Abstract Background: N6-methyladenosine (m6A) RNA methylation is an important epigenetic modification affecting alternative splicing (AS) patterns of genes to regulate gene expression. AS drives protein diversity and its imbalance may be an important factor in tumorigenesis. However, the clinical significance of m6A RNA methylation regulator-related AS in the tumor microenvironment has not been investigated in low-grade glioma (LGG). Methods: We used 12 m6A methylation modulatory genes (WTAP, FTO, HNRNPC, YTHDF2, YTHDF1, YTHDC2, ALKBH5, YTHDC1, ZC3H13, RBM15, METTL14, and METTL3) from The Cancer Genome Atlas (TCGA) database as well as the TCGA-LGG (n = 502) dataset of AS events and transcriptome data. These data were downloaded and subjected to machine learning, bioinformatics, and statistical analyses, including gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Univariate Cox, the Least Absolute Shrinkage and Selection Operator (LASSO), and multivariable Cox regression were used to develop prognostic characteristics. Prognostic values were validated using Kaplan-Maier survival analysis, proportional risk models, ROC curves, and nomograms. The ESTIMATE package, TIMER database, CIBERSORT method, and ssGSEA algorithm in the R package were utilized to explore the role of the immune microenvironment in LGG. Lastly, an AS-splicing factor (SF) regulatory network was examined in the case of considering the role of SFs in regulating AS events. Results: An aggregate of 3,272 m6A regulator-related AS events in patients with LGG were screened using six machine learning algorithms. We developed eight AS prognostic characteristics based on splice subtypes, which showed an excellent prognostic prediction performance. Furthermore, quantitative prognostic nomograms were developed and showed strong validity in prognostic prediction. In addition, prognostic signatures were substantially associated with tumor immune microenvironment diversity, ICB-related genes, and infiltration status of immune cell subtypes. Specifically, UGP2 has better promise as a prognostic factor for LGG. Finally, splicing regulatory networks revealed the potential functions of SFs. Conclusion: The present research offers a novel perspective on the role of AS in m6A methylation. We reveal that m6A methylation regulator-related AS events can mediate tumor progression through the immune-microenvironment, which could serve as a viable biological marker for clinical stratification of patients with LGG so as to optimize treatment regimens. Keywords: low-grade glioma, alternative splicing, machine learning, tumor immune microenvironment, prognosis, gene signature 1 Introduction Low-grade glioma (LGG) is the most prevalent type of progressive and aggressive brain cancer affecting ∼5,000 adults in the United States annually ([46]Li G. et al., 2021). LGG is a heterogeneous group of neuroepithelial tumors derived from the malignant transformation of astrocytes or oligodendrocytes ([47]Sun et al., 2021). According to the criteria of the World Health Organization (WHO), LGC can be classified into grade II (diffuse low grade) and grade III (intermediate grade) glioma tumors. Although the clinical outcome of LGGs is relatively good compared to grade IV tumors, the survival of patients among those with LGGs ranges between 1 and 15 years ([48]Gargini et al., 2020). However, 70% of patients with LGGs experience high-grade gliomas, recurrence, and death within 10 years ([49]Van Den Bent, 2014). Long-term survival of patients with LGGs is not only dependent on histological presentation, the extent of resection, and radiotherapy status but also a myriad of molecular features. These include isocitrate dehydrogenase (IDH) 1 and 2 mutations, 1p19q coding deletion, chromosome 10 loss, chromosome 7 gain, chromosome 19/20 co-gain, as well as mutations in ATRX, TP53, EGFR, and PTEN ([50]Louis et al., 2021). Nonetheless, as the clinical characteristics of patients with LGGs differ considerably, the progression-free survival (PFS) and overall survival (OS) are significantly diverse, posing a challenge to reliably forecasting prognosis. Thus, a thorough investigation of the modulatory processes of the occurrence and progression of LGG is required to identify biological markers for diagnosis, prognosis, and treatment target identification. Generally, LGG is characterized by epigenetic alteration that demonstrates substantial genetic and phenotypic variability ([51]Condelli et al., 2019). Traditional epigenetic studies have focused on non-coding RNAs, chromatin remodeling, histone modifications, and DNA methylation ([52]Xiang et al., 2020). In recent years, various reversible chemical changes of RNA have increasingly received attention as a new epigenetic modality of regulation ([53]Cui et al., 2021). Internal modification of RNA by N6-methyladenosine (m6A) is thought to be the most prevalent, pervasive, and maintained alteration of RNA in nature. Since the discovery of RNA demethylases and the advancement of RNA methylation sequencing methods, RNA methylation has been recognized as a common phenomenon ([54]Huang et al., 2020) and a critical modulator of RNA translation, stability, alternative splicing (AS), processing, as well as transcription ([55]Lichinchi et al., 2016). The m6A modification predominantly takes place on the adenine of the RRACH motif sequence according to three protein complexes known as the “writer”, “eraser”, and “reader” ([56]Fu et al., 2014). The encoder (writer) is a methyltransferase, and the components of this complex are known to be METTL3, METTL14, WTAP, ZC3H13, and RBM15; while FTO and ALKBH5 serve as demethylases (eraser) to revert methylation; moreover, the recognition of m6A is accomplished by m6A-binding proteins (readers) found to be YTH structural domain proteins (i.e., YTHDF2, YTHDF1, YTHDC2, and YTHDC1) and the HNRNP family of nuclear inhomogeneous proteins (HNRNPC) ([57]Bian et al., 2020). The encoder modulates the buildup of the m6A function, whereas the decoder modulates its depletion. Encoders and decoders are essential for the maintenance of a dynamic equilibrium of the levels of m6A in body cells and tissues. In view of the identification of m6A deposits on natural RNA transcripts in the transcriptional process by readers (m6A binding proteins), they could affect post-transcription gene regulation. The abundance and expression of m6A regulators are generally dysregulated in a variety of cancers, and crucial for the incidence, progression, metastases, recurrence of cancers, and the development of drug resistance in gliomas ([58]Du et al., 2021). For example, METTL3-mediated m6A alterations were found to be remarkably increased in GBM cells that were resistant to temozolomide. Furthermore, the functional overexpression of METTL3 in GBM cells can lead to decreased temozolomide responsiveness ([59]Shi et al., 2021). It was also found that YTHDF2 promotes the decay of UBXN1 mRNA through recognition of METTL3-mediated m6A modifications, thereby activating NF-κB and promoting the malignant progression of glioma ([60]Chai et al., 2021). In addition, METTL3 promotes malignant growth in IDH wild-type gliomas via the mechanism of enhancing MALAT1 stability through m6A alteration with the aid of HuR and by means of activating NF-κB. ([61]Chang et al., 2021). It was also discovered that the Jumonji domain-containing 1C (JMJD1C) protein promoted H3K9me1 demethylation in the microRNA miR-302a promoter region, leading to an elevated level of miR-302a expression. MiR-302a has been discovered as a target of the transcription factor METTL3, which can suppress SOCS2 production by modifying the m6A gene ([62]Zhong et al., 2021). Furthermore, research on the m6A modulatory genes has revealed that they serve as mRNA splicing factors (SFs) for AS and that the m6A genes that participate in the regulation mechanism could interface with AS processes as well. AS events are often observed in human cancer cells, and these events are regulated by m6A modulators. For instance, in pancreatic ductal adenocarcinoma, HNRNPC inhibits m6A-dependent antimetastatic AS events ([63]Huang et al., 2021). In the case where the CLK1/SRSF5 pathway is activated, aberrant exon skipping in the METTL14 and Cyclin-L2 genes are induced, triggering pancreatic ductal adenocarcinoma cell proliferation and metastasis while also modulating m6A methylation ([64]Chen S. et al., 2021). The overexpression of SFs in normal cells could result in the generation of particular pro-oncogenic splice isoforms, thus contributing to the occurrence and progression of cancer. In post-transcription modulation, AS is among the most essential processes. It is also a regulatory process in which the RNA antecedents are preferentially spliced and ligated. Moreover, it has the potential to produce extensive biodiversity ([65]Cai et al., 2019). The assembling of spliceosomes on pre-mRNA is normally regulated by the SF and the integration of certain exons into the mRNA. As a consequence, under several AS modes (i.e., mutually exclusive exons [ME], alternate terminator [AT], alternate acceptor site [AA], alternate promoter [AP], alternate donor site [AD], retained intron [RI], and exon skip [ES]), complete exons may be spliced into mRNA or be omitted ([66]Li X. et al., 2021). However, in pathological conditions, variable splicing of transcripts could result in functional and structural variability of proteins. Among them, several transcripts might serve as potential tumorigenesis-inducing drivers ([67]Siva et al., 2014; [68]Papatsirou et al., 2021). An additional characteristic of cancer is the expression of splice isomers that are not balanced, or the inability to express the right isomer ([69]Wollscheid et al., 2016; [70]Xiong et al., 2018). A recent research report described that the m6A RNA methylation regulator-associated AS gene signature has the potential to anticipate prognosis in non-small cell lung cancer. They further identified different AS events and their potential regulatory mechanisms to expand further comprehension of tumors in transcriptomic mechanisms. More importantly, there is growing evidence that AS performs an instrumental function in the establishment of the immune microenvironment ([71]Li Z. et al., 2019; [72]Deng et al., 2021). Alterations in AS not only influence the infiltration of immune cells but also modulate tumor-related immune cytostatic functions ([73]Yu et al., 2021). Although AS is critical in regulating m6A methylation, the clinical significance of m6A regulator-associated AS in the tumor microenvironment (TME) has not been investigated in LGG. In the research, we study might offer a new perspective for the identification of biological markers predicting LGG prognosis and explore their prognostic significance for patients with LGG. Finally, our exploration of the mechanism of m6A methylation-related AS mediating tumor progression in the TME could offer a novel insight into its impact on LGG prognosis. 2 Materials and Methods Data collection and analysis We queried and acquired gene expression patterns of LGG tissue samples from The Cancer Genome Atlas (TCGA) database ([74]https://portal.gdc.cancer.gov/) for analysis. The relevant clinical-pathological data and Genotype-Tissue Expression (GTEx) data in normal tissues were subsequently obtained from the University of California, Santa Cruz database (UCSC, [75]https://xena.ucsc.edu/), which comprised of 529 LGG tissue samples and 1,152 normal samples (containing 103 normal cerebral cortexes). We did not include patients having inadequate clinical records and lacking follow-up duration information. In this way, a total of 502 LGG samples were included in the present research. In addition, AS events in LGG were downloaded from TCGA Splice Seq ([76]https://bioinformatics.mdanderson.org/TCGASpliceSeq/PSIdownload) and afterward the percentage splicing index (PSI) value, which is a quantitative marker of AS, was calculated, after comparison between subgroups of single and multiple samples, i.e., calculating the percentage value of each AS event, which is commonly utilized in the quantification of AS events. Specifically, for every splicing event in a separate gene, the PSI value is defined as a ratio of the standardized read tally denoting the presence of a transcription component to the overall number of standardized reads (including inclusion and exclusion reads) for that event, with a quantified interval (0–1). Our download includes seven major AS types, namely ME, AT, AP, AA, AD, RI, and ES. Identification and profiling of m6A RNA methylation regulatory genes In total, 12 m6A RNA methylation modulatory genes were chosen for the present research, including 5 “writer”-methyltransferases including METTL3, METTL14, WTAP,ZC3H13,RBM15, and 2 “writer”-methyltransferases demethylases including m6A demethylase ALKBH5 and fat mass and FTO, 5 “reader”-binding proteins in the cell including YTHDC1,YTHDC2,YTHDF1,YTHDF2, and HNRNPC. For this purpose, we input the data of these m6A modulators into Cytoscape (version; 3.9.0) and then performed data analysis with the aid of the ClueGO plugin. We then assessed their function in diagnosis, progression prediction, and patient prognosis of LGG. Afterward, we conducted the GO and KEGG pathway and network analyses using the GO terms biological process (BP), molecular function (MF), and cellular component (CC) with adjusted p < 0.05 as the threshold of statistical significance. Afterward, we selected and analyzed the differential expression of m6A RNA methylation-regulated genes in LGG tissue samples with the aid of the “LIMMA” R package with the threshold value of |log2 fold-change (FC)| ≥1 and an adjusted p < 0.05. Tumor samples were clustered using the R packages “euclidean” and “ward.D2” methods, and the findings of clustering analysis were illustrated by performing the differential expression analysis by virtue of the “pheatmap” R function. An investigation on the relationship between clustering and clinical features was performed by Spearman correlation analysis. With the assistance of the “surv” R package, we conducted univariate and multivariate Cox regression analyses for the purpose of anticipating the correlation between all these m6A RNA methylation-regulated genes and the overall survival of patients. Machine learning analysis We acquired an aggregate of 48,050 AS event features of patients with LGG from the TCGA database using Splice-Seq data. Since the number of features is too large and contains a lot of redundant information causing wastage of computational resources, we used a machine learning method to extract key features. We used AS events to construct the machine learning model and train the model by minimizing the following equation: [MATH: min loss function(pred, label) :MATH] [MATH: s.t. data, label :MATH] [MATH: pred=model(data) :MATH] In which six machine learning models were “boosting”, “bagging”, “XGBoost”, “Adaboost”, “GBDT”, and “randomforest”. where data [MATH: ϵ :MATH] R ^m*n is the AS event data, m is the number of data, n is the dimensionality of the characteristic, pred [MATH: ϵ :MATH] R ^m*n is the results of AS event data predicted with the aid of machine learning models, loss_function denotes the loss function, and the difference between pred and label is measured with second-order Euclidean distance as the loss function so as to optimize the model, achieving an accuracy rate of over 85%. Thus, we extracted the key features of each model, sorted their importance, and observed the prediction effect of the model under the different number of features. Finally, the features are screened as effective features when the prediction effect is stable. Identification of gene AS events with OS of patients with LGG We used the “WGCNA” (Weighted gene co-expression network analysis) R software package to associate AS events with OS in patients with LGG. WGCNA is capable of analyzing a large amount of genomic data to discover genomes correlated with tumor phenotypes. Hence, it enabled us to process data on AS events from clinical features and m6A modulatory gene expression profile association information, while avoiding the use of redundant numerous hypothesis tests and correction procedures. For this purpose, we obtained the AS event data according to the formula A [ij ]= power (S [ij ], β) = |S [mn ]|β (j and i denote the AS event of j and i, respectively, whereas n and m denote the node connections counts, and β denote the appropriate soft threshold power) was computed for the standard scale-free network to produce the appropriate value of β. Then, we used the topological overlap matrix TOMij= [MATH: uAi uAju+Ai< /mi>jmin(K i,Kj)+1Aij :MATH] (i and j denote the AS event associated with i and j, respectively, whereas u denotes clinical characteristics and prognosis data). This step involves the establishment of a weighted adjacency matrix, which was translated in the form of TOM. Subsequently, we employed a dynamic tree cutting approach for the purpose of finding the modules that are strongly correlated with AS events on the basis of hierarchical clustering. 1-TOM was used as the AS event distance measure for depth (threshold value of 2) and least size (threshold value of 60). Thereafter, relatively identical modules were merged with the aid of clustering and a height threshold of 0.3 that was determined by earlier research. Eventually, we carried out Spearman correlation as well as module signature gene analyses on the expression of the m6A modulator gene for the purpose of investigating the correlation between clinical characteristics and prognosis of 502 patients with LGG. Considering previous research, we identified and examined the modules that were most remarkably correlated with clinical characteristics and m6A modulator genes, i.e., an absolute correlation coefficient between m6A regulator and AS event modules exceeding 0.4 with adjusted p < 0.05. Finally, we conducted univariate and multivariate Cox regression analyses to examine the correlation between AS events and the OS of patients with LGG. GO and KEGG enrichment analysis of AS genes Following the screening for AS events related to m6A, we analyzed these OS-associated AS genes using GO terms (CC, MF, and BP) and KEGG pathway analyses. The data for m6A-related AS genes and m6A-modulated genes were further explored with the aid of the ClueGO plugin of Cytoscape. In order to determine the threshold for statistical significance, an adjusted p < 0.05 was used. Following that, we constructed functional networks for the associated genes using Cytoscape. Establishment and evaluation of the m6A RNA methylation regulator-related AS gene risk model To discover m6A regulator-related AS events associated with survival, we performed univariate Cox regression analysis to delve into the correlation between m6A regulator-associated AS events and the OS of patients with LGG (p < 0.05). Then, the obtained data were subjected to visualization with the aid of volcano and UpSet plots. In addition, bubble plots were drawn to demonstrate the 20 most significant AS events across the seven classes of survival-associated AS events. Second, for the purpose of screening candidates within every clipping sequence and preventing model overfitting, we conducted minimal Lasso regression analysis. Ultimately, the seven AS events were integrated into the multi-factor Cox regression analysis with the aim of identifying prognosis predictors. Moreover, prognosis risk scores for predicting OS were produced using the seven AS events. The PSI values of the AS events were utilized to develop a multivariate prognostic model. The formula below was utilized to derive the risk scores: [MATH: Risk score=i=1nβi×PSI :MATH] where n denotes the number of survival-associated AS events, βi denotes the coefficient index of those events, and PSI denotes the PSI value of those events. Based on the median value of the risk scores, the LGG sample was classified into two groups, namely the high- and low-risk groups. To contrast the survival rates between the two groups, we employed the Kaplan-Meier survival curves. Moreover, a two-sided log-rank test, as well as the computation of the area under the curve (AUC), were performed with the aid of the “survivor” and “survminer” R packages. The significance criterion was set as p < 0.05. In addition, with the help of the R package “survival ROC” software, we successfully generate time-dependent receiver operating characteristic (ROC) curves to investigate the prognostic significance of this characteristic. Examination of independent prognostic significance of m6A RNA methylation regulator-related AS risk score For the purpose of determining if risk scores independently served as an indicator of prognosis, univariate and multivariate Cox regression analyses were performed utilizing risk values and accessible clinical data acquired from TCGA cohort. Nomogram as a prognostic model based on AS events and clinicopathological characteristics The model incorporated risk scores for m6A modulator-related AS events in addition to the clinical indicators of LGG, including age, gender, grading, diagnosis, and type (primary and recurrent), as well as mutation status of IDH1 (R132), ATRX, TP53, EGFR, and PTEN. This allowed the optimization of its prediction power, and the “rms” R package was used to determine independent prognostic factors, while relevant clinical parameters in the TCGA cohort were used as variables for the construction of column line plots. We created a horizontal straight line for the purpose of denoting the locations of the point of each variable according to the number of variables. Furthermore, the total number of points for each patient was obtained by adding all the points of the variables and standardizing their distribution between 0 and 100. OS values were calculated for patients with LGGs at 1, 2, and 3 years by placing them between each prognostic axis and the total points axis. Subsequently, we drew calibration plots with the aid of the “rms” R package, and the “rmda” and “devtools” R packages were applied to perform clinical decision curve analysis (DCA) to validate the utility of the column line plots in the cohort. Relationship between risk score and characterization of tumor-infiltrating immune cells We obtained information on the immune infiltration of each sample (including neutrophils, CD8^+ T cells, macrophages, dendritic cells, CD4^+ T cells, and B cells) using Tumor Immune Evaluation Resource (TIMER) ([77]https://cistrome.shinyapps.io/timer/) (TIMER). Then, we probed into the correlation between tumor immune cell (TIC) infiltration and prognosis risk scores. We used the “GSEAbase” R package to perform a single-sample gene set enrichment analysis (ssGSEA) of the enrichment of two separate risk groups in a set of 29 immune function-related genes. Subsequently, the “ESTIMATE” R package was used with the aim of measuring the extent and degree of tumor purity and infiltrated cells (i.e., immune and macrophages cells), thus confirming that the tumor immune milieu features of the two risk groups were significantly different. Identification of cell types was performed based on the levels of 22 distinct types of immune cells present in each tumor sample, which was obtained by measuring the relative subset of RNA transcripts (CIBERSORT; [78]https://cibersort.stanford.edu/). Risk score function in immune checkpoint blockade (ICB) therapy The expression of key genes associated with ICB may correlate with the clinical outcomes following immune checkpoint inhibitor treatment of existing studies. Six critical genes for immune checkpoint blockade treatment, including indoleamine 2,3-dioxygenase 1 (ID O 1), cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed death-ligand 1 (PD-L1, commonly referred to as CD274), PD-L2 (also known as PDCD1LG2), PD-1 (also designated PDCD1), and T-cell immunoglobulin structural domain and mucin-containing structural domain molecule-3 (TIM-3, also named HAVCR2). To reveal possible participants in constructing risk profiles in ICB-treated LGG, AS-based prognostic features were correlated with the expression of the six genes that participate in ICB. Furthermore, we analyzed the expression of 47 genes correlated with ICB in patients belonging to the two groups. Correlation between SFs and AS events as well as their modulatory network SFs can modulate AS events in the TME. As a result, developing a prognosis prediction model founded on AS events calls for simultaneous investigation of the correlation between AS events and SFs. Specifically, the PSI levels of AS events and the expression of potential SFs involved in these events were examined by means of Pearson’s correlation test. Statistical significance was set at p < 0.001 and correlation coefficients (r) > 0.6 or < –0.6. Subsequently, according to the analytic outcomes, we constructed a regulatory network between SFs and AS events, which was visualized with the aid of Cytoscape. Statistical analysis All statistical analyses were performed with the aid of R (version: 4.0.3). The “UpSetR” R package was utilized to examine the intersects and clusters of a variety of AS events. The “ClusterProfiler” R package was used for the purpose of conducting the KEGG and GO analyses. The “survival” and “survROC” R packages were used to perform the survival analysis. The LASSO multivariate Cox analysis was performed with the “glmnet” R package. The rates of survival were analyzed utilizing Kaplan-Meier curves and log-rank test, thus determining the possible statistical significance. Pearson correlation tests were used to investigate the correlation between risk scores, clinical factors, the extent of immune cell infiltrates, and immunological checkpoints. A criterion of p < 0.05 denoted statistical significance. 3 Results Clinical characteristics and AS event profiles in LGG We obtained LGG expression profile data from the TCGA database, as well as clinicopathology data from the UCSC database that were used in the present research. We obtained 502 LGG cases for data analysis. For the LGG samples, there were 278 males and 224 females, 232 patients aged <40 years, and 270 individuals aged ≥40 years. The patients were graded as G2 (n = 241) and G3 (n = 261) ([79]Table 1). We observed 48,050 AS events in the LGG tissue sample, with ES being the most common of all AS events. AP ranked second as the most prevalent, with ME ranking last. TABLE 1. Baseline data of all LGG patients. Characteristic Type n Proportion (%) Age <40 232 46.22 ≥40 270 53.78 Gender Female 224 44.62 Male 278 55.38 Grade G2 241 48.01 G3 261 51.99 Diagnoses Astrocytoma, anaplastic 129 25.70 Astrocytoma, NOS 61 12.15 Mixed glioma 128 25.50 Oligodendroglioma, anaplastic 76 15.14 Oligodendroglioma, NOS 108 21.51 Type Primary Tumor 483 96.22 Recurrent Tumor 19 3.78 Chr 19/20 co-gain Gain chr 19/20 12 2.39 No chr 19/20 gain 487 97.01 unknow 3 0.60 Chr 7 gain/Chr 10 loss Gain chr 7 & loss chr 10 55 10.95 No combined CNA 444 88.45 unknow 3 0.60 IDH1 R132status Mutation 386 76.89 Wild 116 23.11 PTEN status Mutation 29 5.78 Wild 473 94.22 EGFR status Mutation 30 5.98 Wild 472 94.02 ATRX status Mutant 185 36.85 WT 317 63.15 TP53 status Mutation 229 45.62 Wild 273 54.38 [80]Open in a new tab Correlation between m6A RNA methylation regulatory gene expressions and LGG prognosis In the present research, we examined 12 m6A genes that were involved in the regulation of RNA methylation. We carried out GO terminology and KEGG pathway analysis and the findings demonstrated a remarkable enrichment of these modulators of m6A in mRNA spliceosome biological processes, RNA modifications (RNA methylation biological processes), and RNA instability biological processes ([81]Figure 1A). In addition, the findings recorded from the Wilcoxon rank-test demonstrated a differential expression in normal and LGG tissues except for WTAP (p < 0.001) ([82]Figure 1B). The results obtained from the univariate Cox regression illustrated that the expression of RBM15, WTAP, YTHDF1, METTL3, YTHDC1, FTO, and YTHDF2 predicted OS in patients with LGG (p < 0.05) ([83]Figure 1C). Therefore, we included BM15, METTL3, YTHDF1, YTHDC1, FTO, and YTHDF2 regulatory genes as subsequent SFs for association analysis. FIGURE 1. [84]FIGURE 1 [85]Open in a new tab Relationship between the methylation status of the m6A RNA modulatory genes and the prognosis of patients with LGG (A) The GO terms and KEGG enrichment pathways analyses of the m6A modulator genes; the various colors reflect the various pathways. (B) In LGG, 12 m6A RNA-methylated regulator genes exhibit differential expression (C) The forest plot representing the analytical data for the univariate Cox regression. The 12 m6A RNA methylation modulators in LGG examined by means of univariate Cox regression and visualized by a forest plot. (D–I) Six algorithms applied to screen the potential AS. The number of features with the maximum accuracy and least margin of error after fivefold cross-validation in “Boosting”, “Bagging”, “XGBoost”, “Adaboost”, “GBDT”, and “Randomforest”. ***p < 0.001, **p < 0.01, and *p < 0.05. Development and verification of the AS events based on multiple machine learning algorithms We used six machine learning models such as “boosting”, “bagging”, “XGBoost”, “Adaboost”, “GBDT”, and “randomforest” to further elucidate the selected AS events. We used the final merged set of valid features for each of the six models as the variable clipping events for the next task, and a total of 3,272 valid features were extracted for the variable clipping events. We then visualized the features filtered by each machine learning model ([86]Figures 1D–I). After comparison, we found that using the above-combined features, the prediction effect is basically comparable to the full number of features but reduces the number of operations by 90%, thus maximizing the prediction effect under the condition of limited computational resources. Identification of m6A RNA methylation regulatory genes with LGG clinical characteristics In the present research, we examined the relationship between AS events and weighted gene co-expression networks. The results demonstrated that they were congruent to the scale-free network ([87]Figure 2B). The log10 transformed RNA-seq scores were discovered by performing hierarchical cluster analysis of the samples using Euclidean distance ([88]Figure 2A), whereas the dynamic tree cutting technique exposed modules with similar expression patterns and combined similar modules ([89]Figure 2C). We then analyzed AS events using the “WGCNA” R package and linked m6A regulator gene expression to clinical traits using Spearman correlation tests. FIGURE 2. [90]FIGURE 2 [91]Open in a new tab Determination of the AS events that are correlated with clinical characteristics and the m6A regulator expression (A) Sample dendrogram and trait indicator. (B) Assessment of the soft-threshold power of WGCNA analysis (C) The AS events in LGG samples are clustered hierarchically. The tree represents an LGG with a distinct name and experiment identification. (D–G) Genes associated with m6A-related AS events in LGG were subjected to analysis using GO terms and KEGG pathways. The GO terms (D–F) and KEGG pathways enrichment (G) analysis of the m6A-related prognostic AS genes in LGG (H) Association of the AS events with m6A methylation regulatory genes, age, gender, grade, Chr 7 gain/Chr 10 loss, as well as Chr 19/20 co-gain, mutations status of IDH1 (R132), IDH2 (R172), PTEN, EGFR, ATRX, and TP53. The findings revealed that the MEyellow module was related to the expression of genes regulating m6A in patients with LGG (RBM15, P = 8e-11, r = –0.3; METTL3, P = 3e-11, r = 0.3; FTO, P = 3e-22, r = –0.43; YTHDF2, P = 8e-05, r = –0.18), and also significantly correlated with survival status (P = 6e-6, r = –0.21), grading (p = 0.02, r = –0.11), and PTEN mutation status (p = 0.03, r = –0.1). In addition, MEred, MEturquoise, MEblue, and MEbrown modules exhibited a remarkable association with the expression of genes regulateing m6A (MEred: RBM15, adjusted P = 2e-29, r = –0.49; METTL3, adjusted p = 0.008, r = 0.12; YTHDF1, adjusted P = 8e-9, r = –0.27; YTHDC1, adjusted P = 4e-5, r = 0.19; FTO, adjusted P = 3e-9, r = 0.27; YTHDF2, adjusted P = 2e-42, r = –0.58; MEturquoise: RBM15 p = 0.04, r = –0.096; METTL3, adjusted P = 4e-64, r = 0.68; YTHDF1, adjusted P = 9e-13, r = 0.32; YTHDC1, adjusted P = 4e-10, r = 0.29; FTO, adjusted P = 3e-20, r = –0.41; YTHDF2, adjusted p = 0.03, r = –0.1; MEblue: RBM15, adjusted P = 2e-10, r = 0.29; METTL3, adjusted P = 8e-6, r = –0.21; YTHDF1, adjusted P = 2e-4, r = 0.17; YTHDC1, adjusted P = 1e-15, r = –0.36; FTO, adjusted P = 4e-9, r = –0.27; YTHDF2, adjusted P = 7e-28, r = 0.48; MEbrown: RBM15, adjusted P = 3e-22, r = 0.43; METTL3, adjusted P = 7e-15, r = 0.35; YTHDF1, adjusted P = 7e-20, r = 0.41; YTHDC1, adjusted P = 1e-10, r = 0.3; FTO, adjusted p = 0.002, r = –0.15; YTHDF2, adjusted P = 7e-5, r = 0.18). In addition to this, the MEred, MEblue, and MEbrown modules were also correlated with the tumor grade of the patient (MEred: adjusted P = 1e-25, r = –0.46; MEblue: adjusted P = 1e-10, r = 0.29; MEbrown: adjusted P = 2e-5, r = 0.2), Chr 7 gain/Chr 10 loss (MEred: adjusted P = 1e-20, r = –0.42; MEblue: adjusted P = 8e-61, r = 0.67; MEbrown: adjusted p = 0.006, r = 0.13), and PTEN mutation status (MEred: adjusted p = 0.003, r = –0.14; MEblue: adjusted P = 3e-8, r = 0.25; MEbrown: adjusted p = 0.03, r = 0.1) ([92]Figure 2H). The above findings revealed that m6A-related AS events in the MEbrown, MEblue, and MEred modules forecast LGG progression and tumor grade, whereas the gain or loss of chromosomes and PTEN mutation status of patients may also influence m6A-related AS events. In addition, the MEred module included 208 AS events, the MEyellow and MEbrown modules both 386 AS events, the MEturquoise module 1,181 AS events, and the MEblue module 768 AS events. Analysis of GO terms and KEGG pathways in the LGG cohort showed that AS events associated with m6A were significantly enriched in mitogen-activated protein kinase (MAPK) signaling pathway, cancer signaling pathway, PI3K-Akt signaling pathway, neurodegenerative ailments, particularly, Alzheimer’s disease, Huntington’s disease, and processes of the nervous system, cellular metabolic processes, intracellular signaling, cell cycle regulation, regulation of catalytic activity, regulation of hydrolase activity, etc. ([93]Figures 2D–G) Identification of m6A-Related AS events with LGG prognosis For the purpose of revealing the correlation between m6A-related AS events and LGG prognosis, we conducted a univariate Cox regression analysis of the data. With the aid of the UpSet plot, we identified 750 prognostic AS events in LGG, with intersecting genomic and splice subtypes (p < 0.05; [94]Figures 3A,B, [95]Supplementary Table S1). ES was the most prevalent pattern compared with other subtypes of AS events, followed by AP and ME, with ME being the least common pattern. Volcano plots were created for the purpose of visualizing the events of AS ([96]Figure 4A). The 20 key AS events associated with survival in the seven subtypes is summarized in [97]Figures 4B–H. FIGURE 3. [98]FIGURE 3 [99]Open in a new tab (A). An upset plot depicting interactions between distinct m6A methylation regulator-related AS types In LGG. (B). Upset plot showing survival-associated m6A methylation regulator-related AS types. FIGURE 4. [100]FIGURE 4 [101]Open in a new tab (A). The volcano plots of AS events associated with the m6A methylation regulator that is survival-relevant. In the TCGA-LGG cohort, the most crucial m6A-relevant RIs MEs, ESs, ATs, APs, ADs, and AAs were identified (B–H). Construction and verification of the prognostic risk score model based on m6A-related AS events To examine the ability of m6A regulator-related AS events in predicting patient prognosis, the above 7 AS events and their combinations were further subjected to LASSO regression and screened for the most important m6A regulator-related AS events. [102]Figures 5A–H shows the results of the LASSO regression analysis. A multivariate Cox regression model was then employed on the independent prognostic indicators. A risk score was generated for every patient, and complete details on the prognostic factors based on the seven AS events are provided in [103]Table 2. Patients with LGG were classified into two groups (low- and high-risk groups) according to their risk scores. [104]Figures 6A–H depicts data on the kinds of potential AS events, as well as data on survival time and survival statuses, which are arranged based on the distributions of risk values. Patients in the low-risk group exhibited a more favorable prognosis as opposed to the ones in the high-risk subgroup, according to the findings of the Kaplan-Meier survival analysis (p < 0.05, [105]Figures 7A–H). For the purpose of assessing the prognostic accuracy of the variables across time, a time-dependent ROC analysis was performed at 1, 3, and 5 years. In all cases with AS event characteristics, the ROC AUC values were over 0.7, indicating strong prognostic prediction ability ([106]Figures 7A–H). The above findings suggest that the AS-based prognostic signature and risk scoring system has the potential to be used as a new method for LGG classification. FIGURE 5. [107]FIGURE 5 [108]Open in a new tab (A–H). AS events associated with the m6A methylation regulator and their correlation with the prognosis of patients with LGG were identified and validated utilizing LASSO regression. The 10-round cross-validation method was used for the purpose of evaluating these 12 m6A-related AS events that were correlated with the prognosis of patients with LGG, as well as the optimum levels of the penalty parameter. TABLE 2. Multivariate Cox analysis of prognostic N6-Methyladenosine-Related AS signatures of each AS type. Type Gene symbol Splice-seq CD AS type Coef HR HR95L HR95H P-value Integrated AS UGP2 53745 AP 4.879983645 131.6285111 19.19928284 902.432924 6.75E-07 SET 87776 AP −4.394829337 0.012340986 0.000431926 0.352606634 0.010187646 SDR39U1 27009 AA 3.2291677 25.25862547 0.527549531 1209.361629 0.101845454 RPAIN 38691 ES 3.338637289 28.18069839 3.858762106 205.8047995 0.000998063 RAD52 19633 AP 1.411443796 4.101873398 0.932391021 18.04539618 0.06185279 AA SDR39U1 27009 AA 6.084249222 438.8901798 15.58619807 12358.66432 0.000353431 SEH1L 44727 AA 3.297355611 27.04103728 3.140548345 232.8312182 0.002684096 GRK6 74765 AA 2.321697441 10.19296158 1.327158745 78.28488208 0.02560975 TMEM198 57724 AA 1.800566228 6.053073916 0.916948689 39.95829241 0.06149485 CARM1 47598 AA −1.506535512 0.221676645 0.071078446 0.69135635 0.009432271 MT1E 36480 AA −1.84909159 0.157380067 0.056506714 0.438328189 0.000402925 PARP3 65117 AA 3.146071769 23.24457494 3.012209603 179.3733953 0.002547726 POLR2J3 81117 AA 2.046993192 7.7445796 1.760038909 34.07794729 0.006773064 NIF3L1 56774 AA 3.053794574 21.19562036 4.631422605 97.00136673 8.31E-05 AD IP6K2 64759 AD 1.922510982 6.838107292 1.876547772 24.91794349 0.003568075 UQCRQ 73319 AD −1.17436915 0.30901386 0.092460845 1.032756793 0.056441926 ATP5J 60271 AD −3.175511473 0.041772733 0.00084905 2.05519287 0.110141838 SEPT4 42700 AD 8.554871125 5191.983732 88.70457802 303892.9408 3.79E-05 STRA6 31688 AD −2.031010147 0.13120292 0.05125151 0.335877052 2.29E-05 PSMC3IP 41077 AD −1.159759969 0.313561436 0.119740086 0.821118285 0.018214528 NSG1 68675 AD 0.850737895 2.341373902 1.007619205 5.440578862 0.047972912 COPS7A 19958 AD 5.018203401 151.1395227 20.88297137 1093.865184 6.72E-07 INPP4A 54629 AD 1.708539296 5.520891197 1.366237351 22.30962254 0.016487465 LRRC23 20008 AD −1.553350033 0.211538126 0.053381317 0.838277909 0.027031513 HHLA3 3405 AD −1.48302156 0.226950905 0.047902498 1.075240657 0.061684026 AP UGP2 53745 AP 4.312239301 74.60737036 8.759537721 635.4513091 7.96E-05 SET 87776 AP −5.016555712 0.006627314 0.000343099 0.128013404 0.000898039 RAD52 19633 AP 1.940344746 6.961150388 1.533485812 31.59964985 0.011940996 CASQ1 8432 AP −1.87466241 0.153406747 0.01906347 1.234488279 0.078074254 CHEK1 19309 AP 2.040848212 7.69713523 1.219051932 48.59997282 0.029958873 AT RGR 12401 AT −1.52922525 0.216703493 0.065338895 0.718720508 0.01242313 SEPT8 73300 AT −1.932573674 0.144775115 0.026213655 0.799576921 0.026657461 CHCHD3 81833 AT 2.770001828 15.95866318 1.355092185 187.942144 0.027703128 NRG1 83312 AT −2.437481846 0.087380613 0.027533352 0.277313543 3.52E-05 LGALS3 27617 AT 3.525390265 33.96702718 2.214634727 520.970308 0.011382795 FANCD2 63307 AT 4.12329134 61.76218845 3.510041307 1086.758698 0.004830142 PSMB7 87532 AT −2.579926544 0.07577957 0.006666101 0.861454613 0.037506205 ES NDRG2 26517 ES 1.855248404 6.39328617 1.530859794 26.70009901 0.010963972 IP6K2 64760 ES 2.795037686 16.3632454 5.32543777 50.27864596 1.06E-06 MYO19 40481 ES −1.29099287 0.274997611 0.089421889 0.845695463 0.024299236 TNFRSF11B 84998 ES −0.944608524 0.38883176 0.168134419 0.899221814 0.027223571 CARD8 50713 ES 1.393842997 4.030308793 1.650740804 9.840060248 0.002209495 CCDC136 81717 ES 1.463262352 4.320030022 1.635017641 11.41434742 0.003159835 ME DLG3 89383 ME 1.675000904 5.338799977 2.052995404 13.88351145 0.000592306 SRGAP1 93242 ME 1.803645895 6.0717441 1.260351051 29.25064124 0.024549824 MAPK10 69825 ME −3.167912676 0.042091365 0.002548083 0.695300238 0.026832968 PCBP4 65134 ME −2.387692442 0.091841369 0.010513434 0.802291368 0.030837457 RAD51 30020 ME −3.274891158 0.037820986 0.004606013 0.310556457 0.002299698 SDR39U1 27012 ME −1.045775084 0.351419334 0.134765333 0.916374752 0.032472257 RI NPIPA5 34148 RI −2.202669028 0.110507816 0.030044922 0.406457286 0.000917135 MED22 88029 RI 1.960669691 7.104083012 0.867257724 58.19261571 0.067664118 ABCC5 67820 RI 6.542764727 694.2032097 31.99629894 15061.68251 3.08E-05 TMEM107 39107 RI 2.317084378 10.1460491 1.982930083 51.91424202 0.005404926 WDR62 49339 RI −1.34359868 0.260905063 0.069045763 0.985888914 0.047600621 GGA3 43399 RI 4.3686116 78.93396369 8.3100749 749.761067 0.000142641 PPP1CC 24503 RI −10.12349163 4.01E-05 4.42E-07 0.003642107 1.08E-05 TEX9 30763 RI −1.760061746 0.172034241 0.030293932 0.976954054 0.047002651 [109]Open in a new tab FIGURE 6. [110]FIGURE 6 [111]Open in a new tab (A–H). The risk scores associated with seven different kinds of m6A methylation regulator-associated AS signatures and combined AS signatures in patients with LGG. The spread of overall survival for patients with different risk scores is shown in the top panel of the figure. The variance trend in patient survival time with risk scores is shown in the center panel. The heatmap of AS events associated with the survival-related m6A methylation modulator is shown in the lower panel. FIGURE 7. [112]FIGURE 7 [113]Open in a new tab (A–H). Treatment outcome in the LGG cohort determined by survival analysis of the patients using the m6A methylation regulator-related AS signature and the Verification of the predictive efficacy of all AS signatures and the combined AS signature. The Kaplan-Meier survival curves of the patients in the high- and low-risk groups are shown in the left panel. The right panel depicts the ROC curve illustrating the prognostic precision of the individual AS signature and the combined AS signature. Identification of m6A-Related AS events signature independence for prognostic prediction and development of AS-clinicopathological nomogram To explore whether the prognostic significance of m6A regulator-associated AS events is independent of clinicopathological variables, we assessed the tumor grade, gender, LGG diagnosis type, age, tumor type (recurrent and primary), as well as the mutation statuses of IDH1 (R132), ATRX, EGFR, TP53, and PTEN. and m6A regulator-associated AS events based on the univariate and multivariate Cox regression analyses that were conducted for the prognostic risk score model. The findings of the multivariate analysis demonstrated that the risk score was remarkably correlated with OS of patients belonging to the two groups in the TCGA set (p < 0.001). In addition, five other clinical factors, namely age, grade, LGG diagnosis type, type (primary and recurrent), and IDH1 (R132) mutation status were also significant (p < 0.05) ([114]Table 3 and [115]Figures 8A,B). We also drew ROC curves comparing risk scores with other clinicopathological factors. The findings illustrated that risk score performed better in contrast with tumor grade, gender, age, LGG diagnosis type, tumor type (recurrent and primary), as well as mutation status of IDH1 (R132), ATRX, EGFR, TP53, and PTEN, along with some other factors ([116]Figures 8C–E). These findings suggest that the prognostic performance of risk score for survival prediction in LGGs is significantly higher. TABLE 3. Univariate and Multivariate Cox Proportional-Hazards analysis for the N6-Methyladenosine-Related AS Events Riskscore and overall survival in LGG patient cohorts. Variables Univariable analysis Multivariable analysis HR 95%CI P-value HR 95%CI P-value N6-Methyladenosine-Related AS Events Riskscore High/Low 1.039 1.029–1.048 <0.001 1.033 1.020–1.045 <0.001 Age 1.060 1.044–1.075 <0.001 1.059 1.042–1.076 <0.001 Gender Female/Male 1.053 0.735–1.509 0.778 1.135 0.782–1.649 0.505 Grade G2/G3 3.316 2.231–4.928 <0.001 1.840 1.167–2.900 0.009 Diagnoses Astrocytoma, anaplastic/Astrocytoma, NOS/Mixed glioma/Oligodendroglioma, anaplastic/Oligodendroglioma, NOS 0.756 0.667–0.856 <0.001 0.795 0.683–0.926 0.003 Type Primary/Recurrent 1.735 0.845–3.560 0.133 2.116 1.010–4.432 0.047 IDH1 R132status Mutation/Wild 2.757 1.901–3.999 <0.001 1.824 1.076–3.092 0.026 EGFR status Mutation/Wild 0.318 0.186–0.544 <0.001 0.716 0.354–1.447 0.352 ATRX status Mutation/Wild 1.398 0.966–2.025 0.076 1.093 0.651–1.836 0.737 TP53 status Mutation/Wild 1.270 0.889–1.815 0.189 0.943 0.576–1.542 0.814 PTEN status Mutation/Wild 0.517 0.283–0.943 0.032 1.927 0.861–4.313 0.111 [117]Open in a new tab FIGURE 8. [118]FIGURE 8 [119]Open in a new tab (A) nomogram based on the AS signature and clinical characteristics associated with the m6A methylation regulator. Cox regression evaluations on univariate (A) and multivariate (B) data, as well as the model, indicating excellent prognostic performance independent of clinical variables. (C–E). ROC curve for anticipating overall survival (OS) at 1, 3, and 5 years used to validate the model’s prognostic accuracy in contrast with other individual components. (F) A nomogram created using the risk score, grade, diagnoses, age, and type data, as well as the IDH1 (R132) status. (G–I). Calibration plots showing agreement in anticipating the OS at 1, 2, and 3 years. (J–L). The DCA curve demonstrating agreement in anticipating OS at 1, 2, and 3 years. The nomogram prognostic score system was created with the aim of effectively applying the findings of the present research to clinical practice. It predicts the 1-, 2-, and 3-years OS of patients according to the above-mentioned clinical parameters. This scoring system includes age, tumor grade, LGG diagnosis type, tumor type (primary and recurrence), IDH1 (R132) mutation status, and the risk score of m6A regulator-related AS events ([120]Figure 8F). Subsequently, to validate the reliability of this model, we performed calibration plots analysis ([121]Figures 8G–I), and the findings of the present research confirmed the applicability of the model in real-world situations. Hence we validated the data presented above by means of clinical DCA to investigate if nomogram plots could accurately anticipate 1-, 2-, and 3-years OS of patients ([122]Figure 8J–L). The findings revealed that nomogram plots exerted remarkably improved performance in anticipating patient prognosis as opposed to any of the independent variables. To highlight the robust prognostic value of nomogram, we compared the efficacy of other signatures from references. Currently there are only