Open Access

Research progress on PRMTs involved in epigenetic modification and tumour signalling pathway regulation (Review)

  • Authors:
    • Kailiang Wu
    • Chen Niu
    • Hanjiao Liu
    • Li Fu
  • View Affiliations

  • Published online on: April 5, 2023     https://doi.org/10.3892/ijo.2023.5510
  • Article Number: 62
  • Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Posttranslational modification (PTM) of proteins is essential for increasing protein diversity and maintaining cellular homeostasis, but uncontrolled modification may lead to tumorigenesis. Arginine methylation is a tumorigenesis‑related PTM that affects protein function through protein‑protein and protein‑nucleic acid interactions. Protein arginine methyltransferases (PRMTs) have vital roles in signalling pathways of tumour‑intrinsic and tumour‑extrinsic microenvironments. The present review summarizes the modifications and functions of PRMTs in histone methylation and nonhistone methylation, their roles in RNA splicing and DNA damage repair and the currently known functions in tumour metabolism and immunotherapy. In conclusion, this article reviews the latest research progress on the role of PRMTs in tumour signal transduction, providing a theoretical basis for clinical diagnosis and treatment. Targeting PRMTs is expected to provide new directions for tumour therapy.

1. Introduction

Posttranslational modification (PTM) is critical for protein diversity (1). Modification of proteins at one or more sites may determine protein conformation, subcellular localization, type of interacting protein, and protein stability and activity. The PTM process is catalysed by a variety of enzymes, including enzymes involved in phosphorylation, acetylation, ubiquitination, methylation and hydroxylation; conversely, the process may be reversed by enzymes that antagonize these processes, such as phosphatases, deubiquitinases, deacetylases and demethylases (1).

Protein arginine methylation is a common PTM catalysed by protein arginine methyltransferases (PRMTs) (2). In 1967, Paik and Kim (3) first discovered methylated arginine in a nuclear protein of calf thymocytes. The first member of the PRMT family, PRMT1, was identified in 1996, followed by other members (4). Protein arginine methylation has a role in the maintenance of key cellular processes, such as tissue homeostasis and disease phenotype (2).

2. Classification, structure and function of PRMTs

Based on the number and position of methyl groups on the ω-guanidino nitrogen atom of the protein arginine, arginine methylation modification may be divided into ω-NG-monomethylarginine (MMA), ω-NG,NG-asymmetric dimethylarginine (ADMA) and ω-NG,N'G-symmetric dimethylarginine (SDMA). The process by which PRMTs methylate arginine to produce MMA, ADMA and SDMA is provided in Fig. 1. Among them, type I PRMTs include PRMT1-4, -6 and -8, which methylate MMA and ADMA; type II PRMTs include PRMT5 and -9, which methylate MMA and SDMA; and type III PRMTs include PRMT7, which methylates MMA (5). The protein structure, modulation function and chromosomal location of each member of the PRMTs are provided in Fig. 2.

Arginine is a basic amino acid with a positively charged guanidine group containing five potential hydrogen bond donors, which may interact with negatively charged molecules (6). PRMTs transfer the methyl group from S-adenosine methionine (SAM) to the guanidino group of arginine in protein substrates, resulting in S-adenosyl homocysteine and methylated proteins. After methylation of arginine residues, the distance between side chains increases and the molecular configuration changes. At the same time, the addition of methyl groups reduces the number of potential hydrogen bond donors, resulting in enhanced hydrophobicity of methylated arginine, which in turn affects intramolecular and intermolecular interactions, such as protein-to-protein, protein-to-nucleic acid, as well as protein structure and stability, ultimately affecting the biological function of the modified protein (7).

PRMT1 is the most widely studied PRMT enzyme due to its powerful methyl transfer function, accounting for >85% of all modifications of PRMTs (8). The residues M48, E100, E144, E153, M155 and H293 on the active site of PRMT1 are critical for substrate and cofactor interactions (9). Mutations in these active sites weaken or result in the loss of the catalytic activity of PRMT1, leading to ADMA synthesis disorders (10). The interaction between PRMT2 and RB downregulates the activity of E2F through multiple mechanisms, including histone methylation, transcription factor (TF) methylation and RNA splicing (11). PRMT3 is localised in the cytoplasm and its protein structure includes a catalytic core and a zinc finger domain. Of note, zinc finger domains not only help to recognise RNA substrates but also regulate catalytic activity by recruiting interacting proteins (12). PRMT4, also known as coactivator-associated arginine methyltransferase 1 (CARM1), is mainly located in the nucleus and normally promotes transcription. CARM1 consists of a unique N-terminal EVH1 domain (residues 28-140) that binds to a proline-rich sequence that is essential for substrate recognition and catalytic activity (13). PRMT5 is a major type II PRMT consisting of four domains: An N-terminal TIM-barrel domain, an intermediate Rossmann fold, a C-terminal β-barrel subunit and a dimerization arm (14). The conserved F379 residue of PRMT5 methylates the production of SDMA. The F379M mutation not only increased the methylation activity of PRMT5 but also altered product specificity by generating SDMA and ADMA of H4R3. By contrast, both F379G and F379A mutations significantly reduced PRMT5 activity (14). PRMT6, a signature tag for epigenetic transcriptional repression, specifically methylates ADMA of histone H3 (15). PRMT7 is the only type III PRMT, and compared with other types of PRMTs, the protein structure of PRMT7 includes a narrower substrate-binding site, which may lead to catalytic production of MMA (16). PRMT8 is significantly and specifically highly expressed in brain and neuronal tissues. The N-terminal region of PRMT8 is able to bind to the plasma membrane, and there is a substrate binding site in the middle, which is crucial for PRMT8 methylation activity (17). PRMT9 is a recently identified type II PRMT that includes MTase domains and forms a pseudodimer for substrate binding (18).

3. Arginine methylation of histones and nonhistones

Histone methylation and transcriptional regulation

Arginine methylation of histone tails is an epigenetic modification catalysed by PRMTs and regulates gene expression. PRMTs may methylate H2AR3, H2AR29, H3R2, H3R8, H3R17/26/42, H4R3 and H4R17/19, as presented in Fig. 3. Structural analysis indicated that electrostatic interactions have a mechanistic role in the process of substrate methylation catalysed by PRMTs. It has been indicated that PRMT1, -3, -5 and -8 preferentially methylate histone H4, while PRMT4/CARM1 preferentially methylates histone H3 (19). The detailed functions of the PRMT-methylated histones H2A, H3 and H4 are presented in Table I.

Table I

Role of PRMTs in histone methylation.

Table I

Role of PRMTs in histone methylation.

TypeResiduesEnzymesFunctions(Refs.)
H2AR3me1/me2sPRMT5Transcriptional repression(22)
R3PRMT7DNA repair(23)
R29me2aPRMT6Transcriptional repression(21)
H3R2me1PRMT5Transcriptional activation(28)
R2me2aPRMT6Transcriptional repression(112,113)
R2me1/me2sPRMT5Transcriptional activation(29,114)
R8me2aPRMT6Transcriptional repression(115)
R8me2aPRMT2Transcriptional activation(24,25)
R8me2sPRMT5Transcriptional activation and repression(33,34,116,117)
R17/R26/ R42me2aCARM1Transcriptional activation(26)
R26me2aCARM1Transcriptional repression(118)
R3me1PRMT5Transcriptional activation(42)
H4R3me2aPRMT1Transcriptional activation(35,41,119)
R3me2aPRMT3Transcriptional activation(44,120)
R3PRMT6Transcriptional activation/ repression(121,122)
R3me2sPRMT5Transcriptional repression, blocking of SHH pathway, promotion of DNA methylation(36-39,42,43,123)
R17me1PRMT7Facilitation of PRMT5-mediated H4R3me2s(124)
R19me1PRMT7Facilitation of PRMT5-mediated H4R3me2s(124)
R3me2aPRMT8Transcriptional activation(125)

[i] PRMT, protein arginine methyltransferase; CARM1, coactivator-associated arginine methyltransferase 1; SHH, Sonic hedgehog.

PRMT5-7 are able to methylate H2A. Since the first five residues of H2A and H4 are identical, it is likely that most of the methylation of H4R3 also applies to H2AR3 (20). A systematic analysis of the H2A methylation status revealed that H2AR29me2 was specifically enriched in genes inhibited by PRMT6, suggesting that H2AR29me2 is involved in transcriptional repression (21). The haemagglutinin-PRMT5 complex was also able to monomethylate and symmetrically dimethylate bovine histone H2A (22). Chromatin immunoprecipitation revealed that PRMT7 dimethylates H2AR3 and H4R3 and is enriched at target DNA repair genes in parental cells (23). Studies have indicated that PRMTs are able to methylate H2A and mainly take part in transcriptional repression, but the underlying regulatory mechanism remains to be elucidated.

H3 methylation exerts transcriptional activation or transcriptional inhibition by PRMT2, CARM1 and PRMT5-7. For instance, PRMT2 is responsible for generating H3R8me2a. H3R8me2a enrichment at the BCL2 promoter may increase its accessibility to STAT3, promoting Bcl2 gene expression (24). PRMT2 acts as a transcriptional coactivator for oncogenic gene expression programs in glioblastoma multiforme (GBM) pathogenesis. PRMT2-mediated H3R8me2a enrichment at promoters and enhancers is closely associated with known active histone marks and is required for the maintenance of target gene expression (25). CARM1 may methylate H3R17, H3R26 and H3R42 and is recognised by tudor domain containing 3 to function as a coactivator (26). It is necessary that PRMT5 accumulation activates H3R2me1/me2s and recruitment of the WD repeat domain 5 (WDR5)/mixed linage leukemia (MLL) complex to promote H3K4me3, which in turn activates transcription (27-30). Recruitment of PRMT5 to the forkhead box (FOX)P1 promoter may increase H3R2me2s and H3K4me3 (29). The potential interaction of PRMT5-mediated H3R2me1 with MLL complexes (absent, small, or homologous 2 and WDR5) may activate the expression of metastasis-related genes, such as vimentin, snail family transcriptional repressor 1, snail family transcriptional repressor 2 and cadherin 2 (28). Genotoxic stress induces interactions among β-catenin, ATM phosphorylated Jun isomerization protein 2 and PRMT5, promoting redox-related gene transcription. During this process, PRMT5-mediated recruitment of H3R2me1/H3R2me2s to the WDR5/MLL complex leads to transcriptional activation of H3K4me3 and redox-related genes (30). H3R2me2a acts as a repressive mark that antagonises H3K4me3, but H3R2 is also symmetrically dimethylated (H3R2me2s) by PRMT5 and PRMT7 and is present in euchromatic regions (31). Profiling of H3-tail interactors indicated that H3R2me2s excludes binding of RBBP7, a central component of the co-repressor complexes SIN3 transcription regulator family member A, nucleosome remodeling and deacetylase and polycomb repressive complex 2. Conversely, H3R2me2s may enhance binding of WDR5, which is a common component of the coactivator complexes MLL, nuclear localization signals 1, Ada two-A containing, SET-domain-containing 1A and SET-domain-containing 1B (31). PRMT6-mediated H3R2me2a inhibits transcription by preventing H3K4me3 readers from binding (32). Of note, H3R8me2s and H4R3me2s are mainly considered repressive markers, but they have also been implicated in the transcriptional activation of certain genes, such as fibroblast growth factor receptor (FGFR)3 and eukaryotic initiation factor 4E expression in colorectal cancer (33) and androgen receptor expression in prostate cancer (34). Overall, for the regulation of H3 methylation, the symmetric arginine dimethylation of H3 generally has a role of transcriptional activation, while the asymmetric arginine dimethylation frequently has a role in transcriptional inhibition.

The methylation of H4 also has a key role in regulating the activation and repression of transcription. For instance, SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4, the ATPase subunit of the switch/sucrose nonfermentable (SWI/SNF) chromatin remodelling complex, acts as a binder for PRMT1 to methylate H4R3me2a and upregulate epidermal growth factor receptor (EGFR) in colorectal cancer (35). PRMT5-mediated H4R3me2s have been found on promoters of tumour suppressor and cyclin-dependent kinase (CDK) suppressor genes, which silence cancer-cell proliferation (36,37). Menin (MEN1) has an essential role in both repressing and activating gene expression. In MEN1-excised cells, the levels of both PRMT5 binding and H4R3m2s were decreased at the GLI1 promoter (38). MEN1 is a crucial factor for binding of the Sonic Hedgehog ligand to its receptor Patched 1 and subsequent activation of the Hedgehog signalling pathway. Of note, MEN1 mutants have reduced binding to PRMT5 and fail to impart the repressive H4R3m2s mark at the growth arrest specific 1 promoter, resulting in its elevated expression (38,39). Pharmacologic inhibition of Hedgehog signalling significantly reduces the proliferation of insulinoma cells and promotes the expression of Hedgehog signalling targets (39). Zinc finger E-box binding homeobox 1 (ZEB1), a zinc finger TF, is a key factor for epithelial to mesenchymal transition (EMT) (40). PRMT1 impacted the EMT process by mediating the asymmetric dimethylation of H4R3me2as at the ZEB1 promoter to activate its transcription, indicating the essential roles of this epigenetic control in EMT (41). Otherwise, exogenous TGFβ promotes EMT through PRMT5-MEP50 catalysing arginine monomethylation and dimethylation (42). PHD finger protein 1 (PHF1) recognizes H4R3me2s and recruits CUL4Bring E3 ligase through PHF to form a complex, silencing the expression of E-cad and FBXW7 to promote cell growth and migration (43). In addition, in 293 cells, overexpression of PRMT3 may increase the level of H4R3me2a, but the specific mechanism remains to be clarified (44). In conclusion, these findings reveal a novel link between PRMTs and H4, whereby PRMTs epigenetically regulate tumour signalling pathways, revealing them as targets for treating tumours.

Dynamic crosstalk between different histone modification types may affect gene expression. PRMT7 methylates H4R17me1 and allosterically enhances H4R3me2 of PRMT5, which in turn inhibits subsequent H3K4me3, H3Ac and H4Ac (45). Deacetylation of H3K9 by H3R8me2s is a transcriptional repression marker (46), whereas H4R3me2s is associated with H4K5 acetylation and may serve as a transcriptional activation marker (47). Of note, an increasing number of studies have demonstrated the existence of nonlinear crosstalk between different histone modification types, leading to the diversity of protein functions.

PRMTs may also affect protein tail modification by targeting microRNA (miRNA/miR) and regulating tumour gene expression. PRMT5 repressed the transcription of the miR-99 family by symmetrical dimethylation of H4R3, which increased FGFR3 expression and in turn activated Erk1/2 and Akt, leading to cell growth and metastasis in lung cancer (48). PRMT5 knockdown results in miR33b, miR96 and miR503 derepression through loss of repressive complex recruitment targeting miRNA promoters. PRMT5 is overexpressed in B-cell lymphoma and promotes the binding of miR33b, miR96 and miR503 to the 3'-untranslated region of cyclin D1 and c-MYC mRNAs, indirectly leading to enhanced cyclin D1 and c-MYC expression, which reinforces the relevance of PRMT5 in promoting lymphoma cell growth and survival (49).

Methylation of nonhistones

According to the function of non-histones after methylation, PRMTs may be divided into five types, including TFs, RNA-binding proteins (RBPs), DNA damage repair proteins, RNA splicing proteins and functional proteins in cell signalling pathways. The detailed functions of PRMT-methylated nonhistones are provided in Table II.

Table II

Role of PRMTs in nonhistone methylation.

Table II

Role of PRMTs in nonhistone methylation.

TypeEnzymesSubstrateFunction(Refs.)
TFsPRMT1TWIST1Activation of EMT(126,127)
PRMT1TAF15Affects TAF15 cellular localization and expression of TAF15-targeted genes(128)
PRMT1RUNX1Enhanced transcription activity to maintain the peripheral T-cell count(129)
PRMT1FOXO1Enhanced cell apoptosis and gluconeogenesis(130)
PRMT1/5E2F-1Transcriptional activation promotes apoptosis; increased cell viability(50,78)
PRMT1C/EBPαPromotes cyclin D1 expression(131)
PRMT1SMAD7Facilitates the dissociation of Smad7 from type I receptors(55)
PRMT5SREBP1Increases adipogenesis and tumour growth in hepatocellular carcinoma(51)
PRMT5BCL6Inhibits the activity of BCL6 target genes(52)
RBPsPRMT1PABPN1Reduces the affinity of PABPN1 for transportin, promotes the PABPN1-RNA interaction(54,132)
PRMT1ICP27Nuclear foci-like structure formation, RNA-binding affinity and SRPK interactions of ICP27(53)
PRMT5hnRNPA1Promotes hnRNPA1 interaction with ITAD, accompanied by translation of cyclin D1 and c-MYC(93)
DNA damage responsePRMT153BP1Interaction with single- and double-stranded DNA(133)
PRMT1MRE11Induction of the intra-S-phase checkpoint defect(134)
PRMT1BRCA1Influences the interaction of BRCA1 with specific promoters or proteins(135)
PRMT1/3/6TOP3BTranslational regulation(136)
PRMT6RCC1Chromatin binds and activates RAN(67)
RNA splicingPRMT5Sm proteinsAssembles into snRNPs(137)
PRMT5SRSF1Differential binding of SRSF1 to its alternative pre-mRNA splicing targets(75)
PRMT5TIP60/KATEnsuring error-free HR DNA repair and maintaining genomic integrity(57)
PRMT1RBM15Aberrant RNA splicing(138)
PRMT9SF3B2Maintains splicing fidelity(79)
PRMT1EZH2Regulates stability and promotes breast cancer metastasis(139)
Signalling pathwaysPRMT1ERαActivates the cytoplasmic signalling(140)
PRMT1EGFRActivates the EGFR signalling pathway(141)
PRMT5P65Activates NF-κB(82)
PRMT5TRIM21Activates NF-κB(83)
PRMT5SKIAltered TGFβ signalling-mediated transcriptional regulation(86)
PRMT3HIF1αActivation of the HIF1/VEGF signalling pathway(88)

[i] TF, transcription factor; RBP, RNA binding protein; PRMT, protein arginine methyltransferase; EMT, epithelial to mesenchymal transition. TWIST1, twist family bHLH transcription factor 1; TAF15, TATA-box binding protein associated factor 15; RUNX1, RUNX family transcription factor 1; FOXO1, forkhead box O1; E2F-1, E2F transcription factor 1; C/EBPα, CCAAT enhancer binding protein α; SMAD7, SMAD family member 7; SREBP1, Sterol regulatory element binding protein 1; BCL6, BCL6 transcription repressor; PABPN1, poly(A)-binding protein 1; ICP27, infected cell protein 27; hnRNPA1, heterogeneous nuclear ribonucleoprotein A1; 53BP1, p53 binding protein 1; MRE11, meiotic recombination 11; BRCA1, breast cancer 1; TOP3B, DNA topoisomerase III beta; RCC1, regulator of chromosome condensation 1; RAN, ras-related nuclear protein; SRSF1, serine and arginine rich splicing factor 1; TIP60, Tat interactive protein 60kDa; KAT5, lysine acetyltransferase 5; RNM15, RNA binding motif protein 15; SF3B2, splicing factor 3b subunit 2; EZH2, enhancer of zeste 2 polycomb repressive complex 2 subunit; ERα, estrogen receptor alpha; EGFR, epidermal growth factor receptor; TRIM21, tripartite motif containing 21; TGFβ, transforming growth factor beta; HIF1α, hypoxia inducible factor 1 subunit alpha; VEGF, vascular endothelial growth factor.

TFs may combine with RNA polymerase to form a transcription initiation complex, which jointly participates in the process of transcription initiation. E2F-1 may regulate transcription in a methylation-dependent manner (50). PRMT5 and -1 methylate E2F-1 to generate functionally opposite effects. The DNA damage response (DDR) induces E2F-1 methylation by PRMT1, increases E2F-1 expression levels and activates apoptotic gene transcription. Conversely, PRMT5 methylation of E2F-1 is recognised by Tudor domain protein, p100/tudor-SN (TSN), which reduces the E2F-1 half-life and increases cell viability. During cell cycle progression, the binding of E2F-1 to cyclin A masks the PRMT1 methylation of E2F-1, thereby inhibiting apoptosis (50). Sterol regulatory element binding protein 1 (SREBP1) is a TF regulated for de novo fatty acid synthesis. PRMT5 methylation of SREBP1 prevents its phosphorylation by glycogen synthase kinase (GSK)3β, but it is subsequently ubiquitinated by F-box and WD repeat domain containing 7 (FBXW7), increasing adipogenesis and promoting tumour growth in hepatocellular carcinoma (HCC) (51). BCL6 is a transcriptional repressor and master regulator of normal germinal centre (GC) formation and GC-derived B-cell lymphomas. PRMT5 methylation of BCL6 at R305 downregulates the activity of BCL6 target genes, inhibiting the growth of diffuse large B-cell lymphoma (52).

ICP27 is an RBP that has a crucial role in the gene expression and replication of herpes simplex virus type 1 (HSV-1) (53). Methylation of ICP27 at residues R138, R148 and R150 by PRMT1 is responsible for the formation of ICP27 nuclear foci, RNA-binding affinity and SRPK interaction. Hypomethylation of ICP27 may significantly inhibit HSV-1 replication, suggesting that PRMT inhibitors have an important role in HSV-2 therapy (53). RNA and transporters compete for binding to nuclear poly(A)-binding protein 1 (PABPN1), but methylation of PABPN1 at R289 reduces the binding capacity between PABPN1 and transporters by ~10-fold, resulting in promotion of the PABPN1-RNA interaction (54). Although eIF4A1 shares >80% sequence similarity with eIF4A2 and -3, only residue R368 of eIF4A1 protein is selectively methylated by PRMT1, whereas other eIF4A isotypes are not (55).

Phosphorylation and ubiquitination are key components of the DDR and arginine methylation is no exception. Deficiency of PRMT1 and PRMT5 leads to spontaneous DNA damage, checkpoint defects and genomic instability in mouse embryonic fibroblasts (56,57). PRMT1 is involved in the methylation of DNA repair proteins, including meiotic recombination 11 (MRE11), p53 binding protein 1, heterogeneous nuclear ribonucleoprotein U like 1, breast cancer 1 (BRCA1) and Separase, which ensure the maintenance of genome stability through homologous recombination (HR) repair and nonhomologous end-joining (NHEJ) (2,58). MRE11 is an essential component of the MRE11-RAD50-NBS1 (MRN) complex that activates the DNA repair pathway. Although methylation of MRE11 does not regulate MRN complex formation, it anchors MRE11 to double-strand breaks (DSBs), preventing nucleases from activating DNA repair. In addition, the TF growth factor independent 1 interacts with PRMT1 and promotes MRE11 methylation (59). Furthermore, CARM1 promotes mitotic arrest deficient 2 like 2 (MAD2L2) silencing by driving the switch from the SWI/SNF complex to EZH2 by methylating the BAF155 subunit of the SWI/SNF complex on the MAD2L2 promoter. EZH2 inhibition upregulates MAD2L2 to decrease DNA end resection, which increases NHEJ and chromosomal abnormalities, ultimately causing mitotic catastrophe in PARP inhibitor-treated HR-proficient cells (60). PRMT5-deficient HeLa cells are sensitive to radiotherapy and accumulate DNA damage (61). Stress-responsive activator of p300 may be recruited to the p53 complex in the DDR, which recruits PRMT5 and promotes methylation of the p53 oligomerization domain to reduce oligomerization and increase nuclear retention (62), while increasing target expression of p21 and p53-up-regulated modulator of apoptosis (63).

Upregulation of key DNA repair genes is the main mechanism of chemotherapy and radiation therapy resistance in tumour cells (64). PRMT5 may promote the survival of tumour cells in the context of genetic damage; therefore, the combination of PRMT5 inhibitors and chemotherapy may be a new strategy to treat cancer resistance. In patients with BRCA1-mutated breast cancer, the HR repair pathway is missing, so the cells rely mainly on PARP-mediated DSB repair. Therefore, Olaparib treatment is effective (65). However, the majority of patients with triple-negative breast cancer (TNBC) do not have BRCA1 mutations and targeting PRMT5 inhibition is similar to HR deficiency in BRCA1 mutations; therefore, targeting PRMT5 inhibition is beneficial for improving patient outcomes (66). Regulator of chromosome condensation 1 (RCC1), as a guanylate exchange factor of RAN, localises in the nucleus and binds to chromatin to regulate DNA damage repair. The methylation of RCC1 at R214 by PRMT6 is necessary for RCC1 to bind to chromatin and activate RAN (67). Inhibition of PRMT6 reduces the tumorigenicity of the cells in GBM and improves the effects of radiation therapy on GBM growth in mice (67).

Interactions between different posttranslational modifications are critical for the DNA damage response. Arginine methylation has an essential role in maintaining genome stability, and arginine methylation and ubiquitination crosstalk control DNA end resection and HR repair (68). Mass spectrometry analysis of PRMT1-interacting proteins revealed that ubiquitin specific peptidase 11 (USP11) has a key role in the early stage of DSB repair by regulating the activity of the PRMT1-MRE11 pathway. USP11 is a substrate of PRMT1 and methylation of USP11 promotes DNA end resection and DSB repair of DNA through HR. PRMT1 is also a ubiquitinated protein that acts as a target of de-ubiquitination to regulate the binding and methylation of PRMT1 to MRE11 (68).

RNA splicing is critical for regulating tumour phenotypes (69,70). Therefore, splicing factors must be tightly controlled genetically and epigenetically to ensure splicing fidelity. It is also important to note that protein arginine methylation usually occurs on the splicing component and that RBPs are required for pre-RNA splicing (71,72). In addition, proteome-wide analysis revealed that the enrichment of arginine-methylated proteins was associated with the control of RNA splicing, trafficking and degradation (73,74). Serine and arginine rich splicing factor 1 (SRSF1) is the substrate and effector of PRMT5 (73,75). Knockdown of PRMT5 resulted in differential binding of SRSF1 to alternative pre-mRNA splicing targets, leading to a decrease in the interaction of SRSF1 with other proteins (73). PRMT5 regulates methylation of the RGG/RG motif in RNA-binding motif protein X and forms a higher-order complex with SRSF1, and splicing generates the short isoform of MDM4 (75). PRMT5 knockdown or inhibition induces aberrant MDM4 splicing, which initiates p53-mediated cell cycle- and apoptosis-related genes, finally promoting tumour growth (76). Furthermore, activation of PRMT5 is critical for sensitivity to CDK4/6 inhibitors, and regulation of the PRMT5-MDM4 axis by palbociclib leads to loss of drug resistance in melanoma (77). By indirectly suppressing PRMT5 activity, palbociclib alters the pre-mRNA splicing of MDM4, a negative regulator of p53, leading to decreased MDM4 protein expression and subsequent p53 activation. In turn, p53 induces p21, leading to inhibition of CDK2, the main kinase substituting for CDK4/6 and a key driver of resistance to palbociclib. Loss of the ability of palbociclib to regulate the PRMT5-MDM4 axis leads to resistance. Importantly, combining palbociclib with the PRMT5 inhibitor GSK3326595 enhances the efficacy of palbociclib in treating naive and resistant models and delays the emergence of resistance. Tat interactive protein 60kDa (TIP60)/lysine acetyltransferase 5 (KAT5) is a histone lysine acetyltransferase that drives the HR of DNA and is regulated by PRMT5-mediated alternative splicing. In response to DNA damage, PRMT5 promotes alternative splicing of the pre-mRNA of TIP60/KAT to the TIP60a isoform (with high H4 lysine acetylase activity), thereby ensuring HR repair of DNA and maintaining genome integrity (57). In addition to PRMT5 methylation altering transcriptional activity, E2F1 is also involved in alternative splicing after PRMT5 methylation. Methylation of E2F1 promotes the recruitment of p100/TSN and small nuclear ribonucleoprotein, which regulates alternative splicing of E2F1 targets (78). PRMT9 is present in the splicing factor (SF)3B2 and -4 complex and methylates SF3B2. Methylated SF3B2 is recognised by the SMN Tudor domain and has an important role in maintaining splicing fidelity (79).

Methylation of arginine-specific proteins may modify the structure or activity of the protein, alter the interaction between specific molecules and then affect tumour cell signalling pathways. Recent cancer-related studies on the role of PRMTs are summarized in Table III. In TNBC, PRMT1 regulates the EGFR and the Wnt signalling pathways (80). Type I PRMT inhibitors decrease breast cancer cell proliferation and have anti-tumour activity. These inhibitors display synergistic interactions with certain chemotherapies used to treat TNBC, as well as erlotinib, an EGFR inhibitor. Therefore, targeting PRMT1 in combination with these chemotherapies may improve existing treatments for TNBC (80). Inhibition of PRMT5, the predominant type II PRMT, produces synergistic cancer-cell growth inhibition when combined with GSK3368715, which is a potent and reversible type I PRMT inhibitor (81). Of note, deletion of the methylthioadenosine phosphorylase gene (MTAP) results in accumulation of the metabolite 2-methylthioadenosine, an endogenous inhibitor of PRMT5, and is associated with sensitivity to GSK3368715. Overall, the MTAP status may serve as a biomarker for patient selection (81). NF-κB has an important role in tumorigenesis and PRMT5 activates NF-κB through methylation of the p65 subunit (82). Although TNFα-induced intracellular signalling pathways have been well studied, the TRAIL signalling pathway remains to be fully elucidated. PRMT5, a novel TRAIL receptor-binding protein, contributes to TRAIL-induced activation of inhibitor of κB kinase (IKK) and NF-κB, leading to induction of several NF-κB target genes (83). PRMT5 methylation of TRIM21 induces selective autophagy, which inhibits TRIM21-dependent monoubiquitination and degradation of IKKβ and activates the NF-κB signalling pathway. Thus, PRMT5 inhibition blocks the NF-κB signalling pathway (84). SKI is a transcriptional repressor that interacts with SMAD and may be methylated by the PRMT5-methylosome protein 50 (MEP50)-SHANK-associated RH domain interactor (SHARPIN) complex, altering transcriptional regulation of the TGFβ signalling pathway (52). In HCC, PRMT9 activates the PI3K/Akt/GSK3β/Snail signalling pathway to regulate Snail, increasing cell migration and invasion through EMT (85). SHARPIN, an adaptor for the linear ubiquitin chain assembly complex, has an important role in the NF-κB signalling pathway. Activated PRMT5 controls the expression of SRY-box transcription factor 10 and melanocyte inducing transcription factor and inhibition of the transcriptional corepressor SKI by SHARPIN-dependent arginine demethylation, contributing to the occurrence of melanomagenesis (86). The expression of PRMT3 is upregulated in colorectal cancer and may stabilise the protein structure of c-MYC, and PRMT3 promotes the expression of c-MYC by interacting with c-MYC through the SAM-dependent MTase-PRMT domain (87). PRMT3 methylates hypoxia-inducible factor (HIF)1α at R282 and stabilizes the structure of HIF1α, while activating the HIF1/VEGFA signalling pathway to promote tumorigenesis (88). PRMT1, -5 and -7 regulate glioma-associated oncogene 1 (GLI1) and GLI2 activity (89). Methylation of GLI1 by PRMT1 upregulates its activity and promotes target gene expression. PRMT5 methylates GLI1 in the cytoplasm and increases GLI1 protein stability. Conversely, nuclear PRMT5 interacts with MENIN to inhibit the expression of growth arrest-specific protein 1, which facilitates Hedgehog ligand binding to Patched and indirectly downregulates GLI1 activity. PRMT7 methylates GLI2 to upregulate its activity through GLI2 dissociation and fusion inhibitors (89). PRMT1 expression is upregulated and promotes tumour cell growth in pancreatic ductal adenocarcinoma (PDAC). PRMT1 promotes β-catenin expression by binding -699 to -874 bp and -1,191 to -1,413 bp of the β-catenin promoter (90). Tumour suppressor protein von Hippel-Lindau interacts with PRMT3 and then forms a protein complex with Auxin response factor and regulates the methylation of p53 (91). PRMT3 protein expression is upregulated in patients with gemcitabine (GEM)-resistant pancreatic cancer. ATP binding cassette subfamily G member 2 (ABCG2) is a newly discovered target of PRMT3, and PRMT3 overexpression increases the methylation of heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) at R31, resulting in enhanced RNA-binding activity of hnRNPA1 and increased expression of ABCG2 mRNA. Therefore, PRMT3 methylates the RNA recognition motif of hnRNPA1 to promote the binding of hnRNPA1 and ABCG2 to enhance the resistance of pancreatic cancer to GEM (92). PRMT5 methylation of hnRNPA1 promotes the interaction of hnRNPA1 with internal ribosome entry site (IRES) RNA to promote IRES-dependent translation of cyclin D1 and c-MYC (93). The PRMT type 1 inhibitor MS023 is a potent inducer of colon cancer-cell differentiation with a wide therapeutic window. This finding may lead to the development of clinically effective anti-cancer drugs based on the mechanism of cancer cell differentiation (94).

Table III

Roles of different PRMTs in cancer.

Table III

Roles of different PRMTs in cancer.

PRMTsCancer typeExpressionBiological mechanismFunction(Refs.)
PRMT1Colorectal cancerHighActivation of EGFR signalling by EGFR methylationOncogenic(35)
HighPromotion of tumour growth by regulation of E2F-1Oncogenic(50)
Breast cancerHighModulation of EMT and cellular senescence through regulation of ZEB1Oncogenic(41)
HighMethylation of EZH2 and regulation of its stabilityOncogenic(139)
HighActivation of cyclin D1 expression by methylation of C/EBPαOncogenic(131)
HighRegulation of IGF-1 signalling by methylation of ERαOncogenic(142)
Pancreatic cancerHighPromotion of β-catenin expression by binding the β-catenin promoterOncogenic(90)
HighStabilisation of BCL2 mRNA by methylation of HSP70Oncogenic(143)
LungHighPromotion of EMT by methylation of Twist1Oncogenic(126)
HNCHighPromotion of proliferation and migrationOncogenic(144)
ESCCHighPromotion of the growth and migration by activating Hedgehog signallingOncogenic(145)
PRMT2Hepatocellular carcinomaHighAcceleration of tumorigenesis by activating Bcl2 via H3R8 methylationOncogenic(24)
GlioblastomaHighPromotion of oncogenic activation and tumorigenesis by methylation of H3R8Oncogenic(25)
Breast cancerHighSplice variants of PRMT2 modulate ERα signallingOncogenic(146)
PRMT3Colorectal cancerHighPromotion of tumorigenesis through regulating c-MYC stabilizationOncogenic(87)
HighPromotion of tumorigenesis by methylating and stabilizing HIF1αOncogenic(88)
Pancreatic cancerHighIncrease of ABCG2 expression by methylation of hnRNPA1Oncogenic(92)
HighInduction of metabolic reprogrammingOncogenic(107)
PRMT4Ovarian cancerHighActivation of Wnt/β-catenin and neoplastic transformationOncogenic(147)
Breast cancerHighUpregulation of cyclin E1 led to the promotion of S-phase entryOncogenic(148)
HighEnhancement of tumour progression and metastasis by methylation of BAF155Oncogenic(149)
HighPromotion of invasion and metastasis by regulation of LSD1 stabilityOncogenic(150)
-Block of tumour cell proliferation and induction of differentiation through ERα-regulated genesTumour suppressive(151)
Pancreatic cancerLowInhibition of glutamine metabolism and tumour growth by methylation of MDH1Tumour suppressive(152)
Liver cancerLowRegulation of glucose metabolism by GAPDH methylationTumour suppressive(152)
PRMT5Lung cancerHigh SHARPIN-PRMT5-H3R2me1 axis activates transcription of metastasis-related genesOncogenic(28)
HighEnhancement of localization to the surface membrane by Enolase-1 methylationOncogenic(153)
HighPromotion of tumour cell proliferation by regulation of AKTOncogenic(154)
Breast cancerHighCritical regulator of breast cancer stem cells via histone methylation and Foxp1 expressionOncogenic(29)
HighPRMT5/WDR77 complex regulates alternative splicing through ZNF326Oncogenic(155)
HighIncrease of sensitivity to chemotherapeutics by governing stemnessOncogenic(156)
Prostate cancerHighPromotion of pICln-dependent androgen receptor transcriptionOncogenic(157)
Gastric cancerHighPRMT5-dependent transcriptional repression of c-MYC target genesOncogenic(158)
HighMediation of epigenetic silencing of IRX1 contributes to tumorigenicity and metastasisOncogenic(159)
AMLHighGenetic deletion or small-molecule inhibitionOncogenic(36)
HighGene activation and repression via histone arginine methylationOncogenic(117)
HCCHighPromotion of invasive activity of tumour cells by regulation of MMP2Oncogenic(160)
HighPromotion of tumour cell proliferation by inhibiting BTG2 expression via the ERK signalling pathwayOncogenic(161)
MelanomaHighRegulation of MDM4 via alternative splicing result in response to CDK4/6 inhibitorsOncogenic(77)
LymphomaHighActivation of WNT/β-catenin and AKT/GSK3β signallingOncogenic(162)
DLBCLHighPRMT5 is upregulated by B-cell receptor signalling and forms a positive feedback loop with PI3K/AKTOncogenic(163)
Bladder cancerHighPromotion of cancer growth through inhibiting NF-κB-dependent apoptosisOncogenic(164)
PRMT6Lung cancerHighActivation of tumour-associated macrophages via interaction with ILF2Oncogenic(165)
Endometrial cancerHighActivation of AKT/mTOR signallingOncogenic(166)
PRMT7Breast cancerHighPromotion of tumour cell invasion through the induction of MMP9 expressionOncogenic(167)
NSCLCHighContribution to tumour cell metastasis by the interaction with HSPA5 and EEF2Oncogenic(168)
PRMT9HCCHighPromotion of tumour cell invasion and metastasis by activating PI3K/Akt/ GSK-3β/Snail signallingOncogenic(85)

[i] AML, acute myeloid leukemia; HNC, head and neck cancer; DLBCL, diffuse large B-cell lymphoma; ESCC, esophageal squamous cell carcinoma; NSCLC, non-small cell lung carcinoma; HCC, hepatocarcinoma; PRMT, protein arginine methyltransferase; EGFR, epidermal growth factor receptor; E2F-1, E2F transcription factor 1; EMT, epithelial to mesenchymal transition; ZEB1, Zinc finger E-box binding homeobox 1; EZH2, enhancer of zeste 2 polycomb repressive complex 2 subunit; C/EBPα, CCAAT enhancer binding protein alpha; IGF-1, IGF like family member 1; ERα, estrogen receptor alpha; BCL2, BCL2 apoptosis regulator; HSP70, heat shock protein 70; ABCG2, ATP binding cassette subfamily G member 2; hnRNPA1, heterogeneous nuclear ribonucleoprotein A1; BAF155, SWI/SNF related, matrix associated, actin dependent regulator of chromatin subfamily c member 1; LSD1, lysine demethylase 1; MDH1, malate dehydrogenase 1; SHARPIN, SHANK-associated RH domain interactor; WDR77, WD repeat domain 77; ZNF326, zinc finger protein 326; IRX1, iroquois homeobox 1; MMP2, matrix metallopeptidase 2; BTG2, BTG anti-proliferation factor 2; ILF2, interleukin enhancer binding factor 2.

4. Participation in tumour immunity

PRMTs can modulate Toll-like receptor and interferon (IFN) activation at multiple levels to modulate immune responses (95). PRMT5 expression was observed to be negatively associated with antitumor immunity. After PRMT5 inhibition, the number of infiltrating immune cells increased and antitumour immunity was enhanced in immunocompetent mice (96). PRMT5 promotes antitumour immunity through two different intertumoral pathways. First, PRMT5-mediated interferon gamma inducible protein 16 (IFI16)/IFI204 methylation attenuates dsDNA-induced TANK binding kinase 1 (TBK1)-interferon regulatory factor 3 (IRF3) activation and chemokine production. dsDNA induced activation of TBK1-IRF3, as reflected by the levels of STING phosphorylation, dimerization and polymerization. PRMT5 methylation of IFI16/IFI204 impacts cyclic GMP-AMP synthase-stimulator of interferon genes (STING) signalling. Ectopically expressed IFI204 in B16 cells activated TBK1-IRF3 signalling and increased the expression of IFNB1 and C-C motif chemokine ligand 5 following dsDNA treatment. IFI204Mt1 (R12A) expression increased STING dimerization and polymerization following dsDNA stimuli, suggesting a critical role of IFI204 methylation on Arg12 in the dsDNA-stimulated STING pathway activation. PRMT5-mediated IFI16/IFI204 methylation attenuates dsDNA-induced TBK1-IRF3 activation and type I interferon and chemokine production. In addition, NLR family CARD domain containing 5, a master regulator of inflammasomes and antigen presentation pathways, was inversely correlated with PRMT5 expression. PRMT5 inhibits immune cell recruitment and activation as well as tumour recognition, thereby influencing tumour immune evasion. Likewise, inhibition of PRMT5 is expected to enhance the response of cold tumours to immune checkpoint therapy (96). MS023, a type I PRMT inhibitor, causes splicing of modulatory drugs, treatment alterations with intron retention and exon skipping, and these alterations result in substantial enrichment of major histocompatibility complex I-binding peptides. Of note, a fairly large proportion (up to 43%) of these putative neoantigens are immunogenic, resulting in neoantigen-specific CD8+ T-cell activation (97).

Controlling PRMT5 activity is a promising strategy for cancer therapy when host immunity against tumours occurs in a FOXP3-dependent manner (98). Arginine methylation occurs frequently at R27, R51 and R146 of FOXP3, but pharmacological inhibition of PRMT5 by DS-437 may reduce T-regulatory cell (Treg) functions and inhibits the methylation of FOXP3. Furthermore, DS-437 significantly enhanced the anti-tumour effects of anti-erbB2/neu monoclonal antibody targeted therapy in BALB/c mice, which bore CT26Her2 tumours, by inhibiting Treg function and induction of tumour immunity (98). Of note, FOXP3 also undergoes methylation on R48 and R51 by interacting with PRMT1. The inhibition of arginine methylation confers gene expression profiles representing type I helper T cells to FOXP3+ T cells, which resulted in attenuated suppressive activity (99). Otherwise, knockout of PRMT1 may enhance anti-programmed cell death receptor-1 immunotherapy in MC38-derived tumours in isogenic C57BL/6 mice (100). Of note, the PRMT1 polymorphism rs975484 modulates programmed cell death ligand-1 (PD-L1) and PD-L2 levels and serves as a predictor of immune checkpoint blockade efficiency in HCC (101). CARM1 was identified as a negative regulator of tumour-specific T cells in B16F10 melanoma-resistant C57BL/6 mice (102).

Furthermore, studies have indicated that type I PRMT inhibitors may enhance the effect of immunotherapy. PT1001B enhances antitumor immunity, and combining it with anti-PD-L1 checkpoint inhibitors provides a potential strategy to overcome anti-PD-L1 resistance in PDAC (103). MS023 treatment significantly improved anti-PD1 therapy in C57BL/6 tumour-bearing mice (104). Therefore, further studies are needed to determine the effect of PRMT inhibition, not only on tumour cells, but also on other cell types in the tumour microenvironment, including immune cells and stromal cells.

5. Participation in metabolic reprogramming

Metabolic reprogramming is an important process by which cancer cells adapt to high energy demands and supplement biosynthetic needs, and numerous cancers switch their tumour cell metabolism to the glycolytic pathway under oxygen-rich conditions. Therefore, inhibition of metabolic reprogramming by modulating different metabolic pathways in tumours provides a new strategy for cancer therapy (105,106).

In pancreatic cancer, PRMT3 may methylate GAPDH at R248 to enhance cancer glycolysis and mitochondrial respiration. PRMT3-overexpressing cancer cells were addicted to GAPDH-mediated metabolism and sensitive to the inhibition of GAPDH and mitochondrial respiration. Both intermediates in the glycolytic pathway and the tricarboxylic acid cycle are enriched in PRMT3-expressed cells. In addition, these cells exhibit an increased extracellular acidification rate and oxygen consumption rate. Double blockade of GAPDH and mitochondrial respiration will be a novel strategy for the treatment of PRMT3-overexpressing pancreatic cancer (107). Esophageal squamous cell carcinoma (ESCC) is associated with elevated asymmetric and systemic arginine dimethylarginine. PRMT1, PRMT5, ornithine decarboxylase 1 and nitric oxide (NO) synthase 2 are overexpressed, and arginase 1, arginase 2 and dimethylarginine dimethylaminohydrolase 1 are downregulated in tumours compared to adjacent tissues. Arginine bioavailability increased and citrulline decreased along with ESCC advancement. In short, metabolic reprogramming in ESCC manifests as alterations in the L-arginine/NO pathway (108). In HCC, PRMT3 mediates ADMA modification of lactate dehydrogenase A (LDHA) at R112. LDHA-R112K-mutant-expressing cells exhibited a decrease in LDH activity, HCC cell glycolysis and proliferation (109). In chronic myeloid leukemia (CML), loss of PRMT7 resulted in reduced expression of glycine decarboxylase, leading to the reprograming of glycine metabolism to generate methylglyoxal, which is detrimental to leukemia stem cells (LSCs). These findings link histone arginine methylation with glycine metabolism, while suggesting PRMT7 as a potential therapeutic target for the eradication of LSCs in CML (109).

6. Methylation profiles of PRMTs

Despite the relevance of PRMTs for signal transduction, metabolism, transcription and other cellular phenotypes, the methylation profiles of protein arginine remain understudied. The function of PRMTs under physiological and pathological conditions often depends on methyltransferase activity. Therefore, comprehensively revealing the substrates of PRMTs is the key to exploring their functions and underlying molecular mechanisms.

Combining two newly developed methylation sequencing methods, immunoaffinity purification and high pH strong cation exchange, may improve the coverage of protein methylation and reveal new PRMT1 targets (110). After knockout of PRMT1, 127 arginine methylation sites on 78 proteins were significantly changed. In contrast, only one lysine methylation site was significantly changed after PRMT1 knockdown, indicating that amino acid methylation was not affected by PRMT1 knockdown. In PRMT1-knockout cells, 114 MMA sites were found to be significantly altered on proteins enriched in the mRNA metabolic process. A high-confidence list of 18 PRMT1 substrates and 12 methylation sites scavenged by other PRMTs in the absence of PRMT1 activity was found through integrative analysis of MMA and DMA. Most importantly, the methylation site hnRNPA1 R206 switched from ADMA to SDMA after PRMT1 knockout (110).

High-resolution mass spectrometry combined with SILAC technology was used to analyse the arginine methylation regulated by PRMT7 in 293 cells (111). A total of 1,031 MMA sites of 513 proteins were detected and a two-fold decrease in monomethylation levels at 297 arginine sites in 174 proteins was found, termed the PRMT7 methylome. During this process, the methylation of 176 MMA sites in 108 proteins disappeared completely. After treatment with the PRMT7-specific inhibitor SGC3027, 503 MMA sites of 274 proteins were more than two-fold reduced, and ~60% of PRMT7-regulated substrates were also inhibited. The same method was also used to identify the methylated substrates of PRMT4 and -5, representing type I and II PRMTs, respectively. The PRMT4 methyl group (two-fold decrease) includes 301 proteins with 660 methylation sites, while the PRMT5 methyl group (two-fold decrease) includes 244 proteins with 429 methylation sites. PRMT4 substrates, such as mediator complex subunit 12, SWI/SNF related, matrix associated, actin dependent regulator of chromatin subfamily c member 1 and E1A binding protein p300, and PRMT5 substrates, such as FUS RNA binding protein, hnRNPA1 and survival of motor neuron 1, have also been identified. Most importantly, PRMT4, -5 and -7 coregulate alternative splicing in an enzyme-dependent manner and share a number of RNA splicing factors, such as hnRNPA1; furthermore, hnRNPA1 arginine methylation is required for the growth of various cancer cells (116). Taken together, the methylation profiles of PRMTs indicated that hnRNAPA1 has a role of co-RNA splicing factor in various arginine methylation modification processes and may also promote the transformation of ADMA to SDMA. Further study of its mechanism will reveal the types of arginine methylation.

7. Future outlook

While much important progress has been made in research on PRMT function, the complexity and significance of gene post-transcriptional processing determine the diversity of the PRMT regulatory mechanisms. The regulatory effect of PRMTs on the function of RBP clearly exemplifies the important regulatory role of arginine methylation in post-transcriptional processing. Although numerous important advances have been made in the study of PRMTs regulating the function of RBP, various questions remain to be answered in the field of cancer, such as how a single arginine methylation site determines the function of specific proteins, how different PRMT family members synergistically regulate the occurrence of specific substrate methylation and how arginine methylation modifications are recognized and removed. In addition, the sequence characteristic of the target RNA directly affected by RBP and how arginine methylation regulates the interaction between RBP and its target RNA at the genome-wide level are also key questions to be solved. In addition, PRMTs directly methylate numerous proteins to control their subcellular localization, protein-protein interactions, stability or activity. Many of these contribute to oncogenic transformation, and thus, evaluation of potential PRMT inhibitors is warranted. What remains to be established is how the inhibition of arginine methylation may be integrated with immunotherapeutic approaches to achieve a maximal, long-lasting therapeutic effect.

Availability of data and materials

Not applicable.

Authors' contributions

KW, CN and HL were involved in the conception of the review. KW wrote the manuscript and performed the literature search. KW and LF reviewed and edited the final manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

This study was supported by grants from the National Natural Science Foundation of China (grant nos. 81672637, 81872164 and 82173344).

References

1 

Vu LD, Gevaert K and De Smet I: Protein language: Post-translational modifications talking to each other. Trends Plant Sci. 23:1068–1080. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Jarrold J and Davies CC: PRMTs and arginine methylation: Cancer's best-kept secret? Trends Mol Med. 25:993–1009. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Paik WK and Kim S: Enzymatic methylation of protein fractions from calf thymus nuclei. Biochem Biophys Res Commun. 29:14–20. 1967. View Article : Google Scholar : PubMed/NCBI

4 

Lin WJ, Gary JD, Yang MC, Clarke S and Herschman HR: The mammalian immediate-early TIS21 protein and the leukemia-associated BTG1 protein interact with a protein-arginine N-methyltransferase. J Biol Chem. 271:15034–15044. 1996. View Article : Google Scholar : PubMed/NCBI

5 

Blanc RS and Richard S: Arginine methylation: The coming of age. Mol Cell. 65:8–24. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Bedford MT and Clarke SG: Protein arginine methylation in mammals: Who, what, and why. Mol Cell. 33:1–13. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Hughes RM and Waters ML: Arginine methylation in a beta-hairpin peptide: Implications for Arg-pi interactions, DeltaCp(o), and the cold denatured state. J Am Chem Soc. 128:12735–12742. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Obianyo O, Causey CP, Jones JE and Thompson PR: Activity-based protein profiling of protein arginine methyltransferase 1. ACS Chem Biol. 6:1127–1135. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Gui S, Wooderchak WL, Daly MP, Porter PJ, Johnson SJ and Hevel JM: Investigation of the molecular origins of protein-arginine methyltransferase I (PRMT1) product specificity reveals a role for two conserved methionine residues. J Biol Chem. 286:29118–29126. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Gui S, Gathiaka S, Li J, Qu J, Acevedo O and Hevel JM: A remodeled protein arginine methyltransferase 1 (PRMT1) generates symmetric dimethylarginine. J Biol Chem. 289:9320–9327. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Yoshimoto T, Boehm M, Olive M, Crook MF, San H, Langenickel T and Nabel EG: The arginine methyltransferase PRMT2 binds RB and regulates E2F function. Exp Cell Res. 312:2040–2053. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Frankel A and Clarke S: PRMT3 is a distinct member of the protein arginine N-methyltransferase family. Conferral of substrate specificity by a zinc-finger domain. J Biol Chem. 275:32974–32982. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Shishkova E, Zeng H, Liu F, Kwiecien NW, Hebert AS, Coon JJ and Xu W: Global mapping of CARM1 substrates defines enzyme specificity and substrate recognition. Nat Commun. 8:155712017. View Article : Google Scholar : PubMed/NCBI

14 

Sun L, Wang M, Lv Z, Yang N, Liu Y, Bao S, Gong W and Xu RM: Structural insights into protein arginine symmetric dimethylation by PRMT5. Proc Natl Acad Sci USA. 108:20538–20543. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Okuno K, Akiyama Y, Shimada S, Nakagawa M, Tanioka T, Inokuchi M, Yamaoka S, Kojima K and Tanaka S: Asymmetric dimethylation at histone H3 arginine 2 by PRMT6 in gastric cancer progression. Carcinogenesis. 40:15–26. 2019. View Article : Google Scholar

16 

Hasegawa M, Toma-Fukai S, Kim JD, Fukamizu A and Shimizu T: Protein arginine methyltransferase 7 has a novel homodimer-like structure formed by tandem repeats. FEBS Lett. 588:1942–1948. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Lee J, Sayegh J, Daniel J, Clarke S and Bedford MT: PRMT8, a new membrane-bound tissue-specific member of the protein arginine methyltransferase family. J Biol Chem. 280:32890–32896. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Hadjikyriacou A, Yang Y, Espejo A, Bedford MT and Clarke SG: Unique features of human protein arginine methyltransferase 9 (PRMT9) and its substrate RNA splicing factor SF3B2. J Biol Chem. 290:16723–16743. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Fulton MD, Cao M, Ho MC, Zhao X and Zheng YG: The macromolecular complexes of histones affect protein arginine methyltransferase activities. J Biol Chem. 297:1011232021. View Article : Google Scholar : PubMed/NCBI

20 

Osborne TC, Obianyo O, Zhang X, Cheng X and Thompson PR: Protein arginine methyltransferase 1: Positively charged residues in substrate peptides distal to the site of methylation are important for substrate binding and catalysis. Biochemistry. 46:13370–13381. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Waldmann T, Izzo A, Kamieniarz K, Richter F, Vogler C, Sarg B, Lindner H, Young NL, Mittler G, Garcia BA and Schneider R: Methylation of H2AR29 is a novel repressive PRMT6 target. Epigenetics Chromatin. 4:112011. View Article : Google Scholar : PubMed/NCBI

22 

Branscombe TL, Frankel A, Lee JH, Cook JR, Yang Z, Pestka S and Clarke S: PRMT5 (Janus kinase-binding protein 1) catalyzes the formation of symmetric dimethylarginine residues in proteins. J Biol Chem. 276:32971–32976. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Karkhanis V, Wang L, Tae S, Hu YJ, Imbalzano AN and Sif S: Protein arginine methyltransferase 7 regulates cellular response to DNA damage by methylating promoter histones H2A and H4 of the polymerase δ catalytic subunit gene, POLD1. J Biol Chem. 287:29801–29814. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Hu G, Yan C, Xie P, Cao Y, Shao J and Ge J: PRMT2 accelerates tumorigenesis of hepatocellular carcinoma by activating Bcl2 via histone H3R8 methylation. Exp Cell Res. 394:1121522020. View Article : Google Scholar : PubMed/NCBI

25 

Dong F, Li Q, Yang C, Huo D, Wang X, Ai C, Kong Y, Sun X, Wang W, Zhou Y, et al: PRMT2 links histone H3R8 asymmetric dimethylation to oncogenic activation and tumorigenesis of glioblastoma. Nat Commun. 9:45522018. View Article : Google Scholar : PubMed/NCBI

26 

Yang Y, Lu Y, Espejo A, Wu J, Xu W, Liang S and Bedford MT: TDRD3 is an effector molecule for arginine-methylated histone marks. Mol Cell. 40:1016–1023. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Xu J and Richard S: Cellular pathways influenced by protein arginine methylation: Implications for cancer. Mol Cell. 81:4357–4368. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Fu T, Lv X, Kong Q and Yuan C: A novel SHARPIN-PRMT5-H3R2me1 axis is essential for lung cancer cell invasion. Oncotarget. 8:54809–54820. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Chiang K, Zielinska AE, Shaaban AM, Sanchez-Bailon MP, Jarrold J, Clarke TL, Zhang J, Francis A, Jones LJ, Smith S, et al: PRMT5 is a critical regulator of breast cancer stem cell function via histone methylation and FOXP1 expression. Cell Rep. 21:3498–3513. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Cao L, Wu G, Zhu J, Tan Z, Shi D, Wu X, Tang M, Li Z, Hu Y, Zhang S, et al: Genotoxic stress-triggered β-catenin/JDP2/PRMT5 complex facilitates reestablishing glutathione homeostasis. Nat Commun. 10:37612019. View Article : Google Scholar

31 

Migliori V, Müller J, Phalke S, Low D, Bezzi M, Mok WC, Sahu SK, Gunaratne J, Capasso P, Bassi C, et al: Symmetric dimethylation of H3R2 is a newly identified histone mark that supports euchromatin maintenance. Nat Struct Mol Biol. 19:136–144. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Mitchell LH, Drew AE, Ribich SA, Rioux N, Swinger KK, Jacques SL, Lingaraj T, Boriack-Sjodin PA, Waters NJ, Wigle TJ, et al: Aryl pyrazoles as potent inhibitors of arginine methyltransferases: Identification of the first PRMT6 tool compound. ACS Med Chem Lett. 6:655–659. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Zhang B, Dong S, Zhu R, Hu C, Hou J, Li Y, Zhao Q, Shao X, Bu Q, Li H, et al: Targeting protein arginine methyltransferase 5 inhibits colorectal cancer growth by decreasing arginine methylation of eIF4E and FGFR3. Oncotarget. 6:22799–22811. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Deng X, Shao G, Zhang HT, Li C, Zhang D, Cheng L, Elzey BD, Pili R, Ratliff TL, Huang J and Hu CD: Protein arginine methyltransferase 5 functions as an epigenetic activator of the androgen receptor to promote prostate cancer cell growth. Oncogene. 36:1223–1231. 2017. View Article : Google Scholar :

35 

Yao B, Gui T, Zeng X, Deng Y, Wang Z, Wang Y, Yang D, Li Q, Xu P, Hu R, et al: PRMT1-mediated H4R3me2a recruits SMARCA4 to promote colorectal cancer progression by enhancing EGFR signaling. Genome Med. 13:582021. View Article : Google Scholar : PubMed/NCBI

36 

Kaushik S, Liu F, Veazey KJ, Gao G, Das P, Neves LF, Lin K, Zhong Y, Lu Y, Giuliani V, et al: Genetic deletion or small-molecule inhibition of the arginine methyltransferase PRMT5 exhibit anti-tumoral activity in mouse models of MLL-rearranged AML. Leukemia. 32:499–509. 2018. View Article : Google Scholar

37 

Yang L, Ma DW, Cao YP, Li DZ, Zhou X, Feng JF and Bao J: PRMT5 functionally associates with EZH2 to promote colorectal cancer progression through epigenetically repressing CDKN2B expression. Theranostics. 11:3742–3759. 2021. View Article : Google Scholar : PubMed/NCBI

38 

Gurung B, Feng Z and Hua X: Menin directly represses Gli1 expression independent of canonical Hedgehog signaling. Mol Cancer Res. 11:1215–1222. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Gurung B, Feng Z, Iwamoto DV, Thiel A, Jin G, Fan CM, Ng JM, Curran T and Hua X: Menin epigenetically represses Hedgehog signaling in MEN1 tumor syndrome. Cancer Res. 73:2650–2658. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Krebs AM, Mitschke J, Lasierra Losada M, Schmalhofer O, Boerries M, Busch H, Boettcher M, Mougiakakos D, Reichardt W, Bronsert P, et al: The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat Cell Biol. 19:518–529. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Gao Y, Zhao Y, Zhang J, Lu Y, Liu X, Geng P, Huang B, Zhang Y and Lu J: The dual function of PRMT1 in modulating epithelial-mesenchymal transition and cellular senescence in breast cancer cells through regulation of ZEB1. Sci Rep. 6:198742016. View Article : Google Scholar : PubMed/NCBI

42 

Chen H, Lorton B, Gupta V and Shechter D: A TGFβ-PRMT5-MEP50 axis regulates cancer cell invasion through histone H3 and H4 arginine methylation coupled transcriptional activation and repression. Oncogene. 36:373–386. 2017. View Article : Google Scholar

43 

Liu R, Gao J, Yang Y, Qiu R, Zheng Y, Huang W, Zeng Y, Hou Y, Wang S, Leng S, et al: PHD finger protein 1 (PHF1) is a novel reader for histone H4R3 symmetric dimethylation and coordinates with PRMT5-WDR77/CRL4B complex to promote tumorigenesis. Nucleic Acids Res. 46:6608–6626. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Siarheyeva A, Senisterra G, Allali-Hassani A, Dong A, Dobrovetsky E, Wasney GA, Chau I, Marcellus R, Hajian T, Liu F, et al: An allosteric inhibitor of protein arginine methyltransferase 3. Structure. 20:1425–1435. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Jain K and Clarke SG: PRMT7 as a unique member of the protein arginine methyltransferase family: A review. Arch Biochem Biophys. 665:36–45. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Pal S, Vishwanath SN, Erdjument-Bromage H, Tempst P and Sif S: Human SWI/SNF-associated PRMT5 methylates histone H3 arginine 8 and negatively regulates expression of ST7 and NM23 tumor suppressor genes. Mol Cell Biol. 24:9630–9645. 2004. View Article : Google Scholar : PubMed/NCBI

47 

Scaglione A, Patzig J, Liang J, Frawley R, Bok J, Mela A, Yattah C, Zhang J, Teo SX, Zhou T, et al: PRMT5-mediated regulation of developmental myelination. Nat Commun. 9:28402018. View Article : Google Scholar : PubMed/NCBI

48 

Jing P, Zhao N, Ye M, Zhang Y, Zhang Z, Sun J, Wang Z, Zhang J and Gu Z: Protein arginine methyltransferase 5 promotes lung cancer metastasis via the epigenetic regulation of miR-99 family/FGFR3 signaling. Cancer Lett. 427:38–48. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Karkhanis V, Alinari L, Ozer HG, Chung J, Zhang X, Sif S and Baiocchi RA: Protein arginine methyltransferase 5 represses tumor suppressor miRNAs that down-regulate CYCLIN D1 and c-MYC expression in aggressive B-cell lymphoma. J Biol Chem. 295:1165–1180. 2020. View Article : Google Scholar :

50 

Cho EC, Zheng S, Munro S, Liu G, Carr SM, Moehlenbrink J, Lu YC, Stimson L, Khan O, Konietzny R, et al: Arginine methylation controls growth regulation by E2F-1. EMBO J. 31:1785–1797. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Liu L, Zhao X, Zhao L, Li J, Yang H, Zhu Z, Liu J and Huang G: Arginine methylation of SREBP1a via PRMT5 promotes de novo lipogenesis and tumor growth. Cancer Res. 76:1260–1272. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Lu X, Fernando TM, Lossos C, Yusufova N, Liu F, Fontá L, Durant M, Geng H, Melnick J, Luo Y, et al: PRMT5 interacts with the BCL6 oncoprotein and is required for germinal center formation and lymphoma cell survival. Blood. 132:2026–2039. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Yu J, Shin B, Park ES, Yang S, Choi S, Kang M and Rho J: Protein arginine methyltransferase 1 regulates herpes simplex virus replication through ICP27 RGG-box methylation. Biochem Biophys Res Commun. 391:322–328. 2010. View Article : Google Scholar

54 

Fronz K, Güttinger S, Burkert K, Kühn U, Stöhr N, Schierhorn A and Wahle E: Arginine methylation of the nuclear poly(a) binding protein weakens the interaction with its nuclear import receptor, transportin. J Biol Chem. 286:32986–32994. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Katsuno Y, Qin J, Oses-Prieto J, Wang H, Jackson-Weaver O, Zhang T, Lamouille S, Wu J, Burlingame A, Xu J and Derynck R: Arginine methylation of SMAD7 by PRMT1 in TGF-β-induced epithelial-mesenchymal transition and epithelial stem-cell generation. J Biol Chem. 293:13059–13072. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Clarke TL, Sanchez-Bailon MP, Chiang K, Reynolds JJ, Herrero-Ruiz J, Bandeiras TM, Matias PM, Maslen SL, Skehel JM, Stewart GS and Davies CC: PRMT5-dependent methylation of the TIP60 coactivator RUVBL1 is a key regulator of homologous recombination. Mol Cell. 65:900–916.e7. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Hamard PJ, Santiago GE, Liu F, Karl DL, Martinez C, Man N, Mookhtiar AK, Duffort S, Greenblatt S, Verdun RE and Nimer SD: PRMT5 regulates DNA repair by controlling the alternative splicing of histone-modifying enzymes. Cell Rep. 24:2643–2657. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Hellmuth S, Gutiérrez-Caballero C, Llano E, Pendás AM and Stemmann O: Local activation of mammalian separase in interphase promotes double-strand break repair and prevents oncogenic transformation. EMBO J. 37:e991842018. View Article : Google Scholar : PubMed/NCBI

59 

Vadnais C, Chen R, Fraszczak J, Yu Z, Boulais J, Pinder J, Frank D, Khandanpour C, Hébert J, Dellaire G, et al: GFI1 facilitates efficient DNA repair by regulating PRMT1 dependent methylation of MRE11 and 53BP1. Nat Commun. 9:14182018. View Article : Google Scholar : PubMed/NCBI

60 

Karakashev S, Fukumoto T, Zhao B, Lin J, Wu S, Fatkhutdinov N, Park PH, Semenova G, Jean S, Cadungog MG, et al: EZH2 inhibition sensitizes CARM1-high, homologous recombination proficient ovarian cancers to PARP inhibition. Cancer Cell. 37:157–167.e6. 2020. View Article : Google Scholar : PubMed/NCBI

61 

Wei X, Yang J, Adair SJ, Ozturk H, Kuscu C, Lee KY, Kane WJ, O'Hara PE, Liu D, Demirlenk YM, et al: Targeted CRISPR screening identifies PRMT5 as synthetic lethality combinatorial target with gemcitabine in pancreatic cancer cells. Proc Natl Acad Sci USA. 117:28068–28079. 2020. View Article : Google Scholar : PubMed/NCBI

62 

Li Y, Chitnis N, Nakagawa H, Kita Y, Natsugoe S, Yang Y, Li Z, Wasik M, Klein-Szanto AJ, Rustgi AK and Diehl JA: PRMT5 is required for lymphomagenesis triggered by multiple oncogenic drivers. Cancer Discov. 5:288–303. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Jansson M, Durant ST, Cho EC, Sheahan S, Edelmann M, Kessler B and La Thangue NB: Arginine methylation regulates the p53 response. Nat Cell Biol. 10:1431–1439. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Rocha CRR, Silva MM, Quinet A, Cabral-Neto JB and Menck CFM: DNA repair pathways and cisplatin resistance: An intimate relationship. Clinics (Sao Paulo). 73(Suppl 1): e478s2018. View Article : Google Scholar : PubMed/NCBI

65 

McCabe N, Turner NC, Lord CJ, Kluzek K, Bialkowska A, Swift S, Giavara S, O'Connor MJ, Tutt AN, Zdzienicka MZ, et al: Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res. 66:8109–8115. 2006. View Article : Google Scholar : PubMed/NCBI

66 

Peshkin BN, Alabek ML and Isaacs C: BRCA1/2 mutations and triple negative breast cancers. Breast Dis. 32:25–33. 2010. View Article : Google Scholar

67 

Huang T, Yang Y, Song X, Wan X, Wu B, Sastry N, Horbinski CM, Zeng C, Tiek D, Goenka A, et al: PRMT6 methylation of RCC1 regulates mitosis, tumorigenicity, and radiation response of glioblastoma stem cells. Mol Cell. 81:1276–1291.e9. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Sanchez-Bailon MP, Choi SY, Dufficy ER, Sharma K, McNee GS, Gunnell E, Chiang K, Sahay D, Maslen S, Stewart GS, et al: Arginine methylation and ubiquitylation crosstalk controls DNA end-resection and homologous recombination repair. Nat Commun. 12:63132021. View Article : Google Scholar : PubMed/NCBI

69 

Dvinge H, Kim E, Abdel-Wahab O and Bradley RK: RNA splicing factors as oncoproteins and tumour suppressors. Nat Rev Cancer. 16:413–430. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Bonnal SC, López-Oreja I and Valcárcel J: Roles and mechanisms of alternative splicing in cancer-implications for care. Nat Rev Clin Oncol. 17:457–474. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Fong JY, Pignata L, Goy PA, Kawabata KC, Lee SC, Koh CM, Musiani D, Massignani E, Kotini AG, Penson A, et al: Therapeutic targeting of RNA splicing catalysis through inhibition of protein arginine methylation. Cancer Cell. 36:194–209.e9. 2019. View Article : Google Scholar : PubMed/NCBI

72 

Guccione E and Richard S: The regulation, functions and clinical relevance of arginine methylation. Nat Rev Mol Cell Biol. 20:642–657. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Radzisheuskaya A, Shliaha PV, Grinev V, Lorenzini E, Kovalchuk S, Shlyueva D, Gorshkov V, Hendrickson RC, Jensen ON and Helin K: PRMT5 methylome profiling uncovers a direct link to splicing regulation in acute myeloid leukemia. Nat Struct Mol Biol. 26:999–1012. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Musiani D, Bok J, Massignani E, Wu L, Tabaglio T, Ippolito MR, Cuomo A, Ozbek U, Zorgati H, Ghoshdastider U, et al: Proteomics profiling of arginine methylation defines PRMT5 substrate specificity. Sci Signal. 12:eaat83882019. View Article : Google Scholar : PubMed/NCBI

75 

Cai T, Cinkornpumin JK, Yu Z, Villarreal OD, Pastor WA and Richard S: Deletion of RBMX RGG/RG motif in Shashi-XLID syndrome leads to aberrant p53 activation and neuronal differentiation defects. Cell Rep. 36:1093372021. View Article : Google Scholar : PubMed/NCBI

76 

Gerhart SV, Kellner WA, Thompson C, Pappalardi MB, Zhang XP, Montes de Oca R, Penebre E, Duncan K, Boriack-Sjodin A, Le B, et al: Activation of the p53-MDM4 regulatory axis defines the anti-tumour response to PRMT5 inhibition through its role in regulating cellular splicing. Sci Rep. 8:97112018. View Article : Google Scholar : PubMed/NCBI

77 

AbuHammad S, Cullinane C, Martin C, Bacolas Z, Ward T, Chen H, Slater A, Ardley K, Kirby L, Chan KT, et al: Regulation of PRMT5-MDM4 axis is critical in the response to CDK4/6 inhibitors in melanoma. Proc Natl Acad Sci USA. 116:17990–18000. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Roworth AP, Carr SM, Liu G, Barczak W, Miller RL, Munro S, Kanapin A, Samsonova A and La Thangue NB: Arginine methylation expands the regulatory mechanisms and extends the genomic landscape under E2F control. Sci Adv. 5:eaaw46402019. View Article : Google Scholar : PubMed/NCBI

79 

Yang Y, Hadjikyriacou A, Xia Z, Gayatri S, Kim D, Zurita-Lopez C, Kelly R, Guo A, Li W, Clarke SG and Bedford MT: PRMT9 is a type II methyltransferase that methylates the splicing factor SAP145. Nat Commun. 6:64282015. View Article : Google Scholar : PubMed/NCBI

80 

Suresh S, Huard S, Brisson A, Némati F, Dakroub R, Poulard C, Ye M, Martel E, Reyes C, Silvestre DC, et al: PRMT1 regulates EGFR and Wnt signaling pathways and is a promising target for combinatorial treatment of breast cancer. Cancers (Basel). 14:3062022. View Article : Google Scholar : PubMed/NCBI

81 

Fedoriw A, Rajapurkar SR, O'Brien S, Gerhart SV, Mitchell LH, Adams ND, Rioux N, Lingaraj T, Ribich SA, Pappalardi MB, et al: Anti-tumor activity of the type I PRMT inhibitor, GSK3368715, synergizes with PRMT5 inhibition through MTAP loss. Cancer Cell. 36:100–114.e25. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Wei H, Wang B, Miyagi M, She Y, Gopalan B, Huang DB, Ghosh G, Stark GR and Lu T: PRMT5 dimethylates R30 of the p65 subunit to activate NF-κB. Proc Natl Acad Sci USA. 110:13516–13521. 2013. View Article : Google Scholar

83 

Tanaka H, Hoshikawa Y, Oh-hara T, Koike S, Naito M, Noda T, Arai H, Tsuruo T and Fujita N: PRMT5, a novel TRAIL receptor-binding protein, inhibits TRAIL-induced apoptosis via nuclear factor-kappaB activation. Mol Cancer Res. 7:557–569. 2009. View Article : Google Scholar : PubMed/NCBI

84 

Gullà A, Hideshima T, Bianchi G, Fulciniti M, Kemal Samur M, Qi J, Tai YT, Harada T, Morelli E, Amodio N, et al: Protein arginine methyltransferase 5 has prognostic relevance and is a druggable target in multiple myeloma. Leukemia. 32:996–1002. 2018. View Article : Google Scholar

85 

Jiang H, Zhou Z, Jin S, Xu K, Zhang H and Xu J, Sun Q, Wang J and Xu J: PRMT9 promotes hepatocellular carcinoma invasion and metastasis via activating PI3K/Akt/GSK-3β/Snail signaling. Cancer Sci. 109:1414–1427. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Tamiya H, Kim H, Klymenko O, Kim H, Feng Y, Zhang T, Han JY, Murao A, Snipas SJ, Jilaveanu L, et al: SHARPIN-mediated regulation of protein arginine methyltransferase 5 controls melanoma growth. J Clin Invest. 128:517–530. 2018. View Article : Google Scholar :

87 

Hu Y, Su Y, He Y, Liu W and Xiao B: Arginine methyltransferase PRMT3 promote tumorigenesis through regulating c-MYC stabilization in colorectal cancer. Gene. 791:1457182021. View Article : Google Scholar : PubMed/NCBI

88 

Zhang X, Wang K, Feng X, Wang J, Chu Y, Jia C, He Q and Chen C: PRMT3 promotes tumorigenesis by methylating and stabilizing HIF1α in colorectal cancer. Cell Death Dis. 12:10662021. View Article : Google Scholar

89 

Abe Y and Tanaka N: Fine-tuning of GLI activity through arginine methylation: its mechanisms and function. Cells. 9:19732020. View Article : Google Scholar : PubMed/NCBI

90 

Song C, Chen T, He L, Ma N, Li JA, Rong YF, Fang Y, Liu M, Xie D and Lou W: PRMT1 promotes pancreatic cancer growth and predicts poor prognosis. Cell Oncol (Dordr). 43:51–62. 2020.

91 

Lai Y, Song M, Hakala K, Weintraub ST and Shiio Y: Proteomic dissection of the von Hippel-Lindau (VHL) interactome. J Proteome Res. 10:5175–5182. 2011. View Article : Google Scholar : PubMed/NCBI

92 

Hsu MC, Pan MR, Chu PY, Tsai YL, Tsai CH, Shan YS, Chen LT and Hung WC: Protein arginine methyltransferase 3 enhances chemoresistance in pancreatic cancer by methylating hnRNPA1 to increase ABCG2 expression. Cancers (Basel). 11:82018. View Article : Google Scholar : PubMed/NCBI

93 

Gao G, Dhar S and Bedford MT: PRMT5 regulates IRES-dependent translation via methylation of hnRNP A1. Nucleic Acids Res. 45:4359–4369. 2017.PubMed/NCBI

94 

Plotnikov A, Kozer N, Cohen G, Carvalho S, Duberstein S, Almog O, Solmesky LJ, Shurrush KA, Babaev I, Benjamin S, et al: PRMT1 inhibition induces differentiation of colon cancer cells. Sci Rep. 10:200302020. View Article : Google Scholar : PubMed/NCBI

95 

Sengupta S, Kennemer A, Patrick K, Tsichlis P and Guerau-de-Arellano M: Protein arginine methyltransferase 5 in T lymphocyte biology. Trends Immunol. 41:918–931. 2020. View Article : Google Scholar : PubMed/NCBI

96 

Kim H, Kim H, Feng Y, Li Y, Tamiya H, Tocci S and Ronai ZA: PRMT5 control of cGAS/STING and NLRC5 pathways defines melanoma response to antitumor immunity. Sci Transl Med. 12:eaaz56832020. View Article : Google Scholar : PubMed/NCBI

97 

Elliott K, Nilsson J and Van den Eynden J: Pharmacologic RNA splicing modulation: A novel mechanism to enhance neoantigen-directed anti-tumor immunity and immunotherapy response. Signal Transduct Target Ther. 6:3732021. View Article : Google Scholar : PubMed/NCBI

98 

Nagai Y, Ji MQ, Zhu F, Xiao Y, Tanaka Y, Kambayashi T, Fujimoto S, Goldberg MM, Zhang H, Li B, et al: PRMT5 associates with the FOXP3 homomer and when disabled enhances targeted p185erbB2/neu tumor immunotherapy. Front Immunol. 10:1742019. View Article : Google Scholar

99 

Kagoya Y, Saijo H, Matsunaga Y, Guo T, Saso K, Anczurowski M, Wang CH, Sugata K, Murata K, Butler MO, et al: Arginine methylation of FOXP3 is crucial for the suppressive function of regulatory T cells. J Autoimmun. 97:10–21. 2019. View Article : Google Scholar

100 

Hou J, Wang Y, Shi L, Chen Y, Xu C, Saeedi A, Pan K, Bohat R, Egan NA, McKenzie JA, et al: Integrating genome-wide CRISPR immune screen with multi-omic clinical data reveals distinct classes of tumor intrinsic immune regulators. J Immunother Cancer. 9:e0018192021. View Article : Google Scholar : PubMed/NCBI

101 

Schonfeld M, Zhao J, Komatz A, Weinman SA and Tikhanovich I: The polymorphism rs975484 in the protein arginine methyltransferase 1 gene modulates expression of immune checkpoint genes in hepatocellular carcinoma. J Biol Chem. 295:7126–7137. 2020. View Article : Google Scholar : PubMed/NCBI

102 

Kumar S, Zeng Z, Bagati A, Tay RE, Sanz LA, Hartono SR, Ito Y, Abderazzaq F, Hatchi E, Jiang P, et al: CARM1 inhibition enables immunotherapy of resistant tumors by dual action on tumor cells and T cells. Cancer Discov. 11:2050–2071. 2021. View Article : Google Scholar : PubMed/NCBI

103 

Zheng NN, Zhou M, Sun F, Huai MX, Zhang Y, Qu CY, Shen F and Xu LM: Combining protein arginine methyltransferase inhibitor and anti-programmed death-ligand-1 inhibits pancreatic cancer progression. World J Gastroenterol. 26:3737–3749. 2020. View Article : Google Scholar : PubMed/NCBI

104 

Lu SX, De Neef E, Thomas JD, Sabio E, Rousseau B, Gigoux M, Knorr DA, Greenbaum B, Elhanati Y, Hogg SJ, et al: Pharmacologic modulation of RNA splicing enhances anti-tumor immunity. Cell. 184:4032–4047.e31. 2021. View Article : Google Scholar : PubMed/NCBI

105 

Tennant DA, Durán RV and Gottlieb E: Targeting metabolic transformation for cancer therapy. Nat Rev Cancer. 10:267–277. 2010. View Article : Google Scholar : PubMed/NCBI

106 

Ganapathy-Kanniappan S and Geschwind JF: Tumor glycolysis as a target for cancer therapy: Progress and prospects. Mol Cancer. 12:1522013. View Article : Google Scholar : PubMed/NCBI

107 

Hsu MC, Tsai YL, Lin CH, Pan MR, Shan YS, Cheng TY, Cheng SH, Chen LT and Hung WC: Protein arginine methyltransferase 3-induced metabolic reprogramming is a vulnerable target of pancreatic cancer. J Hematol Oncol. 12:792019. View Article : Google Scholar : PubMed/NCBI

108 

Bednarz-Misa I, Fortuna P, Fleszar MG, Lewandowski Ł, Diakowska D, Rosińczuk J and Krzystek-Korpacka M: Esophageal squamous cell carcinoma is accompanied by local and systemic changes in L-arginine/NO pathway. Int J Mol Sci. 21:62822020. View Article : Google Scholar : PubMed/NCBI

109 

Lei Y, Han P, Chen Y, Wang H, Wang S, Wang M, Liu J, Yan W, Tian D and Liu M: Protein arginine methyltransferase 3 promotes glycolysis and hepatocellular carcinoma growth by enhancing arginine methylation of lactate dehydrogenase A. Clin Transl Med. 12:e6862022. View Article : Google Scholar : PubMed/NCBI

110 

Hartel NG, Chew B, Qin J, Xu J and Graham NA: Deep protein methylation profiling by combined chemical and immunoaffinity approaches reveals novel PRMT1 targets. Mol Cell Proteomics. 18:2149–2164. 2019. View Article : Google Scholar : PubMed/NCBI

111 

Li WJ, He YH, Yang JJ, Hu GS, Lin YA, Ran T, Peng BL, Xie BL, Huang MF, Gao X, et al: Profiling PRMT methylome reveals roles of hnRNPA1 arginine methylation in RNA splicing and cell growth. Nat Commun. 12:19462021. View Article : Google Scholar : PubMed/NCBI

112 

Iberg AN, Espejo A, Cheng D, Kim D, Michaud-Levesque J, Richard S and Bedford MT: Arginine methylation of the histone H3 tail impedes effector binding. J Biol Chem. 283:3006–3010. 2008. View Article : Google Scholar

113 

Neault M, Mallette FA, Vogel G, Michaud-Levesque J and Richard S: Ablation of PRMT6 reveals a role as a negative transcriptional regulator of the p53 tumor suppressor. Nucleic Acids Res. 40:9513–9521. 2012. View Article : Google Scholar : PubMed/NCBI

114 

Lorton BM, Harijan RK, Burgos ES, Bonanno JB, Almo SC and Shechter D: A binary arginine methylation switch on histone H3 arginine 2 regulates its interaction with WDR5. Biochemistry. 59:3696–3708. 2020. View Article : Google Scholar : PubMed/NCBI

115 

Guccione E, Bassi C, Casadio F, Martinato F, Cesaroni M, Schuchlautz H, Lüscher B and Amati B: Methylation of histone H3R2 by PRMT6 and H3K4 by an MLL complex are mutually exclusive. Nature. 449:933–937. 2007. View Article : Google Scholar : PubMed/NCBI

116 

Dacwag CS, Ohkawa Y, Pal S, Sif S and Imbalzano AN: The protein arginine methyltransferase Prmt5 is required for myogenesis because it facilitates ATP-dependent chromatin remodeling. Mol Cell Biol. 27:384–394. 2007. View Article : Google Scholar :

117 

Tarighat SS, Santhanam R, Frankhouser D, Radomska HS, Lai H, Anghelina M, Wang H, Huang X, Alinari L, Walker A, et al: The dual epigenetic role of PRMT5 in acute myeloid leukemia: Gene activation and repression via histone arginine methylation. Leukemia. 30:789–799. 2016. View Article : Google Scholar

118 

Zhang Z, Nikolai BC, Gates LA, Jung SY, Siwak EB, He B, Rice AP, O'Malley BW and Feng Q: Crosstalk between histone modifications indicates that inhibition of arginine methyltransferase CARM1 activity reverses HIV latency. Nucleic Acids Res. 45:9348–9360. 2017. View Article : Google Scholar : PubMed/NCBI

119 

Cheung N, Fung TK, Zeisig BB, Holmes K, Rane JK, Mowen KA, Finn MG, Lenhard B, Chan LC and So CW: Targeting aberrant epigenetic networks mediated by PRMT1 and KDM4C in acute myeloid leukemia. Cancer Cell. 29:32–48. 2016. View Article : Google Scholar : PubMed/NCBI

120 

Min Z, Xiaomeng L, Zheng L, Yangge D, Xuejiao L, Longwei L, Xiao Z, Yunsong L, Ping Z and Yongsheng Z: Asymmetrical methyltransferase PRMT3 regulates human mesenchymal stem cell osteogenesis via miR-3648. Cell Death Dis. 10:5812019. View Article : Google Scholar : PubMed/NCBI

121 

Zhang Y, van Haren MJ and Martin NI: Peptidic transition state analogues as PRMT inhibitors. Methods. 175:24–29. 2020. View Article : Google Scholar

122 

Hamey JJ, Rakow S, Bouchard C, Senst JM, Kolb P, Bauer UM, Wilkins MR and Hart-Smith G: Systematic investigation of PRMT6 substrate recognition reveals broad specificity with a preference for an RG motif or basic and bulky residues. FEBS J. 288:5668–5691. 2021. View Article : Google Scholar : PubMed/NCBI

123 

Zhao Q, Rank G, Tan YT, Li H, Moritz RL, Simpson RJ, Cerruti L, Curtis DJ, Patel DJ, Allis CD, et al: PRMT5-mediated methylation of histone H4R3 recruits DNMT3A, coupling histone and DNA methylation in gene silencing. Nat Struct Mol Biol. 16:304–311. 2009. View Article : Google Scholar : PubMed/NCBI

124 

Jain K, Jin CY and Clarke SG: Epigenetic control via allosteric regulation of mammalian protein arginine methyltransferases. Proc Natl Acad Sci USA. 114:10101–10106. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Di Lorenzo A and Bedford MT: Histone arginine methylation. FEBS Lett. 585:2024–2031. 2011. View Article : Google Scholar

126 

Avasarala S, Van Scoyk M, Karuppusamy Rathinam MK, Zerayesus S, Zhao X, Zhang W, Pergande MR, Borgia JA, DeGregori J, Port JD, et al: PRMT1 is a novel regulator of epithelial-mesenchymal-transition in non-small cell lung cancer. J Biol Chem. 290:13479–13489. 2015. View Article : Google Scholar : PubMed/NCBI

127 

Zhao Z, Rahman MA, Chen ZG and Shin DM: Multiple biological functions of Twist1 in various cancers. Oncotarget. 8:20380–20393. 2017. View Article : Google Scholar : PubMed/NCBI

128 

Jobert L, Argentini M and Tora L: PRMT1 mediated methylation of TAF15 is required for its positive gene regulatory function. Exp Cell Res. 315:1273–1286. 2009. View Article : Google Scholar : PubMed/NCBI

129 

Mizutani S, Yoshida T, Zhao X, Nimer SD, Taniwaki M and Okuda T: Loss of RUNX1/AML1 arginine-methylation impairs peripheral T cell homeostasis. Br J Haematol. 170:859–873. 2015. View Article : Google Scholar : PubMed/NCBI

130 

Yamagata K, Daitoku H, Takahashi Y, Namiki K, Hisatake K, Kako K, Mukai H, Kasuya Y and Fukamizu A: Arginine methylation of FOXO transcription factors inhibits their phosphorylation by Akt. Mol Cell. 32:221–231. 2008. View Article : Google Scholar : PubMed/NCBI

131 

Liu LM, Sun WZ, Fan XZ, Xu YL, Cheng MB and Zhang Y: Methylation of C/EBPα by PRMT1 inhibits its tumor-suppressive function in breast cancer. Cancer Res. 79:2865–2877. 2019. View Article : Google Scholar : PubMed/NCBI

132 

Fronz K, Otto S, Kölbel K, Kühn U, Friedrich H, Schierhorn A, Beck-Sickinger AG, Ostareck-Lederer A and Wahle E: Promiscuous modification of the nuclear poly(A)-binding protein by multiple protein-arginine methyltransferases does not affect the aggregation behavior. J Biol Chem. 283:20408–20420. 2008. View Article : Google Scholar : PubMed/NCBI

133 

Boisvert FM, Rhie A, Richard S and Doherty AJ: The GAR motif of 53BP1 is arginine methylated by PRMT1 and is necessary for 53BP1 DNA binding activity. Cell Cycle. 4:1834–1841. 2005. View Article : Google Scholar : PubMed/NCBI

134 

Boisvert FM, Déry U, Masson JY and Richard S: Arginine methylation of MRE11 by PRMT1 is required for DNA damage checkpoint control. Genes Dev. 19:671–676. 2005. View Article : Google Scholar : PubMed/NCBI

135 

Guendel I, Carpio L, Pedati C, Schwartz A, Teal C, Kashanchi F and Kehn-Hall K: Methylation of the tumor suppressor protein, BRCA1, influences its transcriptional cofactor function. PLoS One. 5:e113792010. View Article : Google Scholar : PubMed/NCBI

136 

Huang L, Wang Z, Narayanan N and Yang Y: Arginine methylation of the C-terminus RGG motif promotes TOP3B topoisomerase activity and stress granule localization. Nucleic Acids Res. 46:3061–3074. 2018. View Article : Google Scholar : PubMed/NCBI

137 

Friesen WJ, Paushkin S, Wyce A, Massenet S, Pesiridis GS, Van Duyne G, Rappsilber J, Mann M and Dreyfuss G: The methylosome, a 20S complex containing JBP1 and pICln, produces dimethylarginine-modified Sm proteins. Mol Cell Biol. 21:8289–8300. 2001. View Article : Google Scholar : PubMed/NCBI

138 

Zhang L, Tran NT, Su H, Wang R, Lu Y, Tang H, Aoyagi S, Guo A, Khodadadi-Jamayran A, Zhou D, et al: Cross-talk between PRMT1-mediated methylation and ubiquitylation on RBM15 controls RNA splicing. Elife. 4:e079382015. View Article : Google Scholar : PubMed/NCBI

139 

Li Z, Wang D, Lu J, Huang B, Wang Y, Dong M, Fan D, Li H, Gao Y, Hou P, et al: Methylation of EZH2 by PRMT1 regulates its stability and promotes breast cancer metastasis. Cell Death Differ. 27:3226–3242. 2020. View Article : Google Scholar : PubMed/NCBI

140 

Le Romancer M, Treilleux I, Leconte N, Robin-Lespinasse Y, Sentis S, Bouchekioua-Bouzaghou K, Goddard S, Gobert-Gosse S and Corbo L: Regulation of estrogen rapid signaling through arginine methylation by PRMT1. Mol Cell. 31:212–221. 2008. View Article : Google Scholar : PubMed/NCBI

141 

Nakai K, Xia W, Liao HW, Saito M, Hung MC and Yamaguchi H: The role of PRMT1 in EGFR methylation and signaling in MDA-MB-468 triple-negative breast cancer cells. Breast Cancer. 25:74–80. 2018. View Article : Google Scholar

142 

Choucair A, Pham TH, Omarjee S, Jacquemetton J, Kassem L, Trédan O, Rambaud J, Marangoni E, Corbo L, Treilleux I and Le Romancer M: The arginine methyltransferase PRMT1 regulates IGF-1 signaling in breast cancer. Oncogene. 38:4015–4027. 2019. View Article : Google Scholar : PubMed/NCBI

143 

Wang L, Jia Z, Xie D, Zhao T, Tan Z, Zhang S, Kong F, Wei D and Xie K: Methylation of HSP70 orchestrates its binding to and stabilization of BCL2 mRNA and renders pancreatic cancer cells resistant to therapeutics. Cancer Res. 80:4500–4513. 2020. View Article : Google Scholar : PubMed/NCBI

144 

Chuang CY, Chang CP, Lee YJ, Lin WL, Chang WW, Wu JS, Cheng YW, Lee H and Li C: PRMT1 expression is elevated in head and neck cancer and inhibition of protein arginine methylation by adenosine dialdehyde or PRMT1 knockdown downregulates proliferation and migration of oral cancer cells. Oncol Rep. 38:1115–1123. 2017. View Article : Google Scholar : PubMed/NCBI

145 

Zhou W, Yue H, Li C, Chen H and Yuan Y: Protein arginine methyltransferase 1 promoted the growth and migration of cancer cells in esophageal squamous cell carcinoma. Tumour Biol. 37:2613–2619. 2016. View Article : Google Scholar

146 

Zhong J, Cao RX, Zu XY, Hong T, Yang J, Liu L, Xiao XH, Ding WJ, Zhao Q, Liu JH and Wen GB: Identification and characterization of novel spliced variants of PRMT2 in breast carcinoma. FEBS J. 279:316–335. 2012. View Article : Google Scholar

147 

Ou CY, LaBonte MJ, Manegold PC, So AY, Ianculescu I, Gerke DS, Yamamoto KR, Ladner RD, Kahn M, Kim JH and Stallcup MR: A coactivator role of CARM1 in the dysregulation of β-catenin activity in colorectal cancer cell growth and gene expression. Mol Cancer Res. 9:660–670. 2011. View Article : Google Scholar : PubMed/NCBI

148 

El Messaoudi S, Fabbrizio E, Rodriguez C, Chuchana P, Fauquier L, Cheng D, Theillet C, Vandel L, Bedford MT and Sardet C: Coactivator-associated arginine methyltransferase 1 (CARM1) is a positive regulator of the Cyclin E1 gene. Proc Natl Acad Sci USA. 103:13351–13356. 2006. View Article : Google Scholar : PubMed/NCBI

149 

Wang L, Zhao Z, Meyer MB, Saha S, Yu M, Guo A, Wisinski KB, Huang W, Cai W, Pike JW, et al: CARM1 methylates chromatin remodeling factor BAF155 to enhance tumor progression and metastasis. Cancer Cell. 30:179–180. 2016. View Article : Google Scholar : PubMed/NCBI

150 

Liu J, Feng J, Li L, Lin L, Ji J, Lin C, Liu L, Zhang N, Duan D, Li Z, et al: Arginine methylation-dependent LSD1 stability promotes invasion and metastasis of breast cancer. EMBO Rep. 21:e485972020. View Article : Google Scholar

151 

Al-Dhaheri M, Wu J, Skliris GP, Li J, Higashimato K, Wang Y, White KP, Lambert P, Zhu Y, Murphy L and Xu W: CARM1 is an important determinant of ERα-dependent breast cancer cell differentiation and proliferation in breast cancer cells. Cancer Res. 71:2118–2128. 2011. View Article : Google Scholar : PubMed/NCBI

152 

Wang YP, Zhou W, Wang J, Huang X, Zuo Y, Wang TS, Gao X, Xu YY, Zou SW, Liu YB, et al: Arginine methylation of MDH1 by CARM1 inhibits glutamine metabolism and suppresses pancreatic cancer. Mol Cell. 64:673–687. 2016. View Article : Google Scholar : PubMed/NCBI

153 

Zakrzewicz D, Didiasova M, Krüger M, Giaimo BD, Borggrefe T, Mieth M, Hocke AC, Zakrzewicz A, Schaefer L, Preissner KT and Wygrecka M: Protein arginine methyltransferase 5 mediates enolase-1 cell surface trafficking in human lung adenocarcinoma cells. Biochim Biophys Acta Mol Basis Dis. 1864:1816–1827. 2018. View Article : Google Scholar : PubMed/NCBI

154 

Zhang S, Ma Y, Hu X, Zheng Y and Chen X: Targeting PRMT5/Akt signalling axis prevents human lung cancer cell growth. J Cell Mol Med. 23:1333–1342. 2019. View Article : Google Scholar :

155 

Rengasamy M, Zhang F, Vashisht A, Song WM, Aguilo F, Sun Y, Li S, Zhang W, Zhang B, Wohlschlegel JA and Walsh MJ: The PRMT5/WDR77 complex regulates alternative splicing through ZNF326 in breast cancer. Nucleic Acids Res. 45:11106–11120. 2017. View Article : Google Scholar : PubMed/NCBI

156 

Wang Z, Kong J, Wu Y, Zhang J, Wang T, Li N, Fan J, Wang H, Zhang J and Ling R: PRMT5 determines the sensitivity to chemotherapeutics by governing stemness in breast cancer. Breast Cancer Res Treat. 168:531–542. 2018. View Article : Google Scholar

157 

Beketova E, Fang S, Owens JL, Liu S, Chen X, Zhang Q, Asberry AM, Deng X, Malola J, Huang J, et al: Protein arginine methyltransferase 5 promotes pICln-dependent androgen receptor transcription in castration-resistant prostate cancer. Cancer Res. 80:4904–4917. 2020. View Article : Google Scholar : PubMed/NCBI

158 

Liu M, Yao B, Gui T, Guo C, Wu X, Li J, Ma L, Deng Y, Xu P, Wang Y, et al: PRMT5-dependent transcriptional repression of c-Myc target genes promotes gastric cancer progression. Theranostics. 10:4437–4452. 2020. View Article : Google Scholar : PubMed/NCBI

159 

Liu X, Zhang J, Liu L, Jiang Y, Ji J, Yan R, Zhu Z and Yu Y: Protein arginine methyltransferase 5-mediated epigenetic silencing of IRX1 contributes to tumorigenicity and metastasis of gastric cancer. Biochim Biophys Acta Mol Basis Dis. 1864:2835–2844. 2018. View Article : Google Scholar : PubMed/NCBI

160 

Jeon JY, Lee JS, Park ER, Shen YN, Kim MY, Shin HJ, Joo HY, Cho EH, Moon SM, Shin US, et al: Protein arginine methyltransferase 5 is implicated in the aggressiveness of human hepatocellular carcinoma and controls the invasive activity of cancer cells. Oncol Rep. 40:536–544. 2018.PubMed/NCBI

161 

Jiang H, Zhu Y, Zhou Z and Xu J, Jin S, Xu K, Zhang H, Sun Q, Wang J and Xu J: PRMT5 promotes cell proliferation by inhibiting BTG2 expression via the ERK signaling pathway in hepatocellular carcinoma. Cancer Med. 7:869–882. 2018. View Article : Google Scholar : PubMed/NCBI

162 

Chung J, Karkhanis V, Baiocchi RA and Sif S: Protein arginine methyltransferase 5 (PRMT5) promotes survival of lymphoma cells via activation of WNT/β-catenin and AKT/GSK3β proliferative signaling. J Biol Chem. 294:7692–7710. 2019. View Article : Google Scholar : PubMed/NCBI

163 

Zhu F, Guo H, Bates PD, Zhang S, Zhang H, Nomie KJ, Li Y, Lu L, Seibold KR, Wang F, et al: PRMT5 is upregulated by B-cell receptor signaling and forms a positive-feedback loop with PI3K/AKT in lymphoma cells. Leukemia. 33:2898–2911. 2019. View Article : Google Scholar : PubMed/NCBI

164 

Hu G, Wang X, Han Y and Wang P: Protein arginine methyltransferase 5 promotes bladder cancer growth through inhibiting NF-kB dependent apoptosis. EXCLI J. 17:1157–1166. 2018.

165 

Avasarala S, Wu PY, Khan SQ, Yanlin S, Van Scoyk M, Bao J, Di Lorenzo A, David O, Bedford MT, Gupta V, et al: PRMT6 promotes lung tumor progression via the alternate activation of tumor-associated macrophages. Mol Cancer Res. 18:166–178. 2020. View Article : Google Scholar :

166 

Jiang N, Li QL, Pan W, Li J, Zhang MF, Cao T, Su SG and Shen H: PRMT6 promotes endometrial cancer via AKT/mTOR signaling and indicates poor prognosis. Int J Biochem Cell Biol. 120:1056812020. View Article : Google Scholar

167 

Baldwin RM, Haghandish N, Daneshmand M, Amin S, Paris G, Falls TJ, Bell JC, Islam S and Côté J: Protein arginine methyltransferase 7 promotes breast cancer cell invasion through the induction of MMP9 expression. Oncotarget. 6:3013–3032. 2015. View Article : Google Scholar : PubMed/NCBI

168 

Cheng D, He Z, Zheng L, Xie D, Dong S and Zhang P: PRMT7 contributes to the metastasis phenotype in human non-small-cell lung cancer cells possibly through the interaction with HSPA5 and EEF2. Onco Targets Ther. 11:4869–4876. 2018. View Article : Google Scholar : PubMed/NCBI

169 

Yang Y and Bedford MT: Protein arginine methyltransferases and cancer. Nat Rev Cancer. 13:37–50. 2013. View Article : Google Scholar

Related Articles

Journal Cover

May-2023
Volume 62 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu K, Niu C, Liu H and Fu L: Research progress on PRMTs involved in epigenetic modification and tumour signalling pathway regulation (Review). Int J Oncol 62: 62, 2023
APA
Wu, K., Niu, C., Liu, H., & Fu, L. (2023). Research progress on PRMTs involved in epigenetic modification and tumour signalling pathway regulation (Review). International Journal of Oncology, 62, 62. https://doi.org/10.3892/ijo.2023.5510
MLA
Wu, K., Niu, C., Liu, H., Fu, L."Research progress on PRMTs involved in epigenetic modification and tumour signalling pathway regulation (Review)". International Journal of Oncology 62.5 (2023): 62.
Chicago
Wu, K., Niu, C., Liu, H., Fu, L."Research progress on PRMTs involved in epigenetic modification and tumour signalling pathway regulation (Review)". International Journal of Oncology 62, no. 5 (2023): 62. https://doi.org/10.3892/ijo.2023.5510