niu Y, Fedorcsak P, Huang C-M, Li CJ, Vågbø Cathrine B, Shi Y, Wang W-L, Song S-H, et al. 2013. ALKBH5 Is a Mammalian RNA Demethylase that Impacts RNA Metabolism and Mouse Fertility. Molecular Cell 49 18–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng L, Li M, Wei J, Chen S, Xue C, Zhan Y, Duan Y, Deng H, Xiong W, Li G, et al. 2022. The emerging roles of the interaction between m6A modification and c-Myc in driving tumorigenesis and development. Journal of Cellular Physiology [DOI] [PubMed] [Google Scholar]
- Zheng Q, Liu H, Ye J, Zhang H, Jia Z & Cao J 2016. Nuclear distribution of eIF3g and its interacting nuclear proteins in breast cancer cells. Mol Med Rep 13 2973–2980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou H, Lv Q & Guo Z 2018. Transcriptomic signature predicts the distant relapse in patients with ER+ breast cancer treated with tamoxifen for five years. Mol Med Rep 17 3152–3157. [DOI] [PubMed] [Google Scholar]
- Zhou K, Sun Y, Dong D, Zhao C & Wang W 2021. EMP3 negatively modulates breast cancer cell DNA replication, DNA damage repair, and stem-like properties. Cell Death Dis 12 844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou KI & Pan T 2016. Structures of the m(6)A Methyltransferase Complex: Two Subunits with Distinct but Coordinated Roles. Molecular Cell 63 183–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou KI, Shi H, Lyu R, Wylder AC, Matuszek Ż, Pan JN, He C, Parisien M & Pan T 2019. Regulation of Co-transcriptional Pre-mRNA Splicing by m6A through the Low-Complexity Protein hnRNPG. Molecular Cell 76 70–81.e79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Z, Zhu T, Chen S, Qin S, Huang Y & Liu D 2022. Systematic Analysis of the Expression Profile and Prognostic Significance of the IGF2BP Family in Lung Adenocarcinoma. Curr Cancer Drug Targets 22 340–350. [DOI] [PubMed] [Google Scholar]
- Zhu L, Tian Q, Gao H, Wu K, Wang B, Ge G, Jiang S, Wang K, Zhou C, He J, et al. 2022. PROX1 promotes breast cancer invasion and metastasis through WNT/beta-catenin pathway via interacting with hnRNPK. Int J Biol Sci 18 2032–2046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu P, He F, Hou Y, Tu G, Li Q, Jin T, Zeng H, Qin Y, Wan X, Qiao Y, et al. 2021. A novel hypoxic long noncoding RNA KB-1980E6.3 maintains breast cancer stem cell stemness via interacting with IGF2BP1 to facilitate c-Myc mRNA stability. Oncogene 40 1609–1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
