Open Access

m1A regulator‑mediated methylation modifications and gene signatures and their prognostic value in multiple myeloma

  • Authors:
    • Jiaqi Fu
    • Xingjun Han
    • Wei Gao
    • Manya Yu
    • Xing Cui
  • View Affiliations

  • Published online on: November 18, 2024     https://doi.org/10.3892/etm.2024.12768
  • Article Number: 18
  • Copyright: © Fu 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

N1‑methyladenosine (m1A), a methylation of RNA, is gaining attention for its role in diverse biological processes. However, the potential roles of m1A regulatory‑mediated methylation modifications in multiple myeloma (MM) remain unclear. The mRNA expression of m1A regulators in normal plasma (NP; n=9) and MM (n=174) bone marrow plasma cells was investigated and the m1A modification patterns of 559 MM samples based on the expression of 10 m1A‑related regulatory genes were comprehensively evaluated. Univariate Cox regression, Kaplan‑Meier survival curve, unsupervised clustering and gene enrichment analyses were used to explore the associations between m1A‑related regulatory genes and MM patient survival/prognosis. The m1A score model was subsequently constructed to quantify the m1A modification patterns of individual tumours, and its predictive performance was further assessed via receiver operating characteristic curves. Immunohistochemistry, Cell Counting Kit‑8, flow cytometry and m1A dot blot assays were performed to investigate the potential role of YTH domain‑containing family protein 2 (YTHDF2) in MM. Moreover, bioinformatics analysis was performed to predict the potential downstream regulatory mechanism of YTHDF2. In total, seven differentially expressed genes [tRNA methyltransferase 61A/B (TRMT61A/B), YTH N6‑methyladenosine RNA binding protein (YTHD)F1/F2/F3/C1 and Alkb homolog 1, histone H2A dioxygenase (ALKBH1)] were identified in MM samples compared with those in NP samples. The forest map and Kaplan‑Meier curve revealed that the expression of m1A‑related regulatory genes could be favourable prognostic factors for patients with MM. A total of three distinct m1A modification patterns were determined, and Cluster B exhibited the worst outcome, which was accompanied by increased expression of YTHDF2, tRNA methyltransferase 6 non‑catalytic subunit (TRMT6), tRNA methyltransferase 10 homolog C (TRMT10C) and TRMT61B. The m1Ascore model was subsequently constructed. A high m1A score was associated with clinical benefit and an improved treatment response. High expression of the reader protein YTHDF2 was associated with poor survival in patients with MM and was superior to other assessed proteins in terms of prognosis. Subsequent cell experiments demonstrated that YTHDF2 promoted the proliferation and inhibited the apoptosis of U266 cells. Notably, an evident increase in the m1A level was observed when YTHDF2 was overexpressed. A total of 150 genes related to YTHDF2 were identified, and these genes were enriched in the peroxisome proliferator‑activated receptor signalling pathway, protein export, positive regulation of protein targeting to the membrane, and gamma‑delta intraepithelial T‑cell differentiation. Serine and arginine rich splicing factor 10 (SRSF10) was subsequently selected because SRSF10 expression was significantly positively correlated with YTHDF2 expression and was also associated with poor prognosis in patients with MM. The present study revealed the modification patterns and high prognostic value of m1A regulators and demonstrated that the reader protein YTHDF2 is a potentially crucial target for MM.

Introduction

Multiple myeloma (MM) is one of the most common malignant tumours in the blood system and is characterized by the clonal proliferation of malignant plasma cells in the bone marrow and symptoms related to anaemia, immunosuppression, bone destruction and renal failure (1). In recent decades, treatment strategies have greatly advanced, but MM remains an incurable malignant disease. Considerable evidence has indicated that epigenetic mechanisms are generally involved in the pathogenesis of MM, and these mechanisms can regulate gene expression at the level of DNA and chromatin structural modifications, RNA stability, and transcriptional activity; related modification types include RNA modifications, DNA methylations, histone covalent modifications, and non-coding RNA regulation (2-4). Furthermore, epitranscriptomics is a newly emerging field that focuses mainly on the effects of chemical modifications carried by RNAs and their associated regulators on gene expression (5).

An increasing number of studies have revealed the critical roles of N1-methyladenosine (m1A) modification and its regulators in cancer development, and strategies involving m1A modification and m1A-related regulators have received attention (6,7). m1A, which appears on the first nitrogen atom of adenosine in RNA, is highly enriched in the 5'UTR and tends to be located in highly structured regions (8). Similar to DNA and protein modifications, m1A modification is a type of dynamic reversible process (9). The process of m1A methylation is catalysed by methyltransferases (‘writers’) consisting of tRNA methyltransferase 10 homolog C (TRMT10C), tRNA methyltransferase 61B (TRMT61B), tRNA methyltransferase 6 non-catalytic subunit (TRMT6) and tRNA methyltransferase 61A (TRMT61A), whereas the m1A removal process is mediated by demethylases, including Alkb homolog 1, histone H2A dioxygenase (ALKBH1) and AlkB homolog 3, α-ketoglutarate dependent dioxygenase (ALKBH3) (‘erasers’) (10). In addition, a group of specific RNA-binding proteins composed of YTH domain-containing family protein (YTHD)F1/F2/F3/C1 (‘readers’) can recognize the m1A motif, thus affecting m1A functions (11). The levels of proteins involved in m1A modification are generally greater in gastrointestinal cancer cells than in normal cells (12). The reader protein YTHDF2, which is highly expressed in MM, can promote myeloma cell proliferation through EGR1/p21cip1/waf1/CDK2-cyclin E1 axis-mediated cell cycle transition (13). Methyltransferase 3, N6-adenosine-methyltransferase complex catalytic subunit, as a writer, facilitates multiple myeloma tumorigenesis by enhancing YY1 stability and pri-microRNA-27 maturation (14). TRMT6/61A-dependent base methylation of tRNA-derived fragments regulates tRF-3 silencing activity and the unfolded protein response to promote bladder cancer (15). In addition, cooperation among different methylation molecules has been gradually revealed. For example, AlkB homolog 5, RNA demethylase can regulate STAT3 activity to affect the proliferation and tumorigenicity of osteosarcoma in a YTHDF2-dependent manner (16). FTO α-ketoglutarate dependent dioxygenase also acts as a demethylase to promote MM cell proliferation, migration and invasion in a YTHDF2-dependent manner by targeting heat shock transcription factor 1/heat shock proteins (17). However, only a small number of studies have revealed the involvement of m1A-related genes in the oncogenesis and progression of MM (18). Research focused on the systematic understanding of the role of m1A in MM is warranted.

In the present study, gene expression data and clinical information from the Gene Expression Omnibus (GEO) database were used to explore differentially expressed and prognosis-related m1A regulators, and preliminary verification was carried out.

Materials and methods

MM dataset source and preprocessing

Public gene expression data and full clinical annotations were searched in the GEO database (https://www.ncbi.nlm.nih.gov/geo/). In total, three eligible MM cohorts (GSE13591, GSE47552 and GSE24080) were included (19-21). Specifically, 133 patients with MM and 4 healthy volunteers in GSE13591 and 41 patients with MM and 5 healthy volunteers in GSE47552 were selected for differential gene screening and subsequent analysis. The data were analyzed with the R (v.4.1.2; http://www.R-project.org/) software and R Bioconductor package (https://bioconductor.org/biocLite.R).

Analysis of m1A regulator gene expression

The mRNA expression of m1A regulators in normal plasma and MM bone marrow plasma cells was analyzed based on samples from GSE13591 and GSE47552. Another dataset (GSE24080) with prolific clinical information was subsequently used to elucidate alterations in m1A-related regulatory genes in MM samples. The data were analyzed with the limma R package (http://bioinf.wehi.edu.au/limma).

Unsupervised clustering for 10 m1A regulators

Unsupervised clustering analysis was applied to identify distinct m1A modification patterns based on the expression of m1A regulators, and patients were classified for further analysis. The number of clusters and their stability are determined by the consensus clustering algorithm (22). The Consensus ClusterPlus package (https://bioconductor.org/packages/ConsensusClusterPlus/) was used to perform the aforementioned steps.

Single-sample gene set enrichment analysis (ssGSEA)

The infiltration levels of the different immune cell populations were determined via ssGSEA via the gene set variation analysis (GSVA) R Bioconductor package ((https://github.com/rcastelo/GSVA) with default parameters. The ssGSEA algorithm is a rank-based method that defines a score representing the degree of absolute enrichment of a particular gene set in each sample. The enrichment scores were used to represent the relative abundance of each infiltrating immune cell in each sample.

GSVA and functional annotation

GSVA is a non-parametric and unsupervised method that can easily adapt to the analysis of RNA-seq data (23). GSVA enrichment analysis was performed to investigate the differences in biological processes associated with m1A modification patterns via the GSVA R package. The gene sets of ‘c2.cp.kegg.v6.2.-symbols’ were downloaded from the MSigDB database (https://www.gsea-msigdb.org/gsea/msigdb) for GSVA. Adjusted P-values of <0.05 were considered statistically significant. The clusterProfiler R package (https://yulab-smu.top/contribution-knowledge-mining/) was used to perform functional annotation for m1A-related genes, with cut-off values of |log fold change (FC)| >0.1 and an adjusted P-value of <0.05.

Generation of an m1A gene signature

An m1A gene signature was constructed to quantify the m1A modification pattern. Information on the genes in the signature was used to calculate the m1A score for each MM patient. To identify signature genes, overlapping genes from differentially expressed genes (DEGs) identified in different m1A clusters were first extracted, and the significance criterion was set as an adjusted P-value of <0.001. Unsupervised clustering analysis of these overlapping genes was subsequently performed. The consensus clustering algorithm was used to determine the number and stability of gene clusters, and the patients were divided into three groups for further analysis. Next, univariate regression analysis was used to analyze the prognosis of each overlapping gene and the genes with significant prognostic value were extracted for further analysis. Principal component (PC)1 and PC2 were selected as the gene signature variables, and PC analysis (PCA) was used to construct a scoring model. The m1Ascore was then defined using a method similar to the gene expression grade index (24,25):

m1Ascore = Σ (PC1i+PC2i)

where i is the expression of m1A phenotype-related genes. The feature importance was assessed by the cos2 value and the contributions of the variables.

Validation of the gene signature

The ability of the m1A score to differentiate between patients with MM and controls was assessed in four validation cohorts, GSE24870, GSE27838, GSE46053 and GSE113295 (26-29), via receiver operating characteristic (ROC) curve analysis. The role of m1A regulatory factors in the pathogenesis of MM was also verified via the same method. Visualization of the results was performed via the pROC package (https://xrobin.github.io/pROC/).

Immunohistochemical (IHC) staining

Immunohistochemistry was conducted on bone marrow tissue sections as previously described (30). Bone marrow biopsy sections were obtained from 25 patients with MM at the Department of Oncology and Hematology of the Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine (Jinan, China) and the Department of Hematology of Affiliated Hospital of Shandong University of Traditional Chinese Medicine (Jinan, China) from July 2020 to July 2023. The sections were fixed with formalin, embedded in paraffin, and cut into 4-µm thick sections. The sections were then incubated with 3% H2O2 formaldehyde at room temperature for 10 min, subjected to antigen retrieval in a microwave, cooled at room temperature, and incubated with 10% sheep serum for 10 min. The sections were subsequently incubated overnight at 4˚C with a primary YTHDF2 monoclonal antibody (1:2,000; cat. no. EPR23544-19; Abcam). Biotinylated secondary antibodies (1:2,000; cat. no. ab97080; Abcam) were labelled with streptavidin peroxidase solution, added to the slides, and incubated for 30 min at 37˚C. Finally, the samples were stained (room temperature, 3 min) with a Simple DAB Stain Kit (Abcam) and counterstained with haematoxylin (room temperature, 4 min) according to the manufacturer's instructions. Finally, images were captured directly by optical microscopy (x200) (Leica DM2700 M; Lecia Microsystems GmbH) and analyzed by Image-Pro (Version 7.0; Media Cybernetics, Inc.). All patients provided written informed consent and the protocols were approved from the Ethics Committee of the Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine (Jinan, China; approval no. 2023 Ethics Review-KY-001) and the Ethics Committee of the Affiliated Hospital of Shandong University of Traditional Chinese Medicine (Jinan, China; approval no. 2020 Ethics Review-KY-010).

Cell culture and transfection

The human MM cell line U266 (cat. no. TIB-196; American Type Culture Collection) was cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.) containing 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) and 1% streptomycin-penicillin (Invitrogen; Thermo Fisher Scientific, Inc.) at 37˚C in a 5% humidified CO2 atmosphere. The cells were divided into 5 groups: i) ‘NC’ refers to MM cells without any treatment; ii) ‘si-NC’ refers to MM cells transfected with RNAi that is not homologous to the YTHDF2 sequence; iii) ‘si-YTHDF2’ refers to MM cells transfected with YTHDF2 RNAi; iv) ‘oe-NC’ refers to MM cells transfected with blank plasmid; and v) ‘oe-YTHDF2’ refers to MM cells transfected with the YTHDF2 overexpression plasmid. YTHDF2 RNAi (si-YTHDF2) and YTHDF2-overexpressing vectors (oe-YTHDF2) were designed and synthesized by Shandong Jiehelix Biotechnology Co., Ltd. The sequences of siRNA, including the negative controls are presented in Table SI. U266 cells (1x106 cells/well) were seeded into 6-well plates after transfection with the vectors (50 nM siRNA or 2 µg plasmid DNA) via the Lipofectamine™ 3000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.). After 72 h of transfection (37˚C, 5% CO2), reverse transcription-quantitative PCR (RT-qPCR) was performed to verify the success of the transfection. Total RNA was extracted using SPARKeasy Improved Cell RNAKi (Shandong Sikejie Biotechnology Co., Ltd.), and the concentration and purity of extracted RNA were measured with a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Inc.). SPARKscript II All-in-one RT SuperMix for qPCR Kit and VeritiPro PCR instrument (Thermo Fisher Scientific, Inc.) were used for RT of RNA into cDNA. The RT program was conducted according to the manufacturer's instructions at 50˚C for 15 min and 85˚C for 5 sec. SYBR Green PCR Master Mix (Beijing Solarbio Science & Technology Co., Ltd.) and a Step-One Plus Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used for qPCR analysis. The qPCR program with temperature protocol was conducted according to the manufacturer's instructions as follows: Initial denaturation at 95˚C for 5 min, followed by 40 cycles of denaturation at 95˚C for 30 sec, annealing at 61˚C for 30 sec and extension at 72˚C for 1 min. For the measurement of YTHDF2 mRNA levels, the primers used were as follows: Forward, 5'-GCAAGCAATGTTCCAAAAG-3' and reverse, 5'-GCAATATCAGCCCAAGATG-3'. The relative mRNA levels were normalized to ACTB, the primers used for ACTB were as follows: Forward, 5'-TGACGTGGACATCCGCAAAG-3' and reverse, 5'-CTGGAAGGTGGACAGCGAGG-3'. The relative mRNA expression levels were calculated using the 2-ΔΔCq method (31).

Cell viability assay

Cell viability was determined via the use of Cell Counting Kit-8 (CCK-8) reagents (Beyotime Institute of Biotechnology, Shanghai, China) according to the manufacturer's instructions. Briefly, U266 cells were seeded onto 96-well plates (1,000 cells/well) in the presence of medium containing 10% FBS and penicillin-streptomycin (5,000 U/ml). After 24 h of culture at 37˚C in a 5% humidified CO2 atmosphere, CCK-8 reagent (cat. no. CT0001-B; Shandong Sikejie Biotechnology Co., Ltd.) was added, and the mixture was incubated for 2 h. The absorption at 450 nm was determined with a microplate reader (Tecan Group, Ltd.).

Cell apoptosis assay

The samples were analyzed with a FACSCalibur flow cytometer (BD Biosciences) equipped with an argon laser. A total of 10,000 events were collected for each sample. U266 cells were collected and counted, with the cell concentration adjusted to 1x106 cells/ml, before being resuspended in a cell suspension containing annexin V-PE and 7-AAD (1:100; Dalian Meilun Biology Technology Co., Ltd.). Apoptosis was determined according to the number of Annexin V-PE-positive cells, whereas necrosis was determined according to the 7-AAD staining rate via CellQuest software (version 5.1; BD Biosciences).

Total m1A RNA methylation quantification

Total m1A RNA methylation was quantified as previously described (32). TRIzol (Shandong Sikejie Biotechnology Co., Ltd.) was used to extract total RNA from U266 cells according to the instructions. Total m1A levels in U266 cells were then measured via the Human m1A ELISA kit (cat. no. YJ7129203; Shanghai Enzyme-linked Biotechnology Co., Ltd.) for m1A RNA methylation. In brief, RNA (200 ng) was added to each well, followed by antibody capture and detection. Following incubation, the m1A signal was finally calculated from the calibration curve.

Bioinformatics analysis

Kyoto Encyclopedia of Genes and Genomes enrichment analysis was performed via the Metascape platform (https://metascape.org/). ClueGO (http://apps.cytoscape.org/apps/cluego) was conducted with Cytoscape (version 3.8.2) to explore the biological processes associated with the genes. Pearson correlation analysis was performed and visualized via the corrplot (https://github.com/taiyun/corrplot; v.0.92) and ggplot2 R packages (https://ggplot2.tidyverse.org; v.3.3.5). A correlation coefficient absolute value of >0.7 and P<0.5 were used as the standards to screen related genes. The RMbase database (https://rna.sysu.edu.cn/rmbase3) was used to predict whether YTHDF2 could modify SRSF10 by m1A methylation.

Statistical analysis

The survival curves for the prognostic analysis were generated via the Kaplan-Meier method, and log-rank tests were used to identify the significance of differences. A univariate Cox regression model was adopted to calculate hazard ratios (HRs) for m1A regulators and m1A phenotype-related genes. The independent prognostic factors were ascertained through a multivariable Cox regression model. Patients with detailed clinical data were eligible for inclusion in the final multivariate prognostic analysis. The data processing was performed in R (https://www.r-project.org/; v.4.1.2) software. Two-way ordered categorical variables with different attributes were used to evaluate the differences in the different ISS stage groups among the three clusters using Kruskal-Wallis test and Pearson's correlation analysis. The experimental results were statistically analyzed using SPSS Statistics (version 27.0: IBM Corp.) and GraphPad Prism (Version 10.0; GraphPad; Dotmatics). If each group of data had a normal distribution and homogeneity of variance, quantitative data were presented as the mean ± standard deviation (x̄±s). Differences between two groups were analyzed by Student's t-test, differences among three groups were analyzed by one-way ANOVA with Least Significant Difference post hoc test, differences among >3 groups were analyzed by one-way ANOVA and Kruskal-Wallis test with Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Landscape of genetic variations in m1A regulators in MM

First, the differential expression of m1A regulators between normal plasma (NP; n=9) and MM (n=174) bone marrow plasma cells was summarized. Compared with those in the NP samples, the expression of the reader genes YTHDF1, YTHDF2 and YTHDF3 and the writer genes TRMT61A and TRMT61B were upregulated in the MM samples, whereas the expression of the eraser genes ALKBH1 and YTHDC1 were significantly downregulated (Fig. 1A). Another GEO dataset (GSE24080; n=559) with the most comprehensive clinical annotation was subsequently used to further investigate the prognostic value of m1A regulators. Multivariate survival analysis was performed via the Cox proportional hazard model. The results shown in the forest plot revealed that among the seven differentially expressed m1A regulators, YTHDF2 and YTHDF3 could be independent prognostic factors (Fig. 1B). A subsequent univariate Cox regression model revealed the prognostic value of 10 m1A regulators in patients with MM. The Kaplan-Meier survival curve revealed six regulators that were significantly associated with survival. High expression of five regulators (YTHDF1/2/3, TRMT6 and TRMT61B), especially YTHDF2, was associated with shorter overall survival, whereas YTHDC1 exhibited the opposite pattern (Fig. 1C-H). In addition, the comprehensive landscape of m1A regulator interactions, connections and their prognostic significance in patients with MM was depicted in the m1A regulator network (Fig. 1I). The network diagram revealed that YTHDF2 was significantly positively correlated with the methyltransferase TRMT61B and negatively correlated with the demethylase ALKBH1. These analyses indicated that the variable expression of m1A regulators, especially the high expression of the reader gene YTHDF2, may play a crucial role in MM occurrence and progression.

Figure 1

Landscape of genetic variations of m1A regulators in MM. (A) Alterations of m1A regulatory genes in patients with MM. (B) The forest map of m1A regulators. High expression of TRMT10C, TRMT61B, YTHDF2 and YTHDF3 were associated with poor prognosis in patients with MM. (C-H) High expression of (C) TRMT6, (D) TRMT61B, (F) YTHDF1, (G) YTHDF2 and (H) YTHDF3 is associated with poor prognosis in patients with MM. (E) Low expression of YTHDC1 is associated with a worse prognosis. (I) The interaction among m1A regulators in MM. The circle size represents the effect of each regulator on the prognosis, and the range of values was calculated by the log-rank test. Purple dots in the circle indicate risk factors for prognosis; green dots in the circle indicate protective factors for prognosis. The lines linking regulators showed their interactions, and the thickness shows the correlation strength between regulators. Negative correlations are marked with blue, and positive correlations are marked with red. The regulator erasers, readers and writers are marked with red, orange and gray, respectively. Differences between the MM group and Normal group were analyzed by Student's t-test. A univariate Cox regression model to calculate hazard ratios for m1A regulators and the independent prognostic factors were ascertained through a multivariable Cox regression model. The survival curves for the prognostic analysis were generated via the Kaplan-Meier method, and log-rank tests were used to identify the significance of differences. *P<0.1 and **P<0.01. MM, multiple myeloma; m1A, N1-methyladenosine; TRMT10C, tRNA methyltransferase 10 homolog C; TRMT61B, tRNA methyltransferase 61B; YTHD, YTH domain-containing family protein; TRMT6, tRNA methyltransferase 6 non-catalytic subunit; TRMT61A, tRNA methyltransferase 61A; ALKBH1, Alkb homolog 1, histone H2A dioxygenase; ALKBH3, AlkB homolog 3, α-ketoglutarate dependent dioxygenase.

m1A methylation patterns mediated by 10 regulators

Based on the expression of 10 m1A regulators, three distinct modification patterns were eventually identified via unsupervised clustering: A total of 248 patients exhibited pattern A, 190 patients exhibited pattern B, and 121 patients exhibited pattern C; these groups were termed Clusters A-C, respectively (Fig. 2A-D). There was a significant distinction in the m1A transcriptional profile between Cluster B and the other two clusters (Fig. 2C). The results of PCA feature importance selection revealed that YTHDF2, ALKBH3, TRMT10C and TRMT61B play important roles in clustering differentiation. As revealed in Fig. 2D, the cos2 value in Fig. 2E was used to measure the usefulness of a variable. The higher the value is, the more important the variable is in the PCA. In Fig. 2E, the red dotted line represents the average contribution, and a value higher than the average value can be considered an important variable. As demonstrated in Fig. 2F, Cluster B was characterized by increased expression of YTHDF2, TRMT10C, TRMT61B, TRMT6 and ALKBH3. Prognostic analysis of the three m1A modification subtypes revealed a particular survival disadvantage in Cluster B compared with Clusters A and C (Fig. 2G). Notably, Cluster C, which had better survival than Cluster B, presented a significant decrease in the expression of YTHDF2, indicating that YTHDF2 is likely strongly associated with poor prognosis in patients with MM. The value of m1A clusters in risk stratification prediction for MM was also evaluated, but there was no statistical difference (P=0.057) in the International Staging System risk score among different clusters (Table SII). As expected, subsequent analyses of immune cell infiltration indicated that Cluster B had markedly low levels of infiltration of immune cells, including natural killer cells, macrophages, mast cells, myeloid-derived suppressor cells and immature dendritic cells (Fig. 2H).

Figure 2

m1A methylation modification patterns mediated by 10 regulators. (A and B) Unsupervised clustering analysis. The difference between different clusters was the biggest when k=3. (C) PCA of the transcriptome profiles of the three m1A modification patterns. (D) Variable representative analysis. The length from the center point to the variable represents the proportion of the variable in this dimension. (E) The contribution of variables to PCs. The red dotted line represents the average contribution, and higher than the average value can be considered as an important variable. (F) Unsupervised clustering of 10 m1A regulators. The m1Acluster, ISS staging system, sex, immunoglobulin type and age were used as patient annotations. Blue represents high expression of regulators, and yellow represents low expression. (G) Survival analyses for the three m1A modification patterns. (H) The abundance of immune cells in three m1A modification patterns. Differences among multiple groups were analyzed by one-way ANOVA with LSD post hoc test. The survival curves for the prognostic analysis were generated via the Kaplan-Meier method, and log-rank tests were used to identify the significance of differences. *P<0.1, **P<0.01 and ***P<0.001. m1A, N1-methyladenosine; PCA, principal component analysis; PC, principal component; ISS, International Staging System; ns, no significance; TRMT10C, tRNA methyltransferase 10 homolog C; TRMT61B, tRNA methyltransferase 61B; YTHD, YTH domain-containing family protein; TRMT6, tRNA methyltransferase 6 non-catalytic subunit; TRMT61A, tRNA methyltransferase 61A; ALKBH1, Alkb homolog 1, histone H2A dioxygenase; ALKBH3, AlkB homolog 3, α-ketoglutarate dependent dioxygenase.

To further investigate the potential biological behaviour of each m1A modification pattern, GSVA enrichment analysis was performed between Clusters A and B, which presented prominently different outcomes. As shown in Fig. 3A, Cluster B was markedly enriched in various metabolic pathways, such as ‘purine metabolism’, ‘pyrimidine metabolism’, ‘citrate cycle’, ‘pyruvate metabolism’, ‘cysteine and methionine metabolism’, and ‘fructose and mannose metabolism’. Cluster A was enriched in stromal pathways (Fig. 3A).

Prognostic value and association of the m1A score with immunotherapy response

In the three m1A gene clusters, prominent differences in the expression of m1A regulators were observed. However, these analyses were based on the patient population and could not accurately predict the pattern of m1A methylation in individual patients. Considering the individual heterogeneity and complexity of m1A modification, a scoring system was constructed to quantify the m1A modification patterns of individual patients with MM based on DEGs identified from different m1A clusters. First, the patients were divided into low- and high-m1A score groups with a cut-off value of -2.3971 via the survminer package (https://rpkgs.datanovia.com/survminer/index.html). The Kruskal-Wallis test revealed that patients with a low m1A score had better survival (Fig. 3B). A significantly increased m1A score was detected in Cluster B, and Cluster A presented the lowest median score (Fig. 3C). Patients with high m1A scores had a greater survival rate than those with low m1A scores (74 vs. 54%), which again demonstrated that a low m1A score may be closely associated with a poor prognosis (Fig. 3D and E). Immunotherapies involving PD-L1 and PD-1 blockade have undoubtedly emerged as major breakthroughs in cancer therapy (33). As illustrated in Fig. 3F, patients with a low m1A score exhibited significant increased expression levels of PD-L1, which indicated a potential response to anti-PD-L1 immunotherapy.

Validation of the m1A score in MM

The gene signature-based model was validated via four external datasets containing MM and control samples. Satisfactory model performance was achieved, as determined by the ROC curves, with area under the curve (AUC) values of 0.917, 0.777, 0.667 and 0.625 for the GSE24870, GSE27838, GSE46053 and GSE113295 cohorts, respectively (Fig. 4A-D). Subsequently, two datasets, GSE24870 and GSE27838, which exhibited excellent predictive efficacy, were selected to further demonstrate the important role of m1A regulators in MM and the significance of YTHDF2, YTHDF3, TRMT10C and TRMT6 in the pathogenesis of MM was verified (Fig. 4E and F).

YTHDF2 promotes cellular proliferation and m1A methylation in MM

Through bioinformatics analysis, it was revealed that YTHDF2 expression is associated with poor survival in patients with MM. A series of experiments were subsequently designed to explore the important role of aberrantly expressed YTHDF2 in MM pathogenesis. The IHC results revealed a significant increase in YTHDF2 levels in the high-risk MM patient group compared with those in the low-risk group (Fig. 5A). To further investigate the potential mechanism of action of YTHDF2 in MM, YTHDF2 overexpression and knockdown experiments were performed in U266 cells. CCK-8 assays verified that the si-YTHDF2 group exhibited a significantly lower growth rate than the NC and si-NC groups (Fig. 5B). Flow cytometric analysis revealed that the percentage of apoptotic cells was significantly greater in si-YTHDF2 cells than in control cells (Fig. 5C and D). As revealed in Fig. 5E, a YTHDF2-overexpressing cell model was successfully constructed. Since YTHDF2 serves as a reader of m1A modifications involved in multiple biological processes, including cell differentiation and cancer progression (34), an m1A dot blot assay was performed to assess the global mRNA m1A levels in oe-YTHDF2 cells and si-YTHDF2 cells. An evidently increased m1A level was observed in the oe-YTHDF2 group, whereas YTHDF2 knockdown decreased the overall m1A level of mRNAs (Fig. 5F), indicating that YTHDF2-mediated changes in the m1A level may be involved in MM pathogenesis. Several databases were subsequently used to explore the potential downstream regulatory mechanisms of YTHDF2. First, the aim was to identify the DEGs related to YTHDF2 expression in MM and then to perform functional enrichment analysis on the top 150 related genes associated with the peroxisome proliferator-activated receptor ‘(PPAR) signaling pathway’, ‘protein export’ and ‘various types of N-glycan biosynthesis’ (Fig. 5G). Moreover, ClueGO biological function analysis revealed that these YTHDF2-related genes were enriched in ‘positive regulation of protein targeting to the membrane’, ‘gamma-delta intraepithelial T-cell differentiation’, and ‘positive regulation of anion channel activity’ (Fig. 5H). Next, genes closely related to YTHDF2 expression were screened and it was observed that serine/arginine splicing factor 10 (SRSF10) was significantly positively correlated with YTHDF2 expression (Fig. 5I). SRSF10 is an rRNA-cleaving enzyme that is significantly correlated with a variety of tumours (Fig. 5J). Subsequently, through the RMbase database, it was found that YTHDF2 can install m1A modifications on SRSF10, which is consistent with the findings of a previous study (35). Survival analysis revealed that high expression of SRSF10 indicated a poor prognosis (Fig. 5K), suggesting that YTHDF2 may play a role in MM by methylating SRSF10; however, the underlying mechanism needs to be explored in the future.

Discussion

M1A regulators govern m1A RNA methylation functions. Some research groups have reported that m1A regulators play important roles in tumorigenesis. In the present study, seven differentially expressed m1A regulators from the GEO database were identified, including TRMT61A/B, the reader YTHDF1/2/3 and the eraser ALKBH1 (Fig. 1A). The forest plot and Kaplan-Meier curve revealed that the expression of m1A-related regulatory genes was a reliable prognostic indicator for patients with MM; for example, high expression of YTHDF2 was closely associated with shorter overall survival (Fig. 1B and G). The m1A regulator network diagram revealed a strong positive correlation between YTHDF2 and TRMT61B and a significant negative correlation between YTHDF1 and ALKBH1, suggesting a comprehensive network of different m1A regulators (Fig. 1I). Then, based on the expression of 10 m1A-related genes, three distinct m1A modification patterns were defined via unsupervised cluster analysis (Fig. 2A-C). PCA also revealed that YTHDF2, TRMT10C, TRMT61B and TRMT61A played important roles in the differentiation of the three clusters (Fig. 2D and E). Survival analysis revealed a shorter survival time for patients with MM in Cluster B, which was characterized by a high expression of YTHDF2, TRMT61B and TRMT10C (Fig. 2F and G). Cluster B also exhibited lower immune infiltration, as expected (Fig. 3F). Clusters A and B were subsequently selected for GSVA due to the obvious difference in survival time between the two clusters, and the main differentially enriched pathway was DNA and RNA metabolism progression (Fig. 3A). In addition, m1A score values were established and compared among different m1A clusters. The survival results indicated that patients with MM with low m1A scores had longer survival times, and the m1A score was significantly greater in Cluster B, which was consistent with the survival rate trend in the m1A cluster groups (Fig. 3B-E). Moreover, it was also observed that patients with a high m1A score had lower PD-L1 expression, suggesting the value of the m1A score in predicting the response to anti-PD-L1 immunotherapy (Fig. 3F). Additionally, the m1A score was validated using four external datasets, confirming its robustness with satisfactory AUC values. The identified m1A regulators also exhibited high accuracy in predicting MM diagnosis, suggesting that the developed model may serve as a powerful diagnostic tool for MM.

Considering the aforementioned bioinformatics analysis results, the reader protein YTHDF2 attracted the attention of the authors, due to its high prognostic value in MM. YTHDF2 can directly interact with m1A-modified RNA within the YTH domain, which can disrupt the stability of RNA transcripts (36,37). A previous study revealed that YTHDF2 is overexpressed in acute myeloid leukaemia (AML) and is considered a unique target for the treatment of AML (38). It has also been reported that YTHDF2 can promote MM cell proliferation via the STAT5A/MAP2K2/p-ERK axis and that decreased YTHDF2 expression retards MM tumour growth (39). There is also a YTHDF2-dependent mechanism by which other methylases promote cell proliferation, migration and invasion in MM (17). First, it was verified that YTHDF2 was overexpressed in the high-risk group of patients with MM via immunohistochemistry (Fig. 5A). The CCK-8 and flow cytometric results demonstrated that YTHDF2 plays a proliferation-promoting and apoptosis-inhibiting role in MM (Fig. 5B-D). Gain- and loss-of-function studies revealed that YTHDF2 could increase the level of m1A in U266 cells, which was consistent with previous findings (Fig. 5F). Next, the potential downstream mechanism of YTHDF2 was explored and it was found that its co-expressed genes were enriched in the ‘PPAR signaling pathway’, ‘nucleocytoplasmic transport’, and ‘positive regulation of protein targeting to the membrane’ (Fig. 5G and H). Previous studies have confirmed the crucial role of the PPAR signaling pathway in MM (40). PPARs are nuclear receptor proteins, and there are three main isoforms: PPARα, PPARβ and PPARγ. PPARγ can induce apoptosis in MM cells, and its agonist pioglitazone can enhance the cytotoxic effect of a histone deacetylase inhibitor on MM cells (41). However, YTHDF2 has been reported to bind to PPARs to mediate their degradation, which may be one of the potential mechanisms by which YTHDF2 regulates MM through the PPAR pathway (42). SRSF10 was subsequently selected due to its significant positive correlation with YTHDF2 (Fig. 5I). SRSF10 is a member of the family of mammalian splicing regulators known as SR proteins (43,44). The overexpression of SRSF10 has been verified in a growing list of cancers, as revealed in Fig. 5J, and its contribution to the splicing of transcripts implicated in clinically relevant pathways suggests that SRSF10 could become a treatment target (45). Notably, the activation of SRSF10 was found to dysregulate the polyadenylation of PPAR, and studies have confirmed that YTHDF2 could affect alternative splicing patterns of genes to regulate their expression in a methylation-dependent manner (46-49). Therefore, it was hypothesized that YTHDF2 may regulate SRSF10 through m1A modification to intervene in MM by affecting the PPAR signaling pathway. Moreover, as shown in Fig. 5K, high expression of SRSF10 predicted a shorter survival time, indicating its predictive value for MM prognosis.

However, some limitations should be noted. First, the differentially expressed m1A regulators in MM via the GEO database were identified and their functions were preliminarily predicted via bioinformatics analysis. However, the important role of m1A regulators and the m1A score in MM still needs to be verified in clinical trials and more experiments, such as verification of the expression level of prognosis-related m1A regulators in patients with MM and determination of their predictive ability as clinical prognostic factors via Cox analysis and ROC curves. Second, the role of YTHDF2 in the development, risk stratification and prognosis of MM remains to be further confirmed, and its downstream molecular regulatory mechanism remains to be further explored. Additionally, the regulatory relationship between YTHDF2 and SRSF10 needs to be further verified.

In conclusion, seven differentially expressed m1A regulators and three distinct m1A modification patterns that were associated with differences in overall survival and immune infiltration in patients with MM were identified. The m1A score model was subsequently constructed to quantitatively evaluate the m1A modification patterns of individual patients, which may serve as effective biomarkers for predicting the prognosis of patients with MM. Moreover, the reader protein of m1A, YTHDF2, could increase the m1A level, promote proliferation and inhibit apoptosis in U266 cells, indicating that it may be a potentially crucial target for MM treatment.

Supplementary Material

YTHDF2 siRNA sequences.
Two-way ordered categorical variables with different attributes between different m1A clusters and different risk stratifications.

Acknowledgements

The authors are grateful to Mr. Jingwei Cui (Jiaxuan School of Jinan, Jinan, China) for providing help with data analysis in this paper, especially in the application of R to perform the survival analysis and functional prediction in the bioinformatics section.

Funding

Funding: The present study was supported by the National Natural Science Foundation of China (grant nos. 82074348 and 82274491).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author. The data generated in the present study may be found in the Gene Expression Omnibus (GEO) database under accession numbers GSE13591, GSE47552, GSE24080, GSE24870, GSE27838, GSE46053 and GSE113295 at the following URL: https://www.ncbi.nlm.nih.gov/geo/.

Authors' contributions

XC conceived the project, designed the experiments and revised the paper. JF performed the experiments and wrote the original paper. XH, WG and MY assisted with the experiments, and data analysis and interpretation. All authors read and approved the final version of the manuscript. XC, JF, XH, WG and MY confirm the authenticity of all the raw data.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China (approval no. 2020 Ethics Review-KY-010) and the Ethics Committee of the Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China (approval no. 2023 Ethics Review-KY-001). Written informed consent was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Zhan F, Huang Y, Colla S, Stewart JP, Hanamura I, Gupta S, Epstein J, Yaccoby S, Sawyer J, Burington B, et al: The molecular classification of multiple myeloma. Blood. 108:2020–2028. 2006.PubMed/NCBI View Article : Google Scholar

2 

Allegra A, Casciaro M, Barone P, Musolino C and Gangemi S: Epigenetic crosstalk between malignant plasma cells and the tumour microenvironment in multiple myeloma. Cancers (Basel). 14(2597)2022.PubMed/NCBI View Article : Google Scholar

3 

He L, Yu C, Qin S, Zheng E, Liu X, Liu Y, Yu S, Liu Y, Dou X, Shang Z, et al: The proteasome component PSMD14 drives myelomagenesis through a histone deubiquitinase activity. Mol Cell. 83:4000–4016.e6. 2023.PubMed/NCBI View Article : Google Scholar

4 

Muylaert C, Van Hemelrijck LA, Maes A, De Veirman K, Menu E, Vanderkerken K and De Bruyne E: Aberrant DNA methylation in multiple myeloma: A major obstacle or an opportunity? Front Oncol. 12(979569)2022.PubMed/NCBI View Article : Google Scholar

5 

Liu R, Shen Y, Hu J, Wang X, Wu D, Zhai M, Bai J and He A: Comprehensive Analysis of m6A RNA methylation regulators in the prognosis and immune microenvironment of multiple myeloma. Front Oncol. 11(731957)2021.PubMed/NCBI View Article : Google Scholar

6 

Jin Z, MacPherson K, Liu Z and Vu LP: RNA modifications in hematological malignancies. Int J Hematol. 117:807–820. 2023.PubMed/NCBI View Article : Google Scholar

7 

Zhao BS, Roundtree IA and He C: Post-transcriptional gene regulation by mRNA modifications. Nat Rev Mol Cell Biol. 18:31–42. 2017.PubMed/NCBI View Article : Google Scholar

8 

Dominissini D, Nachtergaele S, Moshitch-Moshkovitz S, Peer E, Kol N, Ben-Haim MS, Dai Q, Di Segni A, Salmon-Divon M, Clark WC, et al: The dynamic N(1)-methyladenosine methylome in eukaryotic messenger RNA. Nature. 530:441–446. 2016.PubMed/NCBI View Article : Google Scholar

9 

Yang Y, Hsu PJ, Chen YS and Yang YG: Dynamic transcriptomic m(6)A decoration: Writers, erasers, readers and functions in RNA metabolism. Cell Res. 28:616–624. 2018.PubMed/NCBI View Article : Google Scholar

10 

Liu Y, Zhang S, Gao X, Ru Y, Gu X and Hu X: Research progress of N1-methyladenosine RNA modification in cancer. Cell Commun Signal. 22(79)2024.PubMed/NCBI View Article : Google Scholar

11 

Zou Z, Sepich-Poore C, Zhou X, Wei J and He C: The mechanism underlying redundant functions of the YTHDF proteins. Genome Biol. 24(17)2023.PubMed/NCBI View Article : Google Scholar

12 

Chen Z, Qi M, Shen B, Luo G, Wu Y, Li J, Lu Z, Zheng Z, Dai Q and Wang H: Transfer RNA demethylase ALKBH3 promotes cancer progression via induction of tRNA-derived small RNAs. Nucleic Acids Res. 47:2533–2545. 2019.PubMed/NCBI View Article : Google Scholar

13 

Liu R, Miao J, Jia Y, Kong G, Hong F, Li F, Zhai M, Zhang R, Liu J, Xu X, et al: N6-methyladenosine reader YTHDF2 promotes multiple myeloma cell proliferation through EGR1/p21cip1/waf1/CDK2-Cyclin E1 axis-mediated cell cycle transition. Oncogene. 42:1607–1619. 2023.PubMed/NCBI View Article : Google Scholar

14 

Che F, Ye X, Wang Y, Wang X, Ma S, Tan Y, Mao Y and Luo Z: METTL3 facilitates multiple myeloma tumorigenesis by enhancing YY1 stability and pri-microRNA-27 maturation in m6A-dependent manner. Cell Biol Toxicol. 39:2033–2050. 2023.PubMed/NCBI View Article : Google Scholar

15 

Su Z, Monshaugen I, Wilson B, Wang F, Klungland A, Ougland R and Dutta A: TRMT6/61A-dependent base methylation of tRNA-derived fragments regulates gene-silencing activity and the unfolded protein response in bladder cancer. Nat Commun. 13(2165)2022.PubMed/NCBI View Article : Google Scholar

16 

Yang Z, Cai Z, Yang C, Luo Z and Bao X: ALKBH5 regulates STAT3 activity to affect the proliferation and tumorigenicity of osteosarcoma via an m6A-YTHDF2-dependent manner. EBioMedicine. 80(104019)2022.PubMed/NCBI View Article : Google Scholar

17 

Xu A, Zhang J, Zuo L, Yan H, Chen L, Zhao F, Fan F, Xu J, Zhang B, Zhang Y, et al: FTO promotes multiple myeloma progression by posttranscriptional activation of HSF1 in an m6A-YTHDF2-dependent manner. Mol Ther. 30:1104–1118. 2022.PubMed/NCBI View Article : Google Scholar

18 

Coira IF, Rincón R and Cuendet M: The Multiple Myeloma Landscape: Epigenetics and Non-Coding RNAs. Cancers (Basel). 14(2348)2022.PubMed/NCBI View Article : Google Scholar

19 

Agnelli L, Mosca L, Fabris S, Lionetti M, Andronache A, Kwee I, Todoerti K, Verdelli D, Battaglia C, Bertoni F, et al: A SNP microarray and FISH-based procedure to detect allelic imbalances in multiple myeloma: An integrated genomics approach reveals a wide gene dosage effect. Genes Chromosomes Cancer. 48:603–614. 2009.PubMed/NCBI View Article : Google Scholar

20 

López-Corral L, Corchete LA, Sarasquete ME, Mateos MV, García-Sanz R, Fermiñán E, Lahuerta JJ, Bladé J, Oriol A, Teruel AI, et al: Transcriptome analysis reveals molecular profiles associated with evolving steps of monoclonal gammopathies. Haematologica. 99:1365–1372. 2014.PubMed/NCBI View Article : Google Scholar

21 

Popovici V, Chen W, Gallas BG, Hatzis C, Shi W, Samuelson FW, Nikolsky Y, Tsyganova M, Ishkin A, Nikolskaya T, et al: Effect of training-sample size and classification difficulty on the accuracy of genomic predictors. Breast Cancer Res. 12(R5)2010.PubMed/NCBI View Article : Google Scholar

22 

Li J, Xie L, Xie Y and Wang F: Bregmannian consensus clustering for cancer subtypes analysis. Comput Methods Programs Biomed. 189(105337)2020.PubMed/NCBI View Article : Google Scholar

23 

Hänzelmann S, Castelo R and Guinney J: GSVA: Gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 14(7)2013.PubMed/NCBI View Article : Google Scholar

24 

Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, Nordgren H, Farmer P, Praz V, Haibe-Kains B, et al: Gene expression profiling in breast cancer: Understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst. 98:262–272. 2006.PubMed/NCBI View Article : Google Scholar

25 

Zhang B, Wu Q, Li B, Wang D, Wang L and Zhou YL: m6A regulator-mediated methylation modification patterns and tumor microenvironment infiltration characterization in gastric cancer. Mol Cancer. 19(53)2020.PubMed/NCBI View Article : Google Scholar

26 

Bruns I, Cadeddu RP, Brueckmann I, Fröbel J, Geyh S, Büst S, Fischer JC, Roels F, Wilk CM, Schildberg FA, et al: Multiple myeloma-related deregulation of bone marrow-derived CD34(+) hematopoietic stem and progenitor cells. Blood. 120:2620–2630. 2012.PubMed/NCBI View Article : Google Scholar

27 

Garg TK, Szmania SM, Khan JA, Hoering A, Malbrough PA, Moreno-Bost A, Greenway AD, Lingo JD, Li X, Yaccoby S, et al: Highly activated and expanded natural killer cells for multiple myeloma immunotherapy. Haematologica. 97:1348–1356. 2012.PubMed/NCBI View Article : Google Scholar

28 

Garcia-Gomez A, De Las Rivas J, Ocio EM, Díaz-Rodríguez E, Montero JC, Martín M, Blanco JF, Sanchez-Guijo FM, Pandiella A, San Miguel JF and Garayoa M: Transcriptomic profile induced in bone marrow mesenchymal stromal cells after interaction with multiple myeloma cells: Implications in myeloma progression and myeloma bone disease. Oncotarget. 5:8284–8305. 2014.PubMed/NCBI View Article : Google Scholar

29 

Liu H, He J, Koh SP, Zhong Y, Liu Z, Wang Z, Zhang Y, Li Z, Tam BT, Lin P, et al: Reprogrammed marrow adipocytes contribute to myeloma-induced bone disease. Sci Transl Med. 11(eaau9087)2019.PubMed/NCBI View Article : Google Scholar

30 

Liu B, Li X, Liu F, Li F, Wei S, Liu J and Lv Y: Expression and Significance of TRIM 28 in Squamous Carcinoma of Esophagus. Pathol Oncol Res. 25:1645–1652. 2019.PubMed/NCBI View Article : Google Scholar

31 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

32 

Frapin M, Guignard S, Meistermann D, Grit I, Moullé VS, Paillé V, Parnet P and Amarger V: Maternal protein restriction in rats alters the expression of genes involved in mitochondrial metabolism and epitranscriptomics in fetal hypothalamus. Nutrients. 12(1464)2020.PubMed/NCBI View Article : Google Scholar

33 

Costa F, Vescovini R, Marchica V, Storti P, Notarfranchi L, Dalla Palma B, Toscani D, Burroughs-Garcia J, Catarozzo MT, Sammarelli G and Giuliani N: PD-L1/PD-1 pattern of expression within the bone marrow immune microenvironment in smoldering myeloma and active multiple myeloma patients. Front Immunol. 11(613007)2021.PubMed/NCBI View Article : Google Scholar

34 

Huang T, Liu Z, Zheng Y, Feng T, Gao Q and Zeng W: YTHDF2 promotes spermagonial adhesion through modulating MMPs decay via m(6)A/mRNA pathway. Cell Death Dis. 11(37)2020.PubMed/NCBI View Article : Google Scholar

35 

Xiao W, Adhikari S, Dahal U, Chen YS, Hao YJ, Sun BF, Sun HY, Li A, Ping XL, Lai WY, et al: Nuclear m(6)A Reader YTHDC1 Regulates mRNA Splicing. Mol Cell. 61:507–519. 2016.PubMed/NCBI View Article : Google Scholar

36 

Dai X, Wang T, Gonzalez G and Wang Y: Identification of YTH Domain-Containing Proteins as the Readers for N1-Methyladenosine in RNA. Anal Chem. 90:6380–6384. 2018.PubMed/NCBI View Article : Google Scholar

37 

Seo KW and Kleiner RE: YTHDF2 Recognition of N1-Methyladenosine (m1A)-Modified RNA Is Associated with Transcript Destabilization. ACS Chem Biol. 15:132–139. 2020.PubMed/NCBI View Article : Google Scholar

38 

Paris J, Morgan M, Campos J, Spencer GJ, Shmakova A, Ivanova I, Mapperley C, Lawson H, Wotherspoon DA, Sepulveda C, et al: Targeting the RNA m6A Reader YTHDF2 selectively compromises cancer stem cells in acute myeloid leukemia. Cell Stem Cell. 25:137–148. e6. 2019.PubMed/NCBI View Article : Google Scholar

39 

Hua Z, Wei R, Guo M, Lin Z, Yu X, Li X, Gu C and Yang Y: YTHDF2 promotes multiple myeloma cell proliferation via STAT5A/MAP2K2/p-ERK axis. Oncogene. 41:1482–1491. 2022.PubMed/NCBI View Article : Google Scholar

40 

Sha Y, Wu J, Paul B, Zhao Y, Mathews P, Li Z, Norris J, Wang E, McDonnell DP and Kang Y: PPAR agonists attenuate lenalidomide's anti-myeloma activity in vitro and in vivo. Cancer Lett. 545(215832)2022.PubMed/NCBI View Article : Google Scholar

41 

Aouali N, Broukou A, Bosseler M, Keunen O, Schlesser V, Janji B, Palissot V, Stordeur P and Berchem G: Epigenetic activity of peroxisome proliferator-activated receptor gamma agonists increases the anticancer effect of histone deacetylase inhibitors on multiple myeloma cells. PLoS One. 10(e0130339)2015.PubMed/NCBI View Article : Google Scholar

42 

Yu JT, Hu XW, Chen HY, Yang Q, Li HD, Dong YH, Zhang Y, Wang JN, Jin J, Wu YG, et al: DNA methylation of FTO promotes renal inflammation by enhancing m6A of PPAR-α in alcohol-induced kidney injury. Pharmacol Res. 163(105286)2021.PubMed/NCBI View Article : Google Scholar

43 

Long JC and Caceres JF: The SR protein family of splicing factors: Master regulators of gene expression. Biochem J. 417:15–27. 2009.PubMed/NCBI View Article : Google Scholar

44 

Longman D, Johnstone IL and Cáceres JF: Functional characterization of SR and SR-related genes in caenorhabditis elegans. EMBO J. 19:1625–1637. 2000.PubMed/NCBI View Article : Google Scholar

45 

Shkreta L, Delannoy A, Salvetti A and Chabot B: SRSF10: An atypical splicing regulator with critical roles in stress response, organ development, and viral replication. RNA. 27:1302–1317. 2021.PubMed/NCBI View Article : Google Scholar

46 

Jobbins AM, Haberman N, Artigas N, Amourda C, Paterson HAB, Yu S, Blackford SJI, Montoya A, Dore M, Wang YF, et al: Dysregulated RNA polyadenylation contributes to metabolic impairment in non-alcoholic fatty liver disease. Nucleic Acids Res. 50:3379–3393. 2022.PubMed/NCBI View Article : Google Scholar

47 

Maimaiti A, Tuersunniyazi A, Meng X, Pei Y, Ji W, Feng Z, Jiang L, Wang Z, Kasimu M, Wang Y and Shi X: N6-methyladenosine RNA methylation regulator-related alternative splicing gene signature as prognostic predictor and in immune microenvironment characterization of patients with low-grade glioma. Front Genet. 13(872186)2022.PubMed/NCBI View Article : Google Scholar

48 

Lai S, Wang Y, Li T, Dong Y, Lin Y, Wang L, Weng S, Zhang X and Lin C: N6-methyladenosine-mediated CELF2 regulates CD44 alternative splicing affecting tumorigenesis via ERAD pathway in pancreatic cancer. Cell Biosci. 12(125)2022.PubMed/NCBI View Article : Google Scholar

49 

Yuan J, Lv T, Yang J, Wu Z, Yan L, Yang J and Shi Y: HDLBP-stabilized lncFAL inhibits ferroptosis vulnerability by diminishing Trim69-dependent FSP1 degradation in hepatocellular carcinoma. Redox Biol. 58(102546)2022.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

January-2025
Volume 29 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fu J, Han X, Gao W, Yu M and Cui X: m1A regulator‑mediated methylation modifications and gene signatures and their prognostic value in multiple myeloma. Exp Ther Med 29: 18, 2025.
APA
Fu, J., Han, X., Gao, W., Yu, M., & Cui, X. (2025). m1A regulator‑mediated methylation modifications and gene signatures and their prognostic value in multiple myeloma. Experimental and Therapeutic Medicine, 29, 18. https://doi.org/10.3892/etm.2024.12768
MLA
Fu, J., Han, X., Gao, W., Yu, M., Cui, X."m1A regulator‑mediated methylation modifications and gene signatures and their prognostic value in multiple myeloma". Experimental and Therapeutic Medicine 29.1 (2025): 18.
Chicago
Fu, J., Han, X., Gao, W., Yu, M., Cui, X."m1A regulator‑mediated methylation modifications and gene signatures and their prognostic value in multiple myeloma". Experimental and Therapeutic Medicine 29, no. 1 (2025): 18. https://doi.org/10.3892/etm.2024.12768