Abstract
N7-methylguanosine (m7G) is one of the most common post-transcriptional epigenetic modifications. Different m7G methyltransferases (writers) load the m7G-cap at the 5’-terminal or inside the RNAs. For example, writers such as methyltransferase-like 1 (METTL1)/WD repeat domain 4 (WDR4) and Williams-Beuren syndrome chromosome region 22 (WBSCR22) have been reported in mammals to promote cell proliferation, EMT, and chemoresistance in massive quantities of cancers. The underlying mechanism includes modulating the RNA secondary structure, preventing RNA degradation from exonucleases, and improving codon-dependent translation. However, some studies have shown that in colorectal and lung cancers, m7G inhibits tumor progression. m7G binding proteins (readers), such as eukaryotic translation initiation factor 4E (eIF4E), promote the efficiency of cap-dependent translation and accelerate the cell cycle to improve cancer progression. Due to the more profound understanding of m7G regulatory proteins in cancer, numerous studies aim to investigate the clinical efficiency of m7G-targeted therapy. eIF4E antisense oligonucleotide drug (4EASO) and Ribavirin are the most mature trials that competitively inhibit the binding of eIF4E to m7G-cap. These drugs have encouraging results in halting cancer progression and improving prognosis, including AML and non-small cell lung cancer, which provide a promising perspective for developing more m7G-targeted drugs. In the future, we look forward to an ongoing investigation into the role of m7G modification in tumors and drug resistance to m7G-related therapies to be solved. Therefore, the clinical application would be put into practice as soon as possible.
Keywords: N7-methylguanosine modification, cancer, cancer therapy, RNA metabolism
Introduction
RNA epigenetic modification regulates gene expression at the post-transcriptional level, significantly affecting RNA properties [1]. To date, more than 150 different RNA modifications have been identified, the most common ones of which are N1-methyladenosine (m1A), 3-methylcytidine (m3C), 5-methylcytosine (m5C), N6-methyladenosine (m6A) and N7-methylguanosine (m7G) modification [2]. m7G modification, methylation in the seventh N on RNA guanylate, is widely present in mRNA, rRNA, tRNA, sRNA, telomerase RNA (TERC), microRNA (miRNA), and long non-coding RNA (lncRNA) [3-5]. However, small cytoplasmic RNA (scRNA), small interfering RNA (siRNA), and circular RNA (circRNA) have not been reported to contain m7G modification. Two main types of m7G modification are classified according to their locations, which are m7G-cap at 5’-terminal and internal m7G modification (Figure 1) [6,7]. And at the N2 site inside the m7G-cap, guanylate can be further hypermethylated to m2,2,7G-trimethylguanosine (TMG) caps [8]. Like other epigenetic modifications, m7G modifications are dynamically regulated by RNA methyltransferases and m7G recognition proteins. Although, Alkane monooxygenase (AlkB) has a general function in demethylating almost all the RNA modifications, including m7G, which is utilized to detect m7G sites inside RNAs [9]. However, despite our curiosity about m7G demethylase, we still did not find any clues on the specific enzyme to the best search. Indeed, we hope that more studies will explore the potential enzymes that demethylase m7G modification and their function in tumor progression.
Figure 1.

Chemical structures of internal m7G modification and m7G-cap (Created with KingDraw).
As a conserved RNA modification in the genome, m7G modification is dynamically regulated under different physiological and pathological conditions. Firstly, m7G modification in ischemic stroke also shows dynamic changes, from the hypomethylation in an ischemic state to the METTL1-mediated hypermethylation during ischemia-reperfusion injury [10]. m7G regulation is not only reflected in the global methylation level but also in the sites of internal methylation, which typically occurs in the GA/GG enriched region in the 5’-untranslated region (UTR) [11]. While in response to oxidative and heat stress, m7G modifications are increased and dynamically accumulate in the coding sequence (CDS) and 3’-UTR [12,13]. Secondly, the abnormal self-renewal and differentiation of cells during nervous system development are usually accompanied by the change of m7G methylation, which affects the expression of stem cell gene signature [14]. Significant differences in m7G methylation are shown in malignant transformation and response to chemotherapeutic drugs [15,16].
Interestingly, whether the methylation is upregulated or downregulated depends on the specific disease type. In the following text, we will explain the role of m7G modification and related regulators in the occurrence and development of various tumors. Hopefully, we aim to provide new insights into cancer diagnosis and treatment.
The history of m7G modification research
As early as 1975, around 14 articles reported the m7G modification in viral RNAs, mRNAs, tRNAs, and rRNAs [17,18]. Hefti E was the first scientist to identify m7G-cap in 5’ nucleotide sequence in sindbis viral RNA [19]. The first m7G methyltransferase in the vaccinia virus was solubilized in 1975 by M J Ensinger [20]. m7G-cap and inner m7G modification and their methyltransferases in diverse RNAs were subsequently discovered after that [21,22]. Three years later, the successful development of m7G-specific antibodies significantly promoted the research of m7G modification [23]. m7G reader was first reported in 1991 as eif4E bound to the m7G base through hydrogen-bond pairing [24]. Later in 2007, the m7G reader, eIF4E, was found to be closely related to various malignant tumors, which opened a new era of exploring the mechanism of cancer progression based on m7G modification [25]. Interestingly, there was an academic debate on the existence of m7G modification in human let-7e in colon cancer in 2020 [26,27]. Jeppe Vinther and Luca Pandolfini had a heated discussion on m7G detection methods (M7G-RIP-seq, BoRed-seq) and analysis methods (MS/MS analysis, etc.). This provides a deeper thinking for scientists devoted to RNA epigenetic modification. Since then, the research on m7G modification has experienced explosive growth. More and more scientists are committed to exploring the relationship between m7G modification and tumorigenesis.
Regulators of m7G modification
Writers: guanine-N7 methyltransferases (MTase)
Most of the m7G methyltransferases belong to the Adomet-dependent methyltransferase family [28,29]. The m7G writers differ in species and RNA substrates (Table 1). TrmB is the first identified m7G writer in E. coli to catalyze m7G modification at the G46 site in tRNA [30]. m7G46, located in a variable region within tRNA, is the most common methylation site in prokaryotic and eukaryotic tRNAs [31]. In yeast, the writer is a complex of catalytic subunit Trm8 and auxiliary subunit Trm82 [32]. In mammals, METTL1/WDR4 complex is responsible for m7G modification in tRNAs, mRNAs, and miRNAs [33,34]. The latest study unraveled the core mechanism of METTL1 catalyzing m7G, that the disordered N-terminal region of METTL1 is the nexus for methyltransferase activity, which can be regulated by Ser27 phosphorylation [35]. WDR4 acts as a scaffold for METTL1 and tRNA T-arm, activating m7G methylation in the presence of S-adenosylmethionine or S-adenosylhomocysteine [36].
Table 1.
M7G regulators for targeted RNAs
| RNA | m7G types | Organisms | guanine-N7 MTase | Reference | 
|---|---|---|---|---|
| tRNA | Internal m7G | Eubacteria | TrmB* | [133] | 
| Internal m7G | Yeast | Trm8*/Trm82 | [134] | |
| Internal m7G | Mammalian | METTL1*/WDR4 | [20] | |
| TMG-cap | Yeast | TGS1 | [39] | |
| mRNA | Internal m7G | Mammalian | METTL1*/WDR4 | [34] | 
| m7G cap | vaccina virus | Abd1 (vD1*/vD2) | [135] | |
| m7G cap | Mammalian | RNMT (HCMT1) | [37] | |
| miRNA | Internal m7G | Mammalian | METTL1*/WDR4 | [33] | 
| 16S rRNA | Internal m7G | Streptomyces tenebrarius | KgmB | [40] | 
| 18S rRNA | Internal m7G | Yeast | Bud23-Trm112 | [41] | 
| Internal m7G | Mammalian | WBSCR22 | [23] | |
| 23S rRNA | Internal m7G | E. coli | YcbY (RlmKL) | [46] | 
| sRNA | TMG-cap | Yeast, Mammalian | TGS1 | [8] | 
| TERC | TMG-cap | Mammalian | TGS1 | [24,25] | 
Indicating catalytic subunits in complexes.
The m7G writer for the cap structure at the 5’ end is a complex of RNMT and RNMT-activated small protein (RAM) [37]. Furthermore, CDK1-cyclinB1 activates RNMT by direct phosphorylation and indirectly blocking the inhibitory protein, nuclear protein subunit α2 (KPNA2) [38]. As for TMG-cap, TGS1 is responsible for its biosynthesis in tRNA, sRNA, and telomerase RNA [39]. For the 16S rRNA in Streptomyces tenebrarius, KgmB writes the m7G modification at the G1405 site [40]. For 18S rRNA in yeast and mammals, Bud23/TRMT112 [41,42] and WBSCR22 [43-45] are identified as the responsible writers. m7G modification of the large subunit in 23S rRNA requires YcbY (RlmKL) [46].
Readers
RNA epigenetic modifications must be bound and recognized by specific proteins before performing corresponding functions [41]. Known readers include the YTH family proteins for identifying m6A modifications [47] and the nuclear regulatory protein ALYREF for m5C modifications [48]. There are mainly two m7G readers, CBC and eIF4E. CBC consists of a nuclear cap-binding protein subunit 2 (NCBP2/CBP20) and an adaptor protein subunit 1 (NCBP1/CBP80) [49]. After the m7G-cap is recognized, RNAs are translocated from the nucleus to the cytoplasm [50]. The interaction between the negative π-electron clouds from two aromatic residues in eIF4E and the positive charge in the m7G cap makes them available to bind to each other [51]. Then, translation initiation complexes are recruited to initiate mRNA translation [52]. Notably, the nuclear localization of eIF4E mediated by importin 8 is essential to maintain its pro-oncogenic activity [53]. When m7G-cap is hypermethylated, snuportin-1 and other factors, such as importin-β, recognize the TMG-cap structure and mediate the translocation of modified RNA from the cytoplasm to the nucleus [54]. Recent studies have revealed that eIF4E directly binds the methyltransferase domain in RNMT to form an m7G cap-eIF4E-RNMT complex and promotes m7G-cap assembly [55]. As mentioned above, various proteins regulate the spatial location of RNAs and RNA biological functions by recognizing m7G modification.
The impact of m7G modification on transcription and translation
m7G modification regulates almost all events in RNA post-transcriptional regulation, including post-transcriptional splicing, RNA cytoplasmic localization, and RNA stability [56]. In addition, the TMP cap in TERC inhibits telomerase assembly and restricts elongation [57]. In precursor tRNAs, G-quadruplex formation is interrupted, and canonical stem-loop structure formation is promoted through m7G modification [33]. m7G-cap enhances the plasticity of RNA cytosolic localization by preparing a docking site for CBC to mediate nuclear export [54]. Conversely, TMG-cap is recognized by snuportin-1 to translocate RNAs from the cytoplasm to the nucleus [58]. m7G modification enhances the stability of RNAs through various mechanisms [59]. Firstly, positive charge or zwitterion is introduced onto the nucleobase by m7G modification [60]. On the other hand, the endogenous m7G46 in tRNA forms a stable tertiary base pair with C13-G22 to maintain the tRNA geometry in the secondary structure [61]. Loss of the m7G modification makes tRNA more susceptible to the rapid tRNA degradation (RTD) pathway, especially under heat stress [62]. In addition, the m7G cap and TMG-cap impart a protective layer at the 5’ end of the RNAs, which effectively prevents 5’ exonuclease activities [63,64]. Thus, m7G modifications contribute to RNA stability, simultaneously enhancing the expression of oncogenes and promoting malignant transformation.
The initiation and process of mRNA translation undergo the regulation of m7G modification [52]. m7G-cap recruits initiation factors like eukaryotic translation initiation factor E (eIF4E) and eIF4F [65]. Additionally, m7G-cap facilitates the “RNA-looping” to generate the recognition of translation initiation codon by 40S ribosome at the 5’-end [66]. Besides, tRNA (m7G46) modification adjusts the translation process by altering tRNA codon decoding ability and ribosome-pausing mechanism [15]. Codon recognition dependent mechanism introduces a theory that tRNAs with higher frequencies of m7G modification enjoys higher translation efficiency [67]. However, whether m7G modification has a direct biological role on ribosomes needs to be further explored. In summary, we listed the regulatory function of m7G modification on RNA metabolism (Figure 2).
Figure 2.
The process and molecular functions of m7G modification in RNA metabolism. Different methyltransferases were essential for introducing internal m7G modification and cap m7G modification at 5’-termial in human, including METTL1/WDR4, RNMT/RAM, WBSC22 and TGS1, relative locations are marked with numbers. In the nucleus, m7G-cap could be recognized by CBC to generate RNA nuclear exportation. In the cytoplasm, the binding of TMG-cap and Snuportin1 associated with importin-β enables RNA to be imported into the nucleus. m7G regulates RNA degradation by maintaining RNA geometry structure and protecting RNAs from 5’ exonucleases and RTD pathway. m7G promotes RNA maturation by affecting secondary structures. m7G regulates mRNA translation through eIF4E recognition, interference of ribosome pausing and tRNA decoding.
Roles of m7G modification in normal cells
Dysregulation of m7G modification directly affects cell function. In the HeLa cell line, CBC, accompanied by ARS2, promotes RNA stability by recognizing m7G-cap in RNAs to enhance cell proliferation [68]. WDR4 mutation and METTL1 dysfunction are closely related to abnormal cell self-renewal and neural differentiation. These regulators have been reported to participate in various neural developmental diseases, including microcephalic primordial dwarfism, Down syndrome, and multiple sclerosis [14]. As observed in Embryonic Stem cells, METTL1 alteration significantly affected the m7G tRNA methylome. METTL1 knockdown dramatically affected tRNA function, mRNA translation, and ribosome pausing in the “RAGGU” motif region [69]. Global m7G profile in pluripotent stem cells (hiPSCs) reveals that reduced m7G modification interferes with pluripotency by promoting embryoid body (EB) formation and slower cell cycling and finally results in mesoderm differentiation [34].
Additionally, m7G modification regulates angiogenesis. It has been reported to be associated with ischemic disorders, idiopathic pulmonary fibrosis, and pulmonary arterial hypertension [70]. There was a general decrease in m7G modification inside mRNA during ischemia. However, in post-ischemic injury, the involvement of METTL1 promotes the proliferation, migration, and tube formation of HUVECs by the m7G-VEGFA pathway, resulting in the increase of local angiogenesis and the recovery of blood circulation [10]. However, as a new RNA modification, the mechanism for regulating angiogenesis is still unclear. Furthermore, bioinformatics analysis showed that the m7G-related gene signature (NUDT16, NUDT4, CYFIP1, LARP1, and DCP2) was closely related to the progression of heart failure [71]. In addition, WDR4 was listed as one of the eight RNA epigenetic modification-related proteins that affect osteoarthritis disease [72].
Roles of m7G regulators in cancer
As a post-transcriptional RNA modification, there is no uniformity in the role of m7G modification on tumors. Most studies suggest that m7G modification promotes the expression of proto-oncogenes to drive the initiation and progression of tumors [69]. Five m7G-related lncRNAs (LINC00924, LINC00944, LINC00865, LINC00702, and ZFAS1) screened out gastric cancer patients who might benefit from immunotherapy, which provides a reference for the treatment and prognosis prediction of cancer [73]. However, some studies indicate an anti-cancer function of m7G modification in lung and colon cancer [74]. Herein, we briefly summarize the biological roles of m7G-related proteins in various tumors (Table 2 and Figure 3).
Table 2.
The role of m7G regulators in cancer
| Molecule | Cancer | Role in cancer | Molecular Mechanism | Phenotype | Reference | 
|---|---|---|---|---|---|
| RNMT | Breast cancer | + | RNMT-PI3KMUT | Proliferation, migration and tumorigenesis | [104] | 
| RNMT | Glioma | + | B7-H6-c-Myc-RNMT | Proliferation, migration and tumorigenesis | [106] | 
| METTL1/WDR4 | Glioblastoma | + | Arg-TCT-4-1↑/cell cycle | Proliferation, migration and tumorigenesis | [16] | 
| METTL1/WDR4 | Hepatocellular carcinoma | + | PTEN↓-AKT | Proliferation and migration | [136] | 
| + | CCNB1-P53/PI3K-AKT | Proliferation, migration and chemorisistance | [82] | ||
| + | m7G tRNA↑-oncogene mRNA↑ | Proliferation, migration and tumorigenesis | [67] | ||
| + | SLUG/SNAIL and TGF-β2-PMN-MDSC | tumor metastasis after (IRFA) | [85,86] | ||
| METTL1/WDR4 | Cholangio-carcinoma | + | m7G tRNA↑-oncogene mRNA↑ | Proliferation, migration and tumorigenesis | [83] | 
| METTL1/WDR4 | Bladder cancer | + | EGFR↑/EFEMP | Proliferation and migration | [89] | 
| METTL1/WDR4 | Cervical cancer | + | tRNA stability | Chemoresistance | [79] | 
| CBC | Cervical cancer | + | CBC/ARS2-let7 | Proliferation | [68] | 
| METTL1/WDR4 | Neck and head squamous cancer | + | m7G tRNA↑-PI3K-AKT-MTOR | Proliferation, migration and tumorigenesis | [87] | 
| METTL1/WDR4 | Nasopharyngeal carcinoma | + | ARNT-METTL1/WDR4↑-WNT-β-catenin | Proliferation, migration, tumorigenesis and therapeutic resistance | [80] | 
| METTL1/WDR4 | AML | + | Arg-TCT-4-1↑-cell cycle | Proliferation, migration and tumorigenesis | [16] | 
| METTL1/WDR4 | Esophageal squamous cell carcinoma | + | MTORC1/RPTOR-ULK/autophagy | Reduced autophagy | [88] | 
| METTL1/WDR4 | Lung cancer | + | tRNA function | Proliferation, migration and tumorigenesis | [77] | 
| Colon cancer | - | miR-149-3p↓/S100A4↑/p53↑ | Chemoresistance | [74] | |
| METTL1/WDR4 | Lung cancer and colorectal cancer | - | let-7e↓-HMGA2/RAS/MYC↑ | Proliferation, migration and tumorigenesis | [5] | 
| eIF4E | AML | + | NF-κB/eIF4E1↑ and eIF4E3↓ | Proliferation, migration and tumorigenesis | [123] | 
| eIF4E | Prostate cancer | + | MTOR/Mnk1/2-eIF4E↑ | Proliferation, migration, tumorigenesis and therapeutic resistance | [96] | 
| CBC (NCBP1) | Lung cancer | + | NCBP1↑-CUL4B↑ | Proliferation, migration and tumorigenesis | [111] | 
(-) for suppressor gene; (+) for oncogene.
Figure 3.
The role of m7G regulators in human cancers. m7G regulators are associated with various kinds of cancers including hepatocellular carcinoma, intrahepatic cholangiocarcinoma, bladder cancer, prostate cancer, cervical cancer, glioblastoma, pancreatic cancer, acute myeloid leukemia, colon cancer, lung cancer, etc. The schema was prepared with object images from Servier Medical Art (https://smart.servier.com).
METTL1/WDR4 complex
METTL1/WDR4 complex has been widely reported to be a parameter of malignant transformation in multiple malignancies, such as breast cancer, glioblastoma, hepatocellular carcinoma (HCC), intra-hepatic cholangiocarcinoma (IHC), acute myeloid leukemia (AML), head and neck squamous cell carcinoma (HNSCC), Nasopharyngeal carcinoma (NPC), bladder cancer, lung adenocarcinoma, and cutaneous melanoma [75-77]. Deleting the Trm8-Trm82 gene in yeast increases its sensitivity to 5-fluorouracil [78]. In the cervical cancer cell (HeLa cells) and NPC, the human homologous METTL1/WDR4 complex induces cell resistance to the chemotherapeutic drugs (paclitaxel and cisplatin) [79,80]. In HCC, high expression of METTL1 is significantly correlated with serum alpha-fetoprotein (AFP) levels and hematogenous metastasis [81]. WDR4 promotes the epithelial-mesenchymal transition (EMT) and confers resistance to sorafenib and lenvatinib in HCC cells [82]. In HCC and IHC, m7G tRNA and mRNA modification endow higher translation efficiency of oncogenes like EGFR [67,83,84]. Targeting METTL1/WDR4-m7G-SLUG/SNAIL pathway and TGF-β2-PMN-MDSC may provide a compensatory address to tumor metastasis after insufficient radiofrequency ablation (IRFA) [85,86]. In the above cancers, METTL1/WDR4 ultimately affects cell cycle-related genes (such as cyclin A2, cyclin D2, cyclin-dependent kinase 4 (CDK4), CDK6, and CDK8) through mRNA “translatome” regulation, to promote the G1/S phase transition. Meanwhile, other proto-oncogenes are simultaneously affected, including epidermic growth factor receptors (EGFR), High Mobility Group at-hook 2 (HMGA2), ASH2 like (ASH2L), Set Domain Bifurcate d Histone Lysine Method Transfer 1 (SETDB1), Ubiquitin Conferring Enzyme E2 t [16]. Regulated by C-MYC, WDR4 activates the PI3K/AKT pathway and accelerates the ubiquitination-mediated degradation of P53 by enhancing the stability and translation of CCNB1 RNA [82]. In NPC, under the regulation of ARNT, METTL1 promotes EMT through the WNT/β-catenin signaling pathway [80]. In HNSCC, apart from influencing the intracellular interaction between malignant epithelial cells and stromal cells, METTL1/WDR4 also dramatically impacts the immune microenvironment [87]. In esophageal squamous cell carcinoma, METTL1/WDR4 impedes MTORC1 and RPTOR-mediated ULK/autophagy pathway in an m7G-tRNA-related codon-dependent manner [88]. The METTL1-m7G-EGFR/EGF-containing Fibulin Extracellular Matrix Protein (EFEMP1) axis is essential in promoting bladder cancer progression [89]. Besides promoting RNA expression, METTL1 has also been proposed to negatively regulate the tumor suppressor PTEN and activate the AKT pathway in liver cancer [81].
Although METTL1/WDR4 has been reported to be a positive driver in most cancers, it has also been reported to have a negative regulatory effect on certain tumors. For example, in colorectal cancer, the overexpression of METTL1 increases drug sensitivity through the miR-149-3p/S100A4/p53 axis in cisplatin-resistant cell lines [74]. Some research revealed an inhibitory role of METTL1 in lung cancer, in contrast with the previous study. Both in lung and colorectal cancers, it has been reported that METTL1 promotes the m7G methylation of the GG-enriched region in the miRNA precursor to stabilize its secondary structure and inhibit cell migration [5,90-92]. Among the regulated miRNAs, let-7e plays a significant role in inhibiting genes such as HMGA2, RAS, and MYC [5].
eIF4E
The overactivation of m7G “reader” eIF4E was observed in 30% malignancies, like M4/M5 subtypes of AML, pancreatic cancer, and prostate cancer [93,94]. Under the regulation of nuclear factor kappa-B (NF-κB), eIF4E enhances proto-oncogene expression by recognizing m7G-cap [95]. In addition, both PI3K/Akt/mTOR pathway and the Ras/MAPK/MNK1/2 pathway contribute to the phosphorylation of eIF4E to increase its binding capacity to m7G-cap [96]. In AML and bladder cancer, the final impact of eIF4E aggregates on the oncogenes like VEGF, whereas implies a slight effect on the constructive ones, like GAPDH [97]. In addition, there is simultaneous overexpression of eIF4E1 and loss function of eIF4E3 in AML. Different amino acid residue sequence of eIF4E3 makes it bind to m7G-cap by electrostatic force and Van der Waals force, thus playing an antagonistic role against eIF4E1. Overexpressing eIF4E3 results in decreased expression of eIF4E1-regulated proto-oncogenes VEGF, MYC, CCND1, and NBN. In pancreatic ductal adenocarcinoma, the MAPK interacting protein kinases (MNK)/eIF4E pathway was reported to be related to chemotherapy drug resistance [98]. And SOX2 was later discovered to be the downstream target of eIF4E to promote tumor proliferation [99]. In prostate cancer, eIF4E can significantly enhance the transcription of certain genes, such as MYC and BCL-2 [100].
Promyelocytic leukemia (PML) protein was a tumor suppressor in a spectrum of cancers, including acute promyelocytic leukemia (APL) [101]. The really interesting new gene (RING) domain of PML has an overwhelming advantage in binding the m7G-cap of mRNAs to eIF4E by over 100-fold, thus reducing the affinity of eIF4E to m7G-cap [102]. The expression of genes, including cell cycle-related genes (such as cyclin D1), is significantly decreased, and the cell cycle was arrested in the G1/S phase [103]. Consequently, it might be declined that blocking eIF4E-mediated translation initiation may play an antioncogenic role.
Other m7G regulators
RNMT can play a specific role in PIK3CA mutant breast cancer to promote tumor growth and proliferation, yet not in PIK3CA wild-type breast cancer [104]. The transcription factor c-Myc/E2F3 was later demonstrated to participate in the positive regulation of RNMT [105]. In addition, there is positive feedback for RNMT recognizing the 5’-cap structure in its mRNA to promote stability and translation efficiency [55]. Besides, through B7-H6/c-Myc/RNMT pathway, the proliferation level of glioma stem cells is significantly increased [106]. Otherwise, WBSCR22 has been reported to induce hypermethylation phenotype in breast cancer, melanoma, and HCC [107,108]. In colorectal cancer, it was demonstrated to be correlated with tumor stemness [109] and drug resistance [110]. However, the concrete regulatory mechanism has not been clarified. In lung cancer, NCBP1/cullin 4B (CUL4B) is significantly upregulated and is correlated with EMT, tumor cell invasion and migration [111].
To sum up, there have been plenty of reports on the role of METTL1/WDR4 and eIF4E as m7G regulators in cancer. Most experiments have proved that they exert a positive role in promoting tumor progression, except for lung cancer and colon cancer. However, more scientific work needs to be conducted for more cognition on other regulators.
Therapeutic strategy based on m7G
The role of m7G modification in cancer provides new perspectives into exploring effective therapeutic strategies for cancer treatment [112]. Although the importance of METTL1/WDR4 in regulating m7G modification and tumor malignancy is self-evident [112], seldom researches has been on developing METTL1/WDR4-targeted therapy in tumor treatment. However, scientists have constructed a 3D model of the METTL1/WDR4 heterodimeric complex, which is expected to facilitate an understanding of the crucial function residues and the development of bioactive inhibitors [75].
Since eIF4E directs RNA into the cytoplasm and initiates translation, blocking the binding between eIF4E and m7G may exert antioncogenic effects. The efficacy of eIF4E inhibitors has an “addictive” effect; that is, cancer cells with high eIF4E levels are more susceptible to eIF4E inhibitors [107]. This “addiction” effect has also been observed in other known oncogenes, such as Her2 and PI3K [112-114]. Therefore, an increasing number of studies are screening patients with high eIF4E expression for phase I/II clinical trials of eIF4E-specific antisense oligonucleotide (4EASO) in combination with conventional chemotherapeutic agents. The therapeutic effect of 4EAS0 drugs has been confirmed to promote apoptosis and cell cycle arrest in mesothelioma and prostate cancer [115,116]. The application of 4EASO in non-small cell lung cancer significantly overcomes the cancer resistance to gemcitabine [117]. However, another type of 4EASO, LY2275796, does not have significant antitumor efficacy in patients with stage IV tumors [118]. ISIS 183750 is a second-generation antisense oligonucleotide drug. Phase I/II clinical trials indicated that co-administration of ISIS 183750 in irinotecan-refractory colorectal cancer reduced the therapeutic dosage to 160 mg/m2 twice weekly [119]. However, whether it can be further applied in clinical practice still needs more laboratory and clinical experiments.
Ribavirin, a commonly used antiviral drug, has been recently found to have antitumor effects as an m7G cap analog that inhibits cell proliferation [120]. The efficacy of ribavirin in AML also depends on the level of eIF4E expression. In tumors with high eIF4E levels (i.e., M4/M5 subtypes of AML), ribavirin has a significant impediment effect. In contrast, it has a limited impact on low-expressing cells (i.e., normal myeloid cells and M1/M2 AML) [112]. Six clinical trials have been conducted to explore the therapeutic effects of ribavirin in cancer treatment with or without combined adjuvant chemotherapy [121]. These clinical trials cover solid tumors, including advanced prostate cancer, head and neck squamous cell carcinoma, recurrent hepatocellular carcinoma, and hematologic malignancies such as AML and lymphoma (ClinicalTrials.gov). A phase II clinical trial in AML showed that ribavirin resulted in clinical remission in 3/11 patients, stabilization in 4/11 patients, and progression in 3/11 patients (NCT00559091). Another clinical trial showed that the combination of ribavirin and low-dose Ara-C was therapeutically effective, but patients eventually met unavoidable ribavirin resistance [122]. Another mechanism of ribavirin is to prevent eIF4E binding to importin 8 and reduce eIF4E nuclear localization. Thus, the accumulated eIF4E in the cytoplasm is later transported back to the nucleus after withdrawal of the drug, making it a possible cause of disease relapse and recrudescence [123]. Therefore, combined inhibition of importin 8 may help to overcome tumor resistance [53]. Recently, studies have found that the active metabolite of ribavirin, ribavirin triphosphate (RTP), could bind eIF4E in a concentration-dependent manner, and its affinity is close to the upper limit of m7G. These studies show that RTP can re-associate to eIF4E at the cap-binding site [124]. In summary, ribavirin has potent anti-cancer and chemo-sensitizing effects. However, the clinical indications for ribavirin application and drug resistance are still urgent issues to be addressed [125].
Conclusion and perspective
RNA epigenetics is a heated spot in scientific research [126]. The m7G modification regulates oncogenes on the post-transcriptional and translational levels efficiently. Dysregulation of m7G modification contributes to tumor pathogenesis and progression. Attention to m7G modification leads to a deeper understanding of its biofunction and underlying molecular mechanism [127]. However, our knowledge of it is far from enough. The exact “erasers” for the m7G modification process remained a mystery. The mechanisms of m7G modification in tumors could be interpreted into two main aspects, one is METTL1/WDR4-mediated m7G modification within tRNAs and mRNAs, and another is eIF4E-related recognition of m7G-cap in the 5’-terminal on mRNAs. m7G-lncRNAs participate in the disease progression in gastric cancer, thyroid cancer, hepatocellular carcinoma, endometrial carcinoma and cutaneous melanoma [76,128-130]. Unfortunately, the role of m7G modification in other RNAs and other m7G modification sites in cancer has few reports. Apart from regulating tumor cells, the impact of m7G modification on tumor microenvironment and tumor immunity is unveiled by multi-omics data [131,132], yet the underlying mechanism requires further investigation. The function of m7G modification varies in tumors. Except for colon cancer and lung cancer, m7G modification generally promotes tumor progression. There are also several clinical studies that intend to investigate the antitumor effect of 4EASO and m7G-analogue, ribavirin. So far, most of them have achieved encouraging results, but also demonstrate the emergence of drug resistance issues. In the end, we look forward that more m7G-related drugs would be applied in cancer therapy practice and drug resistance would be conquered.
Acknowledgements
Kingdraw is used for mapping Figure 1. Servier Medical Art (http://smart.servier.com/) is used for drawing Figure 3, licensed under a Creative Common Attribution 3.0 Generic License (https://creativecommons.org/licenses/by/3.0/). The authors gratefully acknowledge the financial support by the National Natural Science Foundation of China (82203216), the Natural Science Foundation of Jiangsu Province (BK20220724) and Postgraduate Research & Practice Innovation Program of Jiangsu Province (SJCX22-0682).
Disclosure of conflict of interest
None.
References
- 1.Deng X, Su R, Weng H, Huang H, Li Z, Chen J. RNA N(6)-methyladenosine modification in cancers: current status and perspectives. Cell Res. 2018;28:507–517. doi: 10.1038/s41422-018-0034-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Zhang Z, Park E, Lin L, Xing Y. A panoramic view of RNA modifications: exploring new frontiers. Genome Biol. 2018;19:11. doi: 10.1186/s13059-018-1394-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Juhling F, Morl M, Hartmann RK, Sprinzl M, Stadler PF, Putz J. tRNAdb 2009: compilation of tRNA sequences and tRNA genes. Nucleic Acids Res. 2009;37:D159–162. doi: 10.1093/nar/gkn772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Monecke T, Dickmanns A, Ficner R. Structural basis for m7G-cap hypermethylation of small nuclear, small nucleolar and telomerase RNA by the dimethyltransferase TGS1. Nucleic Acids Res. 2009;37:3865–3877. doi: 10.1093/nar/gkp249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pandolfini L, Barbieri I, Bannister AJ, Hendrick A, Andrews B, Webster N, Murat P, Mach P, Brandi R, Robson SC, Migliori V, Alendar A, d’Onofrio M, Balasubramanian S, Kouzarides T. METTL1 promotes let-7 microRNA processing via m7G methylation. Mol Cell. 2019;74:1278–1290. e1279. doi: 10.1016/j.molcel.2019.03.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Dimitrova DG, Teysset L, Carre C. RNA 2’-O-methylation (Nm) modification in human diseases. Genes (Basel) 2019;10:117. doi: 10.3390/genes10020117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ramanathan A, Robb GB, Chan SH. mRNA capping: biological functions and applications. Nucleic Acids Res. 2016;44:7511–7526. doi: 10.1093/nar/gkw551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Yedavalli VS, Jeang KT. Trimethylguanosine capping selectively promotes expression of Rev-dependent HIV-1 RNAs. Proc Natl Acad Sci U S A. 2010;107:14787–14792. doi: 10.1073/pnas.1009490107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lin S, Liu Q, Jiang YZ, Gregory RI. Nucleotide resolution profiling of m(7)G tRNA modification by TRAC-Seq. Nat Protoc. 2019;14:3220–3242. doi: 10.1038/s41596-019-0226-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zhao Y, Kong L, Pei Z, Li F, Li C, Sun X, Shi B, Ge J. m7G methyltransferase METTL1 promotes post-ischemic angiogenesis via promoting VEGFA mRNA translation. Front Cell Dev Biol. 2021;9:642080. doi: 10.3389/fcell.2021.642080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhang LS, Liu C, Ma H, Dai Q, Sun HL, Luo G, Zhang Z, Zhang L, Hu L, Dong X, He C. Transcriptome-wide mapping of internal N(7)-methylguanosine methylome in mammalian mRNA. Mol Cell. 2019;74:1304–1316. e1308. doi: 10.1016/j.molcel.2019.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Malbec L, Zhang T, Chen YS, Zhang Y, Sun BF, Shi BY, Zhao YL, Yang Y, Yang YG. Dynamic methylome of internal mRNA N(7)-methylguanosine and its regulatory role in translation. Cell Res. 2019;29:927–941. doi: 10.1038/s41422-019-0230-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Thongdee N, Jaroensuk J, Atichartpongkul S, Chittrakanwong J, Chooyoung K, Srimahaeak T, Chaiyen P, Vattanaviboon P, Mongkolsuk S, Fuangthong M. TrmB, a tRNA m7G46 methyltransferase, plays a role in hydrogen peroxide resistance and positively modulates the translation of katA and katB mRNAs in Pseudomonas aeruginosa. Nucleic Acids Res. 2019;47:9271–9281. doi: 10.1093/nar/gkz702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Xia X, Wang Y, Zheng JC. Internal m7G methylation: a novel epitranscriptomic contributor in brain development and diseases. Mol Ther Nucleic Acids. 2023;31:295–308. doi: 10.1016/j.omtn.2023.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Katsara O, Schneider RJ. m(7)G tRNA modification reveals new secrets in the translational regulation of cancer development. Mol Cell. 2021;81:3243–3245. doi: 10.1016/j.molcel.2021.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Orellana EA, Liu Q, Yankova E, Pirouz M, De Braekeleer E, Zhang W, Lim J, Aspris D, Sendinc E, Garyfallos DA, Gu M, Ali R, Gutierrez A, Mikutis S, Bernardes GJL, Fischer ES, Bradley A, Vassiliou GS, Slack FJ, Tzelepis K, Gregory RI. METTL1-mediated m(7)G modification of Arg-TCT tRNA drives oncogenic transformation. Mol Cell. 2021;81:3323–3338. e3314. doi: 10.1016/j.molcel.2021.06.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dubin DT, Taylor RH. The methylation state of poly A-containing messenger RNA from cultured hamster cells. Nucleic Acids Res. 1975;2:1653–1668. doi: 10.1093/nar/2.10.1653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Muthukrishnan S, Both GW, Furuichi Y, Shatkin AJ. 5’-terminal 7-methylguanosine in eukaryotic mRNA is required for translation. Nature. 1975;255:33–37. doi: 10.1038/255033a0. [DOI] [PubMed] [Google Scholar]
- 19.Hefti E, Bishop DH, Dubin DT, Stollar V. 5’ nucleotide sequence of sindbis viral RNA. J Virol. 1975;17:149–159. doi: 10.1128/jvi.17.1.149-159.1976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ensinger MJ, Martin SA, Paoletti E, Moss B. Modification of the 5’-terminus of mRNA by soluble guanylyl and methyl transferases from vaccinia virus. Proc Natl Acad Sci U S A. 1975;72:2525–2529. doi: 10.1073/pnas.72.7.2525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Silverman S, Gillam IC, Tener GM, Soll D. The nucleotide sequence of lysine tRNA2 from Drosophila. Nucleic Acids Res. 1979;6:435–442. doi: 10.1093/nar/6.2.435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wei C, Moss B. 5’-terminal capping of RNA by guanylyltransferase from HeLa cell nuclei. Proc Natl Acad Sci U S A. 1977;74:3758–3761. doi: 10.1073/pnas.74.9.3758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Rainen L, Stollar BD. Antibodies distinguishing between intact and alkali-hydrolyzed 7-methylguanosine. Nucleic Acids Res. 1978;5:4877–4889. doi: 10.1093/nar/5.12.4877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ueda H, Iyo H, Doi M, Inoue M, Ishida T, Morioka H, Tanaka T, Nishikawa S, Uesugi S. Combination of Trp and Glu residues for recognition of mRNA cap structure. Analysis of m7G base recognition site of human cap binding protein (IF-4E) by site-directed mutagenesis. FEBS Lett. 1991;280:207–210. doi: 10.1016/0014-5793(91)80294-d. [DOI] [PubMed] [Google Scholar]
- 25.Goldson TM, Vielhauer G, Staub E, Miller S, Shim H, Hagedorn CH. Eukaryotic initiation factor 4E variants alter the morphology, proliferation, and colony-formation properties of MDA-MB-435 cancer cells. Mol Carcinog. 2007;46:71–84. doi: 10.1002/mc.20276. [DOI] [PubMed] [Google Scholar]
- 26.Vinther J. No evidence for N7-methylation of guanosine (m(7)G) in human let-7e. Mol Cell. 2020;79:199–200. doi: 10.1016/j.molcel.2020.05.022. [DOI] [PubMed] [Google Scholar]
- 27.Kouzarides T, Pandolfini L, Barbieri I, Bannister AJ, Andrews B. Further evidence supporting N7-methylation of guanosine (m(7)G) in human microRNAs. Mol Cell. 2020;79:201–202. doi: 10.1016/j.molcel.2020.05.023. [DOI] [PubMed] [Google Scholar]
- 28.Ahn HJ, Kim HW, Yoon HJ, Lee BI, Suh SW, Yang JK. Crystal structure of tRNA(m1G37)methyltransferase: insights into tRNA recognition. EMBO J. 2003;22:2593–2603. doi: 10.1093/emboj/cdg269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Leulliot N, Chaillet M, Durand D, Ulryck N, Blondeau K, van Tilbeurgh H. Structure of the yeast tRNA m7G methylation complex. Structure. 2008;16:52–61. doi: 10.1016/j.str.2007.10.025. [DOI] [PubMed] [Google Scholar]
- 30.Boccaletto P, Machnicka MA, Purta E, Piatkowski P, Baginski B, Wirecki TK, de Crecy-Lagard V, Ross R, Limbach PA, Kotter A, Helm M, Bujnicki JM. MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Res. 2018;46:D303–D307. doi: 10.1093/nar/gkx1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Stange N, Beier H. A cell-free plant extract for accurate pre-tRNA processing, splicing and modification. EMBO J. 1987;6:2811–2818. doi: 10.1002/j.1460-2075.1987.tb02577.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Alexandrov A, Grayhack EJ, Phizicky EM. tRNA m7G methyltransferase Trm8p/Trm82p: evidence linking activity to a growth phenotype and implicating Trm82p in maintaining levels of active Trm8p. RNA. 2005;11:821–830. doi: 10.1261/rna.2030705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Boulias K, Greer EL. Put the pedal to the METTL1: adding internal m(7)G increases mRNA translation efficiency and augments miRNA processing. Mol Cell. 2019;74:1105–1107. doi: 10.1016/j.molcel.2019.06.004. [DOI] [PubMed] [Google Scholar]
- 34.Deng Y, Zhou Z, Ji W, Lin S, Wang M. METTL1-mediated m(7)G methylation maintains pluripotency in human stem cells and limits mesoderm differentiation and vascular development. Stem Cell Res Ther. 2020;11:306. doi: 10.1186/s13287-020-01814-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Li J, Wang L, Hahn Q, Nowak RP, Viennet T, Orellana EA, Roy Burman SS, Yue H, Hunkeler M, Fontana P, Wu H, Arthanari H, Fischer ES, Gregory RI. Structural basis of regulated m(7)G tRNA modification by METTL1-WDR4. Nature. 2023;613:391–397. doi: 10.1038/s41586-022-05566-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ruiz-Arroyo VM, Raj R, Babu K, Onolbaatar O, Roberts PH, Nam Y. Structures and mechanisms of tRNA methylation by METTL1-WDR4. Nature. 2023;613:383–390. doi: 10.1038/s41586-022-05565-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Pillutla RC, Shimamoto A, Furuichi Y, Shatkin AJ. Human mRNA capping enzyme (RNGTT) and cap methyltransferase (RNMT) map to 6q16 and 18p11.22-p11.23, respectively. Genomics. 1998;54:351–353. doi: 10.1006/geno.1998.5604. [DOI] [PubMed] [Google Scholar]
- 38.Aregger M, Kaskar A, Varshney D, Fernandez-Sanchez ME, Inesta-Vaquera FA, Weidlich S, Cowling VH. CDK1-cyclin B1 activates RNMT, coordinating mRNA cap methylation with G1 phase transcription. Mol Cell. 2016;61:734–746. doi: 10.1016/j.molcel.2016.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ohira T, Suzuki T. Precursors of tRNAs are stabilized by methylguanosine cap structures. Nat Chem Biol. 2016;12:648–655. doi: 10.1038/nchembio.2117. [DOI] [PubMed] [Google Scholar]
- 40.Beauclerk AA, Cundliffe E. Sites of action of two ribosomal RNA methylases responsible for resistance to aminoglycosides. J Mol Biol. 1987;193:661–671. doi: 10.1016/0022-2836(87)90349-4. [DOI] [PubMed] [Google Scholar]
- 41.Letoquart J, Huvelle E, Wacheul L, Bourgeois G, Zorbas C, Graille M, Heurgue-Hamard V, Lafontaine DL. Structural and functional studies of Bud23-Trm112 reveal 18S rRNA N7-G1575 methylation occurs on late 40S precursor ribosomes. Proc Natl Acad Sci U S A. 2014;111:E5518–5526. doi: 10.1073/pnas.1413089111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.White J, Li Z, Sardana R, Bujnicki JM, Marcotte EM, Johnson AW. Bud23 methylates G1575 of 18S rRNA and is required for efficient nuclear export of pre-40S subunits. Mol Cell Biol. 2008;28:3151–3161. doi: 10.1128/MCB.01674-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sloan KE, Warda AS, Sharma S, Entian KD, Lafontaine DLJ, Bohnsack MT. Tuning the ribosome: the influence of rRNA modification on eukaryotic ribosome biogenesis and function. RNA Biol. 2017;14:1138–1152. doi: 10.1080/15476286.2016.1259781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Haag S, Kretschmer J, Bohnsack MT. WBSCR22/Merm1 is required for late nuclear pre-ribosomal RNA processing and mediates N7-methylation of G1639 in human 18S rRNA. RNA. 2015;21:180–187. doi: 10.1261/rna.047910.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ounap K, Kasper L, Kurg A, Kurg R. The human WBSCR22 protein is involved in the biogenesis of the 40S ribosomal subunits in mammalian cells. PLoS One. 2013;8:e75686. doi: 10.1371/journal.pone.0075686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wang KT, Desmolaize B, Nan J, Zhang XW, Li LF, Douthwaite S, Su XD. Structure of the bifunctional methyltransferase YcbY (RlmKL) that adds the m7G2069 and m2G2445 modifications in Escherichia coli 23S rRNA. Nucleic Acids Res. 2012;40:5138–5148. doi: 10.1093/nar/gks160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dai XY, Shi L, Li Z, Yang HY, Wei JF, Ding Q. Main N6-methyladenosine readers: YTH family proteins in cancers. Front Oncol. 2021;11:635329. doi: 10.3389/fonc.2021.635329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Yang X, Yang Y, Sun BF, Chen YS, Xu JW, Lai WY, Li A, Wang X, Bhattarai DP, Xiao W, Sun HY, Zhu Q, Ma HL, Adhikari S, Sun M, Hao YJ, Zhang B, Huang CM, Huang N, Jiang GB, Zhao YL, Wang HL, Sun YP, Yang YG. 5-methylcytosine promotes mRNA export - NSUN2 as the methyltransferase and ALYREF as an m(5)C reader. Cell Res. 2017;27:606–625. doi: 10.1038/cr.2017.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Gonatopoulos-Pournatzis T, Cowling VH. Cap-binding complex (CBC) Biochem J. 2014;457:231–242. doi: 10.1042/BJ20131214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Ohno M, Segref A, Bachi A, Wilm M, Mattaj IW. PHAX, a mediator of U snRNA nuclear export whose activity is regulated by phosphorylation. Cell. 2000;101:187–198. doi: 10.1016/S0092-8674(00)80829-6. [DOI] [PubMed] [Google Scholar]
- 51.Brown CJ, McNae I, Fischer PM, Walkinshaw MD. Crystallographic and mass spectrometric characterisation of eIF4E with N7-alkylated cap derivatives. J Mol Biol. 2007;372:7–15. doi: 10.1016/j.jmb.2007.06.033. [DOI] [PubMed] [Google Scholar]
- 52.Topisirovic I, Svitkin YV, Sonenberg N, Shatkin AJ. Cap and cap-binding proteins in the control of gene expression. Wiley Interdiscip Rev RNA. 2011;2:277–298. doi: 10.1002/wrna.52. [DOI] [PubMed] [Google Scholar]
- 53.Volpon L, Culjkovic-Kraljacic B, Osborne MJ, Ramteke A, Sun Q, Niesman A, Chook YM, Borden KL. Importin 8 mediates m7G cap-sensitive nuclear import of the eukaryotic translation initiation factor eIF4E. Proc Natl Acad Sci U S A. 2016;113:5263–5268. doi: 10.1073/pnas.1524291113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Palacios I, Hetzer M, Adam SA, Mattaj IW. Nuclear import of U snRNPs requires importin beta. EMBO J. 1997;16:6783–6792. doi: 10.1093/emboj/16.22.6783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Osborne MJ, Volpon L, Memarpoor-Yazdi M, Pillay S, Thambipillai A, Czarnota S, Culjkovic-Kraljacic B, Trahan C, Oeffinger M, Cowling VH, Borden KLB. Identification and characterization of the interaction between the methyl-7-guanosine cap maturation enzyme RNMT and the cap-binding protein eIF4E. J Mol Biol. 2022;434:167451. doi: 10.1016/j.jmb.2022.167451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Drummond DR, Armstrong J, Colman A. The effect of capping and polyadenylation on the stability, movement and translation of synthetic messenger RNAs in Xenopus oocytes. Nucleic Acids Res. 1985;13:7375–7394. doi: 10.1093/nar/13.20.7375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Chen L, Roake CM, Galati A, Bavasso F, Micheli E, Saggio I, Schoeftner S, Cacchione S, Gatti M, Artandi SE, Raffa GD. Loss of human TGS1 hypermethylase promotes increased telomerase RNA and telomere elongation. Cell Rep. 2020;30:1358–1372. e1355. doi: 10.1016/j.celrep.2020.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hamm J, Mattaj IW. Monomethylated cap structures facilitate RNA export from the nucleus. Cell. 1990;63:109–118. doi: 10.1016/0092-8674(90)90292-m. [DOI] [PubMed] [Google Scholar]
- 59.Dewe JM, Whipple JM, Chernyakov I, Jaramillo LN, Phizicky EM. The yeast rapid tRNA decay pathway competes with elongation factor 1A for substrate tRNAs and acts on tRNAs lacking one or more of several modifications. RNA. 2012;18:1886–1896. doi: 10.1261/rna.033654.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bahr A, Hankeln T, Fiedler T, Hegemann J, Schmidt ER. Molecular analysis of METTL1, a novel human methyltransferase-like gene with a high degree of phylogenetic conservation. Genomics. 1999;57:424–428. doi: 10.1006/geno.1999.5780. [DOI] [PubMed] [Google Scholar]
- 61.Shi H, Moore PB. The crystal structure of yeast phenylalanine tRNA at 1.93 A resolution: a classic structure revisited. RNA. 2000;6:1091–1105. doi: 10.1017/s1355838200000364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Alexandrov A, Chernyakov I, Gu W, Hiley SL, Hughes TR, Grayhack EJ, Phizicky EM. Rapid tRNA decay can result from lack of nonessential modifications. Mol Cell. 2006;21:87–96. doi: 10.1016/j.molcel.2005.10.036. [DOI] [PubMed] [Google Scholar]
- 63.Wang Z, Kiledjian M. Functional link between the mammalian exosome and mRNA decapping. Cell. 2001;107:751–762. doi: 10.1016/s0092-8674(01)00592-x. [DOI] [PubMed] [Google Scholar]
- 64.Shimotohno K, Kodama Y, Hashimoto J, Miura KI. Importance of 5’-terminal blocking structure to stabilize mRNA in eukaryotic protein synthesis. Proc Natl Acad Sci U S A. 1977;74:2734–2738. doi: 10.1073/pnas.74.7.2734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Culjkovic B, Topisirovic I, Skrabanek L, Ruiz-Gutierrez M, Borden KL. eIF4E is a central node of an RNA regulon that governs cellular proliferation. J Cell Biol. 2006;175:415–426. doi: 10.1083/jcb.200607020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Jang SK, Paek KY. Cap-dependent translation is mediated by ‘RNA looping’ rather than ‘ribosome scanning’. RNA Biol. 2016;13:1–5. doi: 10.1080/15476286.2015.1107700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Chen Z, Zhu W, Zhu S, Sun K, Liao J, Liu H, Dai Z, Han H, Ren X, Yang Q, Zheng S, Peng B, Peng S, Kuang M, Lin S. METTL1 promotes hepatocarcinogenesis via m(7) G tRNA modification-dependent translation control. Clin Transl Med. 2021;11:e661. doi: 10.1002/ctm2.661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Gruber JJ, Zatechka DS, Sabin LR, Yong J, Lum JJ, Kong M, Zong WX, Zhang Z, Lau CK, Rawlings J, Cherry S, Ihle JN, Dreyfuss G, Thompson CB. Ars2 links the nuclear cap-binding complex to RNA interference and cell proliferation. Cell. 2009;138:328–339. doi: 10.1016/j.cell.2009.04.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lin S, Liu Q, Lelyveld VS, Choe J, Szostak JW, Gregory RI. Mettl1/Wdr4-mediated m(7)G tRNA methylome is required for normal mRNA translation and embryonic stem cell self-renewal and differentiation. Mol Cell. 2018;71:244–255. e245. doi: 10.1016/j.molcel.2018.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Huang T, He WY. Construction and validation of a novel prognostic signature of idiopathic pulmonary fibrosis by identifying subtypes based on genes related to 7-methylguanosine modification. Front Genet. 2022;13:890530. doi: 10.3389/fgene.2022.890530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ma C, Tu D, Xu Q, Wu Y, Song X, Guo Z, Zhao X. Identification of m(7)G regulator-mediated RNA methylation modification patterns and related immune microenvironment regulation characteristics in heart failure. Clin Epigenetics. 2023;15:22. doi: 10.1186/s13148-023-01439-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Chen Z, Wang W, Hua Y. Expression patterns of eight RNA-modified regulators correlating with immune infiltrates during the progression of osteoarthritis. Front Immunol. 2023;14:1019445. doi: 10.3389/fimmu.2023.1019445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ma M, Li J, Zeng Z, Zheng Z, Kang W. Integrated analysis from multicentre studies identities m7G-related lncRNA-derived molecular subtypes and risk stratification systems for gastric cancer. Front Immunol. 2023;14:1096488. doi: 10.3389/fimmu.2023.1096488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Liu Y, Yang C, Zhao Y, Chi Q, Wang Z, Sun B. Overexpressed methyltransferase-like 1 (METTL1) increased chemosensitivity of colon cancer cells to cisplatin by regulating miR-149-3p/S100A4/p53 axis. Aging (Albany NY) 2019;11:12328–12344. doi: 10.18632/aging.102575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Campeanu IJ, Jiang Y, Liu L, Pilecki M, Najor A, Cobani E, Manning M, Zhang XM, Yang ZQ. Multi-omics integration of methyltransferase-like protein family reveals clinical outcomes and functional signatures in human cancer. Sci Rep. 2021;11:14784. doi: 10.1038/s41598-021-94019-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Rong J, Wang H, Yao Y, Wu Z, Chen L, Jin C, Shi Z, Wu C, Hu X. Identification of m7G-associated lncRNA prognostic signature for predicting the immune status in cutaneous melanoma. Aging (Albany NY) 2022;14:5233–5249. doi: 10.18632/aging.204151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ma J, Han H, Huang Y, Yang C, Zheng S, Cai T, Bi J, Huang X, Liu R, Huang L, Luo Y, Li W, Lin S. METTL1/WDR4-mediated m(7)G tRNA modifications and m(7)G codon usage promote mRNA translation and lung cancer progression. Mol Ther. 2021;29:3422–3435. doi: 10.1016/j.ymthe.2021.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Gustavsson M, Ronne H. Evidence that tRNA modifying enzymes are important in vivo targets for 5-fluorouracil in yeast. RNA. 2008;14:666–674. doi: 10.1261/rna.966208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Okamoto M, Fujiwara M, Hori M, Okada K, Yazama F, Konishi H, Xiao Y, Qi G, Shimamoto F, Ota T, Temme A, Tatsuka M. tRNA modifying enzymes, NSUN2 and METTL1, determine sensitivity to 5-fluorouracil in HeLa cells. PLoS Genet. 2014;10:e1004639. doi: 10.1371/journal.pgen.1004639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chen B, Jiang W, Huang Y, Zhang J, Yu P, Wu L, Peng H. N(7)-methylguanosine tRNA modification promotes tumorigenesis and chemoresistance through WNT/beta-catenin pathway in nasopharyngeal carcinoma. Oncogene. 2022;41:2239–2253. doi: 10.1038/s41388-022-02250-9. [DOI] [PubMed] [Google Scholar]
- 81.Tian QH, Zhang MF, Zeng JS, Luo RG, Wen Y, Chen J, Gan LG, Xiong JP. METTL1 overexpression is correlated with poor prognosis and promotes hepatocellular carcinoma via PTEN. J Mol Med (Berl) 2019;97:1535–1545. doi: 10.1007/s00109-019-01830-9. [DOI] [PubMed] [Google Scholar]
- 82.Xia P, Zhang H, Xu K, Jiang X, Gao M, Wang G, Liu Y, Yao Y, Chen X, Ma W, Zhang Z, Yuan Y. MYC-targeted WDR4 promotes proliferation, metastasis, and sorafenib resistance by inducing CCNB1 translation in hepatocellular carcinoma. Cell Death Dis. 2021;12:691. doi: 10.1038/s41419-021-03973-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Dai Z, Liu H, Liao J, Huang C, Ren X, Zhu W, Zhu S, Peng B, Li S, Lai J, Liang L, Xu L, Peng S, Lin S, Kuang M. N(7)-methylguanosine tRNA modification enhances oncogenic mRNA translation and promotes intrahepatic cholangiocarcinoma progression. Mol Cell. 2021;81:3339–3355. e3338. doi: 10.1016/j.molcel.2021.07.003. [DOI] [PubMed] [Google Scholar]
- 84.Huang M, Long J, Yao Z, Zhao Y, Zhao Y, Liao J, Lei K, Xiao H, Dai Z, Peng S, Lin S, Xu L, Kuang M. METTL1-mediated m7G tRNA modification promotes lenvatinib resistance in hepatocellular carcinoma. Cancer Res. 2023;83:89–102. doi: 10.1158/0008-5472.CAN-22-0963. [DOI] [PubMed] [Google Scholar]
- 85.Zhu S, Wu Y, Zhang X, Peng S, Xiao H, Chen S, Xu L, Su T, Kuang M. Targeting N(7)-methylguanosine tRNA modification blocks hepatocellular carcinoma metastasis after insufficient radiofrequency ablation. Mol Ther. 2022 doi: 10.1016/j.ymthe.2022.08.004. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zeng X, Liao G, Li S, Liu H, Zhao X, Li S, Lei K, Zhu S, Chen Z, Zhao Y, Ren X, Su T, Cheng AS, Peng S, Lin S, Wang J, Chen S, Kuang M. Eliminating METTL1-mediated accumulation of PMN-MDSCs prevents hepatocellular carcinoma recurrence after radiofrequency ablation. Hepatology. 2023;77:1122–1138. doi: 10.1002/hep.32585. [DOI] [PubMed] [Google Scholar]
- 87.Chen J, Li K, Chen J, Wang X, Ling R, Cheng M, Chen Z, Chen F, He Q, Li S, Zhang C, Jiang Y, Chen Q, Wang A, Chen D. Aberrant translation regulated by METTL1/WDR4-mediated tRNA N7-methylguanosine modification drives head and neck squamous cell carcinoma progression. Cancer Commun (Lond) 2022;42:223–244. doi: 10.1002/cac2.12273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Han H, Yang C, Ma J, Zhang S, Zheng S, Ling R, Sun K, Guo S, Huang B, Liang Y, Wang L, Chen S, Wang Z, Wei W, Huang Y, Peng H, Jiang YZ, Choe J, Lin S. N(7)-methylguanosine tRNA modification promotes esophageal squamous cell carcinoma tumorigenesis via the RPTOR/ULK1/autophagy axis. Nat Commun. 2022;13:1478. doi: 10.1038/s41467-022-29125-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ying X, Liu B, Yuan Z, Huang Y, Chen C, Jiang X, Zhang H, Qi D, Yang S, Lin S, Luo J, Ji W. METTL1-m(7) G-EGFR/EFEMP1 axis promotes the bladder cancer development. Clin Transl Med. 2021;11:e675. doi: 10.1002/ctm2.675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Mirihana Arachchilage G, Dassanayake AC, Basu S. A potassium ion-dependent RNA structural switch regulates human pre-miRNA 92b maturation. Chem Biol. 2015;22:262–272. doi: 10.1016/j.chembiol.2014.12.013. [DOI] [PubMed] [Google Scholar]
- 91.Rouleau SG, Garant JM, Bolduc F, Bisaillon M, Perreault JP. G-Quadruplexes influence pri-microRNA processing. RNA Biol. 2018;15:198–206. doi: 10.1080/15476286.2017.1405211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Pandey S, Agarwala P, Jayaraj GG, Gargallo R, Maiti S. The RNA stem-loop to G-quadruplex equilibrium controls mature microRNA production inside the cell. Biochemistry. 2015;54:7067–7078. doi: 10.1021/acs.biochem.5b00574. [DOI] [PubMed] [Google Scholar]
- 93.Topisirovic I, Guzman ML, McConnell MJ, Licht JD, Culjkovic B, Neering SJ, Jordan CT, Borden KL. Aberrant eukaryotic translation initiation factor 4E-dependent mRNA transport impedes hematopoietic differentiation and contributes to leukemogenesis. Mol Cell Biol. 2003;23:8992–9002. doi: 10.1128/MCB.23.24.8992-9002.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ruggero D, Pandolfi PP. Does the ribosome translate cancer? Nat Rev Cancer. 2003;3:179–192. doi: 10.1038/nrc1015. [DOI] [PubMed] [Google Scholar]
- 95.Hariri F, Arguello M, Volpon L, Culjkovic-Kraljacic B, Nielsen TH, Hiscott J, Mann KK, Borden KL. The eukaryotic translation initiation factor eIF4E is a direct transcriptional target of NF-kappaB and is aberrantly regulated in acute myeloid leukemia. Leukemia. 2013;27:2047–2055. doi: 10.1038/leu.2013.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.D’Abronzo LS, Ghosh PM. eIF4E phosphorylation in prostate cancer. Neoplasia. 2018;20:563–573. doi: 10.1016/j.neo.2018.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Swaminathan M, Wang ES. Novel therapies for AML: a round-up for clinicians. Expert Rev Clin Pharmacol. 2020;13:1389–1400. doi: 10.1080/17512433.2020.1850255. [DOI] [PubMed] [Google Scholar]
- 98.Adesso L, Calabretta S, Barbagallo F, Capurso G, Pilozzi E, Geremia R, Delle Fave G, Sette C. Gemcitabine triggers a pro-survival response in pancreatic cancer cells through activation of the MNK2/eIF4E pathway. Oncogene. 2013;32:2848–2857. doi: 10.1038/onc.2012.306. [DOI] [PubMed] [Google Scholar]
- 99.Yu Y, Tian L, Feng X, Cheng J, Gong Y, Liu X, Zhang Z, Yang X, He S, Li CY, Huang Q. eIF4E-phosphorylation-mediated Sox2 upregulation promotes pancreatic tumor cell repopulation after irradiation. Cancer Lett. 2016;375:31–38. doi: 10.1016/j.canlet.2016.02.052. [DOI] [PubMed] [Google Scholar]
- 100.Yu D, Scott C, Jia WW, De Benedetti A, Williams BJ, Fazli L, Wen Y, Gleave M, Nelson C, Rennie PS. Targeting and killing of prostate cancer cells using lentiviral constructs containing a sequence recognized by translation factor eIF4E and a prostate-specific promoter. Cancer Gene Ther. 2006;13:32–43. doi: 10.1038/sj.cgt.7700885. [DOI] [PubMed] [Google Scholar]
- 101.Rego EM, Wang ZG, Peruzzi D, He LZ, Cordon-Cardo C, Pandolfi PP. Role of promyelocytic leukemia (PML) protein in tumor suppression. J Exp Med. 2001;193:521–529. doi: 10.1084/jem.193.4.521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Cohen N, Sharma M, Kentsis A, Perez JM, Strudwick S, Borden KL. PML RING suppresses oncogenic transformation by reducing the affinity of eIF4E for mRNA. EMBO J. 2001;20:4547–4559. doi: 10.1093/emboj/20.16.4547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Borden KL. Pondering the promyelocytic leukemia protein (PML) puzzle: possible functions for PML nuclear bodies. Mol Cell Biol. 2002;22:5259–5269. doi: 10.1128/MCB.22.15.5259-5269.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Dunn S, Lombardi O, Lukoszek R, Cowling VH. Oncogenic PIK3CA mutations increase dependency on the mRNA cap methyltransferase, RNMT, in breast cancer cells. Open Biol. 2019;9:190052. doi: 10.1098/rsob.190052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Cowling VH. Enhanced mRNA cap methylation increases cyclin D1 expression and promotes cell transformation. Oncogene. 2010;29:930–936. doi: 10.1038/onc.2009.368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Chen H, Guo Y, Sun J, Dong J, Bao Q, Zhang X, Fu F. Preferential expression of B7-H6 in glioma stem-like cells enhances tumor cell proliferation via the c-Myc/RNMT axis. J Immunol Res. 2020;2020:2328675. doi: 10.1155/2020/2328675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Nakazawa Y, Arai H, Fujita N. The novel metastasis promoter Merm1/Wbscr22 enhances tumor cell survival in the vasculature by suppressing Zac1/p53-dependent apoptosis. Cancer Res. 2011;71:1146–1155. doi: 10.1158/0008-5472.CAN-10-2695. [DOI] [PubMed] [Google Scholar]
- 108.Stefanska B, Cheishvili D, Suderman M, Arakelian A, Huang J, Hallett M, Han ZG, Al-Mahtab M, Akbar SM, Khan WA, Raqib R, Tanvir I, Khan HA, Rabbani SA, Szyf M. Genome-wide study of hypomethylated and induced genes in patients with liver cancer unravels novel anti-cancer targets. Clin Cancer Res. 2014;20:3118–3132. doi: 10.1158/1078-0432.CCR-13-0283. [DOI] [PubMed] [Google Scholar]
- 109.Zhao H, Su W, Sun Y, Wu Z. WBSCR22 competes with long non-coding RNA Linc00346 for miR-509-5p binding site to regulate cancer stem cell phenotypes of colorectal cancer. Biochem Genet. 2020;58:384–398. doi: 10.1007/s10528-020-09949-y. [DOI] [PubMed] [Google Scholar]
- 110.Yan D, Tu L, Yuan H, Fang J, Cheng L, Zheng X, Wang X. WBSCR22 confers oxaliplatin resistance in human colorectal cancer. Sci Rep. 2017;7:15443. doi: 10.1038/s41598-017-15749-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Zhang H, Wang A, Tan Y, Wang S, Ma Q, Chen X, He Z. NCBP1 promotes the development of lung adenocarcinoma through up-regulation of CUL4B. J Cell Mol Med. 2019;23:6965–6977. doi: 10.1111/jcmm.14581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kentsis A, Topisirovic I, Culjkovic B, Shao L, Borden KL. Ribavirin suppresses eIF4E-mediated oncogenic transformation by physical mimicry of the 7-methyl guanosine mRNA cap. Proc Natl Acad Sci U S A. 2004;101:18105–18110. doi: 10.1073/pnas.0406927102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005;4:988–1004. doi: 10.1038/nrd1902. [DOI] [PubMed] [Google Scholar]
- 114.Xu Q, Simpson SE, Scialla TJ, Bagg A, Carroll M. Survival of acute myeloid leukemia cells requires PI3 kinase activation. Blood. 2003;102:972–980. doi: 10.1182/blood-2002-11-3429. [DOI] [PubMed] [Google Scholar]
- 115.Graff JR, Konicek BW, Lynch RL, Dumstorf CA, Dowless MS, McNulty AM, Parsons SH, Brail LH, Colligan BM, Koop JW, Hurst BM, Deddens JA, Neubauer BL, Stancato LF, Carter HW, Douglass LE, Carter JH. eIF4E activation is commonly elevated in advanced human prostate cancers and significantly related to reduced patient survival. Cancer Res. 2009;69:3866–3873. doi: 10.1158/0008-5472.CAN-08-3472. [DOI] [PubMed] [Google Scholar]
- 116.Graff JR, Konicek BW, Vincent TM, Lynch RL, Monteith D, Weir SN, Schwier P, Capen A, Goode RL, Dowless MS, Chen Y, Zhang H, Sissons S, Cox K, McNulty AM, Parsons SH, Wang T, Sams L, Geeganage S, Douglass LE, Neubauer BL, Dean NM, Blanchard K, Shou J, Stancato LF, Carter JH, Marcusson EG. Therapeutic suppression of translation initiation factor eIF4E expression reduces tumor growth without toxicity. J Clin Invest. 2007;117:2638–2648. doi: 10.1172/JCI32044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Thumma SC, Jacobson BA, Patel MR, Konicek BW, Franklin MJ, Jay-Dixon J, Sadiq A, De A, Graff JR, Kratzke RA. Antisense oligonucleotide targeting eukaryotic translation initiation factor 4E reduces growth and enhances chemosensitivity of non-small-cell lung cancer cells. Cancer Gene Ther. 2015;22:396–401. doi: 10.1038/cgt.2015.34. [DOI] [PubMed] [Google Scholar]
- 118.Hong DS, Kurzrock R, Oh Y, Wheler J, Naing A, Brail L, Callies S, Andre V, Kadam SK, Nasir A, Holzer TR, Meric-Bernstam F, Fishman M, Simon G. A phase 1 dose escalation, pharmacokinetic, and pharmacodynamic evaluation of eIF-4E antisense oligonucleotide LY2275796 in patients with advanced cancer. Clin Cancer Res. 2011;17:6582–6591. doi: 10.1158/1078-0432.CCR-11-0430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Duffy AG, Makarova-Rusher OV, Ulahannan SV, Rahma OE, Fioravanti S, Walker M, Abdullah S, Raffeld M, Anderson V, Abi-Jaoudeh N, Levy E, Wood BJ, Lee S, Tomita Y, Trepel JB, Steinberg SM, Revenko AS, MacLeod AR, Peer CJ, Figg WD, Greten TF. Modulation of tumor eIF4E by antisense inhibition: a phase I/II translational clinical trial of ISIS 183750-an antisense oligonucleotide against eIF4E-in combination with irinotecan in solid tumors and irinotecan-refractory colorectal cancer. Int J Cancer. 2016;139:1648–1657. doi: 10.1002/ijc.30199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kentsis A, Volpon L, Topisirovic I, Soll CE, Culjkovic B, Shao L, Borden KL. Further evidence that ribavirin interacts with eIF4E. RNA. 2005;11:1762–1766. doi: 10.1261/rna.2238705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Casaos J, Gorelick NL, Huq S, Choi J, Xia Y, Serra R, Felder R, Lott T, Kast RE, Suk I, Brem H, Tyler B, Skuli N. The use of ribavirin as an anti-cancer therapeutic: will it go viral? Mol Cancer Ther. 2019;18:1185–1194. doi: 10.1158/1535-7163.MCT-18-0666. [DOI] [PubMed] [Google Scholar]
- 122.Assouline S, Culjkovic-Kraljacic B, Bergeron J, Caplan S, Cocolakis E, Lambert C, Lau CJ, Zahreddine HA, Miller WH Jr, Borden KL. A phase I trial of ribavirin and low-dose cytarabine for the treatment of relapsed and refractory acute myeloid leukemia with elevated eIF4E. Haematologica. 2015;100:e7–9. doi: 10.3324/haematol.2014.111245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Assouline S, Culjkovic B, Cocolakis E, Rousseau C, Beslu N, Amri A, Caplan S, Leber B, Roy DC, Miller WH Jr, Borden KL. Molecular targeting of the oncogene eIF4E in acute myeloid leukemia (AML): a proof-of-principle clinical trial with ribavirin. Blood. 2009;114:257–260. doi: 10.1182/blood-2009-02-205153. [DOI] [PubMed] [Google Scholar]
- 124.Volpon L, Osborne MJ, Zahreddine H, Romeo AA, Borden KL. Conformational changes induced in the eukaryotic translation initiation factor eIF4E by a clinically relevant inhibitor, ribavirin triphosphate. Biochem Biophys Res Commun. 2013;434:614–619. doi: 10.1016/j.bbrc.2013.03.125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Culjkovic B, Topisirovic I, Borden KL. Controlling gene expression through RNA regulons: the role of the eukaryotic translation initiation factor eIF4E. Cell Cycle. 2007;6:65–69. doi: 10.4161/cc.6.1.3688. [DOI] [PubMed] [Google Scholar]
- 126.Barbieri I, Kouzarides T. Role of RNA modifications in cancer. Nat Rev Cancer. 2020;20:303–322. doi: 10.1038/s41568-020-0253-2. [DOI] [PubMed] [Google Scholar]
- 127.Tomikawa C. 7-methylguanosine modifications in transfer RNA (tRNA) Int J Mol Sci. 2018;19:4080. doi: 10.3390/ijms19124080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Zhou Y, Dai X, Lyu J, Li Y, Bao X, Deng F, Liu K, Cui L, Cheng L. Construction and validation of a novel prognostic model for thyroid cancer based on N7-methylguanosine modification-related lncRNAs. Medicine (Baltimore) 2022;101:e31075. doi: 10.1097/MD.0000000000031075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Wang T, Zhou Z, Wang X, You L, Li W, Zheng C, Zhang J, Wang L, Kong X, Gao Y, Sun X. Comprehensive analysis of nine m7G-related lncRNAs as prognosis factors in tumor immune microenvironment of hepatocellular carcinoma and experimental validation. Front Genet. 2022;13:929035. doi: 10.3389/fgene.2022.929035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Sun J, Li L, Chen H, Gan L, Guo X, Sun J. Identification and validation of an m7G-related lncRNAs signature for prognostic prediction and immune function analysis in endometrial cancer. Genes (Basel) 2022;13:1301. doi: 10.3390/genes13081301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Li X, Dong H, Chen L, Wang Y, Hao Z, Zhang Y, Jiao Y, Zhao Z, Peng X, Zhan X. Identification of N7-methylguanosine related subtypes and construction of prognostic model in gastric cancer. Front Immunol. 2022;13:984149. doi: 10.3389/fimmu.2022.984149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Dong K, Gu D, Shi J, Bao Y, Fu Z, Fang Y, Qu L, Zhu W, Jiang A, Wang L. Identification and verification of m(7)G modification patterns and characterization of tumor microenvironment infiltration via multi-omics analysis in clear cell renal cell carcinoma. Front Immunol. 2022;13:874792. doi: 10.3389/fimmu.2022.874792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Zegers I, Gigot D, van Vliet F, Tricot C, Aymerich S, Bujnicki JM, Kosinski J, Droogmans L. Crystal structure of Bacillus subtilis TrmB, the tRNA (m7G46) methyltransferase. Nucleic Acids Res. 2006;34:1925–1934. doi: 10.1093/nar/gkl116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Alexandrov A, Martzen MR, Phizicky EM. Two proteins that form a complex are required for 7-methylguanosine modification of yeast tRNA. RNA. 2002;8:1253–1266. doi: 10.1017/s1355838202024019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Mao X, Schwer B, Shuman S. Yeast mRNA cap methyltransferase is a 50-kilodalton protein encoded by an essential gene. Mol Cell Biol. 1995;15:4167–4174. doi: 10.1128/mcb.15.8.4167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Yang YF, Zhang MF, Tian QH, Fu J, Yang X, Zhang CZ, Yang H. SPAG5 interacts with CEP55 and exerts oncogenic activities via PI3K/AKT pathway in hepatocellular carcinoma. Mol Cancer. 2018;17:117. doi: 10.1186/s12943-018-0872-3. [DOI] [PMC free article] [PubMed] [Google Scholar]


