Skip to main content

m6A RNA methylation regulator-based signature for prognostic prediction and its potential immunological role in uterine corpus endometrial carcinoma

Abstract

Background

Uterine corpus endometrial carcinoma (UCEC) is the most common female pelvic malignancy worldwide. N6-methyladenosine (m6A) plays an important role in various cellular responses, especially in cancer progression. However, the correlation between prognostic UCEC and m6A RNA methylation regulators remains unclear.

Methods

We used The Cancer Genome Atlas (TCGA) to provide a gene signature that could improve the prognostic evaluation of UCEC patients according to the distinct genetic trait of m6A RNA methylation regulators from a bioinformatics perspective. After comparing UCEC subgroups with different genetic profiles of m6A regulators, we identified 71 differentially expressed genes associated with overall survival (OS) and generated a nine-gene signature through least absolute shrinkage and selection operator (LASSO) Cox regression analysis. Finally, we used in vitro and in vivo tumor cell experiments as well as the immune correlation analysis to verify the function of each gene in the proposed gene signature.

Results

Time-dependent receiver operating characteristic (ROC) curves revealed that the proposed gene signature could predict the outcome of UCEC patients accurately. We found that CDKN2A mainly acted from the perspective of tumor cells, while COL4A4, PXDN, TIGIT, CHODL, LMO3, KCNJ12, L1CAM, and EPHB1 might play a role in UCEC from an immunological point of view.

Conclusions

From an epigenetics perspective, the m6A RNA methylation regulator-based gene signature can predict the prognosis of UCEC patients and immune therapeutic efficacy.

Peer Review reports

Background

Uterine corpus cancer is the most common female pelvic malignancy (with an estimated 65,620 new cases) and ranks as the third leading cause of gynecological cancer-related death (estimated to be 12,940, following breast cancer at 43,600 and ovarian cancer at 22,950) [1]. Endometrial carcinomas account for most of these cases and encompass more than 83% of uterine corpus cancers [2]. The incidence of uterine corpus endometrial carcinoma (UCEC) is increasing due to prolonged life expectancy and early diagnosis. However, most patients face a risk of recurrence after surgery or multi-therapies, leading to a poor prognosis. Thus, it is urgent to identify novel therapeutic targets and prognostic biomarkers to provide personalized treatment for UCEC. In recent years, comprehensive studies of new prognostic signatures based on epigenetic characteristics have been elucidated [3, 4]. Cancer cell-intrinsic epigenetic alterations have been associated with carcinogenesis and tumor progression. However, few studies have reported prognostic signatures based on epigenetics in UCEC, possibly because epigenetics is not widely studied in UCEC.

N6-methyladenosine (m6A) methylation modification refers to the addition of a methylated group to the N6 position of adenosine, which is the most abundant internal modification of eukaryotic mRNA and lncRNA relative to N1-methyladenosine (m1A) and 5-methylcytosine (m5C) [5]. Modification by m6A is a dynamic and reversible process, which is regulated by m6A methyltransferases, m6A demethylases, and m6A-binding proteins, i.e., writers, erasers, and readers (WERs). In the process, methylation and demethylation can occur simultaneously. The installation of m6A can affect alternative pre-mRNA splicing, 3′-end processing, mature mRNA nuclear export, and mRNA degradation. Therefore, it can affect gene expression at the transcriptional level in several ways and is directly linked with protein translation [6, 7]. Methylation of mRNA m6A is essential for different physiological activities, including human diseases, by impacting gene transcription [8]. The m6A regulators are closely associated with the malignancy in various cancers because m6A regulators can directly affect the expression of oncogenes or tumor suppressors, and certain m6A regulators serve as prognosis biomarkers for certain cancers [9, 10]. The m6A methylation in nearly 70% of endometrial tumors is reduced, thus promoting the proliferation and oncogenicity of endometrial cancer cells [11]. However, the correlation between UCEC prognostics and m6A RNA methylation status remains unknown.

Recently, numerous studies have found that m6A modification can participate in regulating the composition of the tumor microenvironment (TME) infiltrating cells to modulate the immune response [12]. For example, RNA methyltransferase METTL3-mediated m6A methylation can promote dendritic cell (DC) activation and antigen-presenting cells (APC)-based T cell response [13]. YTHDF1 depletion can enhance CD8+ T cell-related response and immuno-therapeutic efficacy of PD-L1 checkpoint blockade [14]. Therefore, m6A modification status can modulate the efficiency of the antitumor immune response, the immune-therapeutic efficacy, and the overall survival (OS). Since epigenetic reprogramming is more likely manipulated by targeting epigenetic modifiers, epigenetic biomarkers may offer additional advantages [15]. However, until now, the relevance between m6A regulators and TME in UCEC remains unclear. Thus, an inquiry into m6A methylation may provide more possibilities for predicting immune therapeutic efficacy and the prognosis of UCEC.

With the rapid growth of high throughput sequencing technologies, there is an opportunity to obtain an intact picture of the mutation and transcription landscape of most tumors with even more clarity. Herein, we explored the landscape of m6A regulators in a UCEC cohort from The Cancer Genome Atlas (TCGA) database. We subgrouped UCEC patients according to their genetic alterations in m6A regulators because alteration in even one m6A regulator could lead to a dysfunction of m6A RNA modification, resulting in dysregulation of gene expression [16]. Then, we compared the molecular, clinical, and immunological characteristics between groups with distinctions of m6A regulators using a least absolute shrinkage and selection operator (LASSO) Cox regression analysis. A nine-gene prognostic mRNA signature was generated to predict the OS of UCEC patients. Finally, the function of each gene in the proposed gene signature was verified using in vitro and in vivo tumor cell experiments as well as the immune correlation analysis. The result shows that CDKN2A mainly acts from the perspective of tumor cells while the remaining genes mainly act by regulating the immune response. Therefore, the m6A RNA methylation regulator-based signature can provide a robust and stable prognostic tool and help probe the potential therapeutic target for UCEC treatment from the perspective of epigenetics.

Methods

Datasets for analysis

All paired clinical data and transcript profiles of UCEC samples were obtained and trimmed from the TCGA Data Portal by R package “GDCRNATools” and all the somatic mutations were available from the mutation annotation format (MAF) file on the TCGA website directly. A total of 530 UCEC samples were retained, and only patients with somatic mutation data and complete follow-up clinical information were screened for subsequent studies. Based on the R package “caret”, TCGA UCEC profiles were randomly separated into two parts: 75% was the training set, and 25% was the testing set. All methods were carried out in accordance with relevant guidelines.

Visualization of genetic alterations in m6A regulators

A total of 20 representative m6A regulators, including ten readers, eight writers, and two erasers, were selected from recently published studies. Before further analysis, all gene symbols of these m6A regulators were converted into HUGO Gene Nomenclature Committee (HGNC) symbols by manual curation from Ensembl (http://asia.ensembl.org/index.html). The R package “maftools” was used to summarize, analyze, and visualize the somatic mutations [17]. The summary of the MAF file was visualized as a waterfall plot, showing the number of variants in each sample. The variant allele frequency of gene mutations, shown as a boxplot, was defined as the reading of variants divided by the total reading.

Recognition of m6A-related differentially expressed genes (DEGs)

Alterations in m6A regulatory genes were screened in each UCEC sample as above. Subsequently, one cohort was separated into two subgroups: one group with m6A alterations and one without m6A alterations. The R package “Deseq2” was adopted to generate the DEGs, which depended upon a negative binomial distribution between the two groups. P < 0.05 and |log2 Fold-change| ≥ 1 were taken as significance criteria for DEGs. The R package “pheatmap” was adopted to conduct unsupervised learning clustering and plot the heatmap.

Immune cell composition estimation and gene ontology (GO) enrichment analysis

Tissue-infiltrating immune and other stromal subpopulation abundances based on mRNA expression were estimated by the MCP-counter method implemented by the R package “MCPcounter”. The immune cell composition estimation results were visualized using the R package “vioplot” in a violin plot. The online web server g:Profiler (https://biit.cs.ut.ee/gprofiler/gost) was used to perform an ordered GO enrichment analysis, where genes are in the order of decreasing importance [18]. Then, pathway enrichment was visualized and interpreted with the Cytoscape (V3.8.0) desktop application and the “EnrichmentMap Pipeline Collection” plug-in.

Modeling of m6A-related signatures for forecasting OS

The R package “glmnet” was used to conduct the LASSO Cox regression model via penalized maximum likelihood. The optimal cut-off point for distinguishing between high- and low-risk rating groups was determined using the R package “survminer” based on log-rank statistics. The m6A score formula was generated, and the risk score of each patient was calculated as follows:

$$\boldsymbol{Risk}\ \boldsymbol{score}=\sum_{\boldsymbol{i}=\textbf{1}}^{\boldsymbol{n}}{\boldsymbol{Coef}}_{\boldsymbol{i}}\times {\boldsymbol{Exp}}_{\boldsymbol{i}}$$

where Coefi means the coefficient and Expi is the expression of each m6A-related mRNA. Receiver operating characteristic (ROC) curves and area under the ROC curve (AUC) were used to quantify the sensitivity and specificity of the prognostic signature through the R package “timeROC”.

Tools for further analysis of the m6A signature

GETPIA 2 (http://gepia2.cancer-pku.cn/) [19], an enhanced web server for large-scale expression profiling and interactive analysis, was used to determine the relevance of the m6A signature gene expression and clinical outcome across various cancer types. CIBERSORT (https://cibersort.stanford.edu/) [20], an analytical tool to estimate the abundances of member cell types, was used to calculate the correlation between the indicated gene and tumor-infiltrating immune cells. RMBase v2.0 (https://rna.sysu.edu.cn/rmbase/) [21], a comprehensive database to integrate epi-transcriptome sequencing data for exploring post-transcriptionally modifications of RNAs, was used to decipher different RNA modifications of the signature, including m6A, N1-Methyladenosines (m1A), pseudouridine (Ψ) modifications, 5-methylcytosine (m5C) modifications, 2′-O-methylations (2′-O-Me). M6A2Target (http://m6a2target.canceromics.org/) [22], a comprehensive database for the target gene of WERs of m6A modification, was used to illustrate the correlation between WERs and the m6A signature.

Cell transfection to obtain knockdown cell lines

The lentivirus construction used to knockdown COL4A4, PXDN, CDKN2A, TIGIT, CHODL, LMO3, KCNJ12, L1CAM, and EPHB1was purchased from Genepharma (Shanghai, China). UCEC cell line Ishikawa was plated in 6-well microplates at 40–50% confluence and then infected with the above 9 lentiviruses (termed as shCOL4A4, shPXDN, shCDKN2A, shTIGIT, shCHODL, shLMO3, shKCNJ12, shL1CAM, and shEPHB1), or control (termed as shCtrl), respectively. Stable transduction pools were generated by puromycin selection for 2 weeks. The above cell transfection protocol was based on the manufacturer’s instructions.

Western blot assay

Western blot assay was performed as previously reported [23]. In brief, the proteins from Ishikawa cells were exacted with RIPA lysis buffer (Sigma-Aldrich, St Louis, MO). The concentration of proteins was measured with a BCA assay kit (Bio-Rad Laboratories, Hercules, CA, USA). The 10% SDS-PAGE isolated protein was transferred to a 0.22-μm nitrocellulose (NC) membrane (GE Healthcare, Piscataway, NJ, USA). The membranes were blocked at room temperature with 5% non-fat milk for 2 h. Then, the membranes were incubated in specific primary antibodies including COL4A4 (1:1000, MBS2032561, MyBioSource, San Diego, CA, USA), PXDN(1:1000, sc-293,408, Santa Cruz, CA, USA), CDKN2A(1:1000, ab270058, Abcam, Cambridge, MA, USA), TIGIT(1:1000, ab243903, Abcam, Cambridge, MA, USA), CHODL(1:1000, ab236742, Abcam, Cambridge, MA, USA), LMO3(1:1000, ab230490, Abcam, Cambridge, MA, USA), KCNJ12(1:1000, PA5–68685, Invitrogen, Carlsbad, CA, USA), L1CAM(1:1000, ab24345, Abcam, Cambridge, MA, USA), EPHB1(1:1000, ab129103, Abcam, Cambridge, MA, USA) and β-Actin (1:3000, A1978, Sigma, Victoria, BC, Canada) at 4 °C overnight. The membranes were washed with 0.1% TBST three times for 5 min, and then incubated with anti-mouse or anti-rabbit horseradish peroxidase-conjugated secondary antibody (Cell Signaling Technology, Danvers, MA, USA) for 2 h, and washed with 0.1% TBST three times for 5 min each. Chemiluminescent ECL Plus reagents (Pierce, USA) were added to visualize the reaction products. The membranes were scanned with Tanon 5200 (Tanon, Shanghai, PR China). The band intensity was measured by densitometry using the Quantity One Software (Tanon, Shanghai, PR China). The protein levels were normalized with that of β-actin. All experiments were repeated in triplicate, and the representative results were shown.

MTT, Colony formation, Transwell, and wound healing assays

MTT, Colony formation and Transwell assays were performed as described previously [23]. Cell migration was also performed with a wound healing assay. Ishikawa cells transfected with the shCOL4A4, shPXDN, shCDKN2A, shTIGIT, shCHODL, shLMO3, shKCNJ12, shL1CAM, shEPHB1 and shCtrl were seeded in 6-well microplates, and scratches were generated using micropipette tips when 90% confluence was reached. Cells were washed 3 times using sterile PBS to wash off non-adherent cells generated by the scratch, and a fresh serum-free medium was replaced to continue culturing the cells. The wound status was observed at 0 h and 72 h after scratching with an X71 inverted microscope. The means of intercellular distances were calculated using the ImageJ software. All experiments were duplicated thrice.

Xenografted tumor model

Four to 6 weeks old (average weight: 15 g) BALB/c nude mice (male/female ratio 1:1) were obtained from the Shanghai Institute of Materia Medica, Chinese Academy of Science, and maintained under specific pathogen-free conditions. No statistical method was applied for the sample size estimation for the animal study. Each experimental group had enrolled 4 nude mice in an unrandomized manner to ensure the precision of the results. The experimental protocol was approved by the Wannan Medical College Animal Experimental Ethics Committee and reporting of these experiments complied with the ARRIVE (Animal Research: Reporting of In Vivo Experiments) guidelines. The cells (2 × 106 Ishikawa -shCDKN2A cells and Ishikawa-shCtrl cells) were injected subcutaneously into the right dorsal flank. The tumor sizes were measured using a Vernier caliper every day when the tumors were readily visualized. Tumor formation in nude mice was observed by measuring the tumor volume calculated by the following formula: volume = (length × width^2)/2. On day 35, animals were euthanized, and tumors were excised and weighed. The exclusion criteria of animal experiments are that the bodyweight of the mouse was statistically significantly changed compared to the others. The xenograft tumor was festered seriously and influenced the measurement of tumor volume.

Statistical analysis

Correlation analysis was conducted using the Pearson correlation method. The chi-square test was applied to compare the clinical-pathological features among different groups. The Mann-Whitney U test was adopted to compare the difference between genetic patterns. All statistical analyses were performed using R software (version 3.6.3) in the RStudio program (version 1.3.1073).

Results

Landscape of genetic alterations in m6A regulators in TCGA UCEC patients

Among 530 TCGA UCEC cases that were analyzed, 153 samples from all cases had a genetic alteration in the m6A regulator with a frequency of 28.9%, and missense mutations had a leading role. The frequency of genetic alteration in the m6A writer gene ZC3H13 (11.9%) was the highest, followed by YTHDC2 (8.7%), YTHDC1 (6.6%), IGF2BP1 (6.0%), and RBM15 (5.3%) (Fig. 1A; Table 1). Figure 1B shows the variant allele frequency of gene mutations, which refers to the ratio between the number of mutants and wild-type DNA copies, which estimates the clonal status of mutated genes (Fig. 1B). We searched for alterations in m6A regulatory genes in each TCGA UCEC sample. Then, TCGA cohorts were separated into two subgroups: one group with m6A regulator mutations and one group without m6A regulator mutations. Considering that an alteration in only one m6A regulator can lead to a dysfunction of m6A RNA modification, these two groups can represent two scenarios (a normal status and an abnormal one) of the m6A level.

Fig. 1
figure 1

Genetic alterations in m6A regulators in uterine corpus endometrial carcinoma (UCEC). A The waterfall plot shows the number of altered m6A regulators in each UCEC sample. B The boxplot shows the variant allele frequency of gene mutations, defined as the reading of variant reads separated by the total reading

Table 1 Different genetic alteration patterns of m6A regulators in UCEC samples (n = 530)

Relationship among genetic alterations, m6A gene expression, and clinical phenotypes

We explored the relevance of genetic alterations, m6A gene expression, and the clinical phenotypes of UCEC patients. The results demonstrated that alterations in m6A regulators were only significantly related to age, not clinical stage and race. In Table 2, values in bold with an asterisk indicate a statistically significant difference (P < 0.05) using the chi-square test and demonstrate that genetic alterations in m6A regulators are more likely to occur in UCEC patients younger than 60 years of age. Supplementary Fig. 1 shows the decreasing trend of mRNA levels of ALKBH5, FTO, METTL16, and RBM15 by the Mann-Whitney U test. It seems that genetic alterations, especially single-nucleotide substitutions by missense mutations, may not impair most mRNA expression. However, a single nucleotide alteration in the DNA sequence of a gene can produce the wrong amino acid, thus affecting the three-dimensional structure and biological function of an m6A regulator protein.

Table 2 Clinical phenotypes between UCEC patients with or without mutation of m6A regulators

GO analysis for differentially expressed mRNAs

A DEG analysis was conducted to recognize distinct patterns between subgroups with these two different m6A profiles. A total of 124 mRNAs passed the threshold screening. As shown in the heatmap (Fig. 2A), 23 up-regulated genes and 101 down-regulated genes are ranked by Pearson distance, indicating that m6A regulator-related genetic alterations can decrease most gene expression. GO analyses were conducted on these 124 genes. The results show that these differentially expressed mRNAs are mainly enriched in biological processes associated with T cell activation. A word cloud analysis shows that cell, regulation, and proliferation are the most frequent words enriched in GO terms (Fig. 2B). Therefore, m6A modification can affect the occurrence and development of tumors.

Fig. 2
figure 2

The m6A-related differences between the group with genetic alterations in the m6A regulator and the group without genetic alterations in the m6A regulator. A The heatmap shows that 124 mRNAs are differentially expressed between the two groups. B Gene Ontology (GO) enrichment of the differentially expressed mRNAs

Relationship between genetic alterations and immunological characteristics

Considering the potential roles of T cells above, the tissue-infiltrating immune and other stromal subpopulations abundances in UCEC groups with distinct m6A regulator traits were estimated to further explore the immunological role of m6A modification. The results are shown in the violin plot in Fig. 3, suggesting that cytotoxic lymphocytes and natural killer (NK) cells both can kill target cells and exhibit higher proportions in the UCEC group with alterations in the m6A regulator (Fig. 3). The results are consistent with the GO enrichment results above. In contrast, endothelial cells have the opposite role. There were no significant inter-group differences for other types of immune cells. These results show that distinct levels of m6A can lead to differential immune cell composition, thereby influencing the immune responses.

Fig. 3
figure 3

Immunological characteristics between UCEC patients with or without mutations in m6A regulators. Significant results are marked in red

Generation of m6A gene signature from the training set

A total of 71 prognostic genes were generated by manipulating a univariate Cox model, revealing an association between the expression level of mRNAs and the OS of patients (P < 0.05). To reduce dimensionality and select representative m6A-related markers for forecasting the OS of UCEC patients, a LASSO Cox regression model was performed according to the minimum criterion in the training set. Nine m6A-related genes, CDKN2A, TIGIT, COL4A4, PXDN, CHODL, LMO3, KCNJ12, L1CAM, and EPHB1were selected and further weighted by their regression coefficients (Figs. 4A-B). The forest plot in Fig. 4C shows that TIGIT is an independent protective factor with a hazard ratio (HR) < 1 while CHODL is an independent risk factor with HR > 1 for UCEC patients (Fig. 4C). The results suggest that patients with a high expression of CHODL have a poor survival while those with a high expression of TIGIT have a good survival. The whole Cox model also exhibited statistical significance with a concordance index of 0.75.

Fig. 4
figure 4

The mRNA signature associated with the m6A trait. A The profile of the LASSO coefficients. B The partial likelihood of deviance is shown against log (Lambda). At the value fitting 10-fold cross-validation, a vertical line was drawn. C Forest plots of the prognostic ability of the nine m6A-related mRNAs are included in the prognostic signature. Time-dependent ROC curves and Kaplan-Meier survival analysis between UCEC patients with high- and low-risk depend upon our mRNA signature. (D, E) Training cohort. (G, H) Testing cohort. We used AUCs at 1, 3, 5, and 7 years to assess prognostic accuracy and calculated P values using the log-rank test. F Gene Ontology (GO) enrichment of the m6A signature

In addition, time-dependent ROC curve analyses in the training set were performed at 1, 3, 5, and 7 years to evaluate the accuracy of prognostic prediction of the m6A-related mRNA signature. Figure 4D shows that the maximum AUC is 0.79 at 3249 days, indicating that m6A-related genes can serve as biomarkers in the prognosis of UCEC (Fig. 4D). Subsequently, the risk score for each UCEC patient was calculated with the formula generated by the coefficients. The UCEC patients in the training set were separated into high- and low-risk subgroups using the median risk score as the cut-off value. Consistent with ROC curve analysis, the Kaplan-Meier curve shows that the UCEC patient risk score is inversely proportional to the clinical outcome (Fig. 4E). Figure 4F shows that the gene signature is enriched in the pathways: collagen type IV sulfilimine cross-linking by peroxidasin, axon guidance receptor activity, cell adhesion molecules, positive regulation of cell morphogenesis involved in differentiation, and negative regulation of lymphocyte activation (Fig. 4F).

Validation of m6A gene signature for OS prediction in the testing set

The risk score for each sample in the testing cohort was calculated using the same formula in training set to verify the prognostic ability of the m6A gene signature. The UCEC patients in the testing dataset were separated into high- and low-risk groups based on the median risk score. The results were consistent with those in the training dataset. As shown in Fig. 4G, the max AUC is 0.74 at 1314 days (Fig. 4G). Figure 4H shows that patients with higher risk scores have shorter survival and increased death status (P < 0.0001), suggesting that the m6A gene signature has a robust and stable OS predictive ability (Fig. 4H). Furthermore, Fig. 5 shows that the expression pattern of the m6A gene signature is tumor-specific and most closely related to clinical outcomes in UCEC (Fig. 5). The increased risk is marked in red, and the decreased risk is marked in blue. Except for TIGIT with a decreased risk, the m6A signature has an increased risk for survival. The result is consistent with the finding that abnormal m6A levels can lead to tumor genesis [9, 11].

Fig. 5
figure 5

Heatmap shows the relevance of the m6A signature gene expression and clinical outcome across various cancer types using the Cox proportional hazard model. The increased risk is marked in red, and the decreased risk is marked in blue

Further analysis of the proposed m6A gene signature

Figure 6 is the alluvial plot to illustrate the correlation between WERs of m6A modification and the m6A signature (Fig. 6). Yellow wire represents high confidential targets validated by low-throughput experiments; purple wire represents binding evidence indicated by high-throughput sequencings such as CLIP-Seq, RIP-seq, and ChIP-seq; green wire represents m6A WER perturbation followed by high-throughput sequencings such as RNA-Seq, m6A-Seq, and Ribo-Seq [22]. The results show that the proposed m6A gene signature closely relates to m6A regulators. Therefore, reprogramming m6A levels by restoring dysfunction of the m6A regulators can directly target the proposed m6A gene signature and return to normal expression levels. We explored different RNA modifications other than m6A methylation, including N1-methyladenosines (m1A), pseudouridine (Ψ) modifications, 5-methylcytosine (m5C) modifications, and 2′-O-methylations (2′-O-Me) (Table 3) to better understand the proposed m6A signature. The results show that m6A methylation is the main type of modification site in the proposed signature.

Fig. 6
figure 6

The alluvial plot illustrates the correlation between writers, erasers, and readers (WERs) of m6A modification and the m6A signature

Table 3 RNA Modifications of the Proposed Signature from RMBase v2.0

In vitro and in vivo experiments to validate the proposed m6A gene signature

Then, some in vitro experiments were performed to verify the above assumption. The Ishikawa cell line is by far the most commonly used xenograft model of UCEC. To elucidate the biological functions of CDKN2A, TIGIT, COL4A4, PXDN, CHODL, LMO3, KCNJ12, L1CAM, and EPHB1 in UCEC tumor cells, we knocked down the expression of the 9-gene signature using shRNA or the negative control in Ishikawa cell lines to assess cell proliferation, migration, and invasion in vitro (Fig. 7A). The MTT assay and the colony formation assay demonstrated that tumor proliferation was significantly inhibited in the shCDKN2A group compared with the shCtrl group (P < 0.05, Figs. 7B-C). Transwell assays revealed that Ishikawa cell invasion was significantly reduced after downregulation of CDKN2A (Fig. 7D). Cell migration was evaluated by wound-healing assay. The result shows that decreased expression of CDKN2A significantly inhibits the migration of Ishikawa cells (Fig. 7E). The above results indicate that knockdown of CDKN2A can inhibit the proliferation, migration, and invasion of UCEC tumor cells in vitro. The mouse xenograft models were established to determine whether CDKN2A contributes to tumor progression in vivo. The shRNA or CDKN2A shRNA Ishikawa cells were injected at flank into nude mice. After 5 weeks of injection, the tumor volume and weight of the CDKN2A knockdown group were significantly reduced compared to those of the control group (Figs. 7F-H). These findings indicated that CDKN2A functions from the perspective of tumor cells since it is significantly up-regulated in metastatic UCEC, and its knockdown attenuates the ability of tumor cells to invade metastases in vitro and in vivo.

Fig. 7
figure 7

Silencing each gene of the m6A signature expression in UCEC inhibits cell proliferation, migration, and invasion in vitro and in vivo. A Western blot analysis of each gene (COL4A4, PXDN, CDKN2A, TIGIT, CHODL, LMO3, KCNJ12, L1CAM, and EPHB1) expression in Ishikawa cells expressing each gene shRNA (shCOL4A4, shPXDN, shCDKN2A, shTIGIT, shCHODL, shLMO3, shKCNJ12, shL1CAM, and shEPHB1). β-actin is adopted as a loading control. B MTT assays were used to investigate the proliferation rates of the COL4A4, PXDN, CDKN2A, TIGIT, CHODL, LMO3, KCNJ12, L1CAM, EPHB1-silenced Ishikawa cells. C Colony formation assay was used to investigate the proliferation capacity of the COL4A4, PXDN, CDKN2A, TIGIT, CHODL, LMO3, KCNJ12, L1CAM, EPHB1-silenced Ishikawa cells. Representative pictures are shown on the left, and the number of colonies has been counted on the right. D Transwell assay was used to investigate the metastasis capacity of the COL4A4, PXDN, CDKN2A, TIGIT, CHODL, LMO3, KCNJ12, L1CAM, EPHB1-silenced Ishikawa cells. Representative pictures are shown on the left, and the number of metastasis cells has been counted on the right. E Wound healing assay was used to investigate the migration capacity of the COL4A4, PXDN, CDKN2A, TIGIT, CHODL, LMO3, KCNJ12, L1CAM, EPHB1-silenced Ishikawa cells. Representative pictures are shown on the left, and the wound width has been measured on the right. F Macroscopic view of tumor harvested of two indicates treatment groups (n = 4). G Comparison of the tumor weight of two indicated treatment groups at the end-point (n = 4). H Comparison of the growth curve of the tumors of two indicates treatment groups at the each-point (n = 4). Bar graph data are presented as mean ± SEM; *, P < 0.05

Potential immunological role of the m6A gene signature

Since only CDKN2A among all 9 genes was validated to be associated with tumor cells in in vitro and in vivo assays, we speculated that the rest of the genes function from the perspective of immune cells. The correlation between various types of immune cell signatures and each gene of the m6A gene signature was explored (Fig. 8). Using the CIBERSORT algorithm, we found that except for CDKN2A, other genes had a certain degree of correlation with immune cells. TIGIT was the gene most strongly associated with immune function and was associated with CD8+ T cells, CD4+ T cells, NK cells, and Macrophage M1 cells with P < 0.001. In contrast, its trend was the opposite for other genes. The result is consistent with the previous results: TIGIT is an independent protective factor with a hazard ratio (HR) < 1 while others are risk factors. These results indicate that CDKN2A may function from the perspective of tumor cells, and CDKN2A, TIGIT, COL4A4, PXDN, CHODL, LMO3, KCNJ12, L1CAM, and EPHB1 may play a role in UCEC from an immunological point of view.

Fig. 8
figure 8

Correlation between each gene of the m6A gene signature and the signatures of various types of immune cells

Discussion

UCEC is the most common female pelvic malignancy worldwide. The m6A modification, a dynamic and reversible process involving m6A regulators, can directly regulate the expression of oncogenes or tumor suppressor genes at the transcriptional level [8], which is closely associated with malignancies. A better understanding of the potential role of m6A modification during the tumorigenesis can provide potential biomarkers for early diagnosis and prognosis assessment and new ideas for therapeutic targets [24, 25]. Abnormal expression patterns of m6A regulators and a reduction in m6A methylation have already been found in endometrial cancer [11]. However, the genetic variations, tumor-infiltrating immunological characteristics, and clinical implications involving m6A regulators in UCEC remain unknown.

In the present study, we examined the landscape of genetic alterations in the m6A regulators of TCGA UCEC patients and found that 28.9% of m6A regulators were mutated, suggesting a close relationship between m6A methylation modification and endometrial cancer. Among them, the highest frequency of genetic alteration was found in the m6A writer gene ZC3H13 (11.9%). The result shows that the function of ZC3H13 is suppressed in UCEC patients, and this dysfunction may be caused by the occurrence of UCEC. ZC3H13 has also been reported to act as a tumor suppressor in breast and colorectal cancer [26, 27]. Then, we explored the relevance of genetic alterations, m6A gene expression, and the clinical phenotypes of patients with UCEC. Most indicators did not differ significantly between two distinct traits of m6A regulator alteration, except that UCEC patients younger than 60 are more likely to have genetic alterations of m6A regulators. The result implies that although older patients are more likely to be informed of the risks and symptoms of endometrial cancer, m6A modifications may play a role in the occurrence of endometrial cancer in younger patients [28]. Alteration in only one m6A regulator can lead to the dysfunction of m6A RNA modification. Therefore, we generated an mRNA signature from the outcome-related DEGs by univariate Cox regression followed by LASSO Cox regression after the separation of UCEC patients into two subgroups depending upon the distinction of the m6A regulator. Subsequently, a nine m6A-related prognostic signature, CDKN2A, TIGIT, COL4A4, PXDN, CHODL, LMO3, KCNJ12, L1CAM, and EPHB1, was developed and performed well for predicting the survival outcome of UCEC patients.

To better understand the proposed m6A signature, we explored different RNA modifications other than m6A methylation and illustrated the correlation between m6A regulators and the proposed signature. The results show that the proposed m6A gene signature did have a close relationship with m6A regulators. Furthermore, m6A methylation is the main modification site in the proposed signature since the direct effects of m6A modification on modified transcript expression are established, and the epigenetic reprogramming is more likely manipulated by targeting epigenetic modifiers [29]. Therefore, this m6A signature can provide new ideas for diagnosing and treating UCEC from the perspective of epigenetics.

Since RNA-seq data in TCGA is performed using bulk RNA extracted from homogenized tissues, which provide an average number of gene expressions in the pooled population of diverse cells [30], we cannot determine whether the proposed gene signature works in tumor cells or tumor-infiltrating immune cells. Therefore, we used in vitro and in vivo tumor cell experiments and immune correlation analysis to verify the function of each gene in the proposed gene signature. The results show that CDKN2A mainly acts from the perspective of tumor cells, while COL4A4, PXDN, TIGIT, CHODL, LMO3, KCNJ12, L1CAM, and EPHB1 may play a role in UCEC from an immunological point of view. Numerous studies have demonstrated that m6A regulators correlate with malignant tumor progression and immunomodulatory abnormality, revealing the special correlation between TME infiltrating immune cells and m6A modification [12]. A recent study also shows that abnormal modification behavior of m6A can slow down the kinetic rates of mRNA during T cell differentiation due to the increase in naïve T cell numbers from lymph nodes [31]. Therefore, our proposed prognostic m6A gene signature may predict immune therapeutic efficacy. Since the present study focused on mining the m6A associated gene signature and defined their roles from tumor and immune perspective separately without the mechanistic studies that shows the downstream targets of each gene. We will use our constructed knockdown cell lines for RNA-seq to obtain gene expression profiles of KD cell lines relative to control cell lines that have been subjected to subsequent mechanistic studies.

Conclusions

The abnormal m6A modifications are closely associated with the clinical outcome of patients with UCEC. The m6A RNA methylation regulator-based prognostic signature can predict the prognosis of UCEC patients and immune therapeutic efficacy from the perspective of epigenetics. Our results will provide a direction for the further exploration of the pathogenesis of UCEC.

Availability of data and materials

Publicly available datasets were analyzed in this study. These datasets can be found here: https://portal.gdc.cancer.gov/.

Abbreviations

UCEC :

Uterine corpus endometrial carcinoma

m 6 A :

N6-methyladenosine

TCGA :

The Cancer Genome Atlas

OS :

Overall survival

LASSO :

Least absolute shrinkage and selection operator

ROC :

Receiver operating characteristic

m 1 A :

N1-methyladenosine

m 5 C :

5-methylcytosine

WERs :

Writers, erasers, and readers

TME :

Tumor microenvironment

APC :

Antigen-presenting cells

MAF :

Mutation annotation format

HGNC :

HUGO Gene Nomenclature Committee

DEGs :

Differentially expressed genes

GO :

Gene ontology

AUC :

Area under the ROC curve

Ψ :

Pseudouridine

2′-O-Me :

2′-O-methylations

References

  1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33.

    Article  Google Scholar 

  2. Casey MJ, Summers GK, Crotzer D. Endometrial Cancer. Treasure Island (FL): StatPearls; 2021.

    Google Scholar 

  3. Liu S, Wu B, Li X, Zhao L, Wu W, Ai S. Construction and validation of a potent epigenetic modification-related prognostic signature for osteosarcoma patients. J Oncol. 2021;2021:2719172.

    Article  Google Scholar 

  4. Zheng S, Han H, Lin S. N (6)-methyladenosine (m (6) a) RNA modification in tumor immunity. Cancer Biol Med. 2022;19(4):385–97.

  5. Liu ZX, Li LM, Sun HL, Liu SM. Link between m6A modification and cancers. Front Bioeng Biotechnol. 2018;6:89.

    Article  Google Scholar 

  6. Meyer KD, Patil DP, Zhou J, Zinoviev A, Skabkin MA, Elemento O, et al. 5′ UTR m (6) a promotes cap-independent translation. Cell. 2015;163(4):999–1010.

    Article  Google Scholar 

  7. Zhao BS, Roundtree IA, He C. Post-transcriptional gene regulation by mRNA modifications. Nat Rev Mol Cell Biol. 2017;18(1):31–42.

    Article  Google Scholar 

  8. Roignant JY, Soller M. M (6) a in mRNA: an ancient mechanism for fine-tuning gene expression. Trends Genet. 2017;33(6):380–90.

    Article  Google Scholar 

  9. Chen M, Wong CM. The emerging roles of N6-methyladenosine (m6A) deregulation in liver carcinogenesis. Mol Cancer. 2020;19(1):44.

    Article  Google Scholar 

  10. Li Z, Li Y, Zhong W, Huang P. m6A-related lncRNA to develop prognostic signature and predict the immune landscape in bladder Cancer. J Oncol. 2021;2021:7488188.

    Google Scholar 

  11. Liu J, Eckert MA, Harada BT, Liu SM, Lu Z, Yu K, et al. M (6) a mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer. Nat Cell Biol. 2018;20(9):1074–83.

    Article  Google Scholar 

  12. Shulman Z, Stern-Ginossar N. The RNA modification N (6)-methyladenosine as a novel regulator of the immune system. Nat Immunol. 2020;21(5):501–12.

    Article  Google Scholar 

  13. Wang H, Hu X, Huang M, Liu J, Gu Y, Ma L, et al. Mettl3-mediated mRNA m (6) a methylation promotes dendritic cell activation. Nat Commun. 2019;10(1):1898.

    Article  Google Scholar 

  14. Han D, Liu J, Chen C, Dong L, Liu Y, Chang R, et al. Author correction: anti-tumour immunity controlled through mRNA m (6) a methylation and YTHDF1 in dendritic cells. Nature. 2019;568(7751):E3.

    Article  Google Scholar 

  15. Kumar S, Nagpal R, Kumar A, Ashraf MU, Bae YS. Immunotherapeutic potential of m6A-modifiers and MicroRNAs in controlling acute myeloid Leukaemia. Biomedicines. 2021;9(6):690.

  16. Zeng J, Zhang H, Tan Y, Wang Z, Li Y, Yang X. Genetic alterations and functional networks of m6A RNA methylation regulators in pancreatic cancer based on data mining. J Transl Med. 2021;19(1):323.

    Article  Google Scholar 

  17. Mayakonda A, Lin DC, Assenov Y, Plass C, Koeffler HP. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 2018;28(11):1747–56.

    Article  Google Scholar 

  18. Raudvere U, Kolberg L, Kuzmin I, Arak T, Adler P, Peterson H, et al. G:profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 2019;47(W1):W191–W8.

    Article  Google Scholar 

  19. Tang Z, Kang B, Li C, Chen T, Zhang Z. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 2019;47(W1):W556–W60.

    Article  Google Scholar 

  20. Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. Profiling tumor infiltrating immune cells with CIBERSORT. Methods Mol Biol. 2018;1711:243–59.

    Article  Google Scholar 

  21. Xuan JJ, Sun WJ, Lin PH, Zhou KR, Liu S, Zheng LL, et al. RMBase v2.0: deciphering the map of RNA modifications from epitranscriptome sequencing data. Nucleic Acids Res. 2018;46(D1):D327–D34.

    Article  Google Scholar 

  22. Deng S, Zhang H, Zhu K, Li X, Ye Y, Li R, et al. M6A2Target: a comprehensive database for targets of m6A writers, erasers and readers. Brief Bioinform. 2021;22(3):bbaa055.

  23. Liu X, Zhang M, Zhu X, Wang Y, Lv K, Yang H. Loss of FAM60A attenuates cell proliferation in glioma via suppression of PI3K/Akt/mTOR signaling pathways. Transl Oncol. 2021;14(11):101196.

    Article  Google Scholar 

  24. Li Y, Gu J, Xu F, Zhu Q, Chen Y, Ge D, et al. Molecular characterization, biological function, tumor microenvironment association and clinical significance of m6A regulators in lung adenocarcinoma. Brief Bioinform. 2021;22(4):bbaa225.

  25. Li Z, Li F, Peng Y, Fang J, Zhou J. Identification of three m6A-related mRNAs signature and risk score for the prognostication of hepatocellular carcinoma. Cancer Med. 2020;9(5):1877–89.

    Article  Google Scholar 

  26. Gong PJ, Shao YC, Yang Y, Song WJ, He X, Zeng YF, et al. Analysis of N6-Methyladenosine methyltransferase reveals METTL14 and ZC3H13 as tumor suppressor genes in breast Cancer. Front Oncol. 2020;10:578963.

    Article  Google Scholar 

  27. Zhu D, Zhou J, Zhao J, Jiang G, Zhang X, Zhang Y, et al. ZC3H13 suppresses colorectal cancer proliferation and invasion via inactivating Ras-ERK signaling. J Cell Physiol. 2019;234(6):8899–907.

    Article  Google Scholar 

  28. Braun MM, Overbeek-Wager EA, Grumbo RJ. Diagnosis and Management of Endometrial Cancer. Am Fam Physician. 2016;93(6):468–74.

    Google Scholar 

  29. Roundtree IA, Evans ME, Pan T, He C. Dynamic RNA modifications in gene expression regulation. Cell. 2017;169(7):1187–200.

    Article  Google Scholar 

  30. Ding S, Chen X, Shen K. Single-cell RNA sequencing in breast cancer: understanding tumor heterogeneity and paving roads to individualized therapy. Cancer Commun (Lond). 2020;40(8):329–44.

    Article  Google Scholar 

  31. Furlan M, Galeota E, de Pretis S, Caselle M, Pelizzola M. m6A-dependent RNA dynamics in T cell differentiation. Genes (Basel). 2019;10(1):28.

Download references

Acknowledgments

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (81870017), the Key University Science Research Project of Anhui Province (NO. KJ2021A0848), and the Key Research and Development Plan of Anhui Province, China (No. 2022a05020011).

Author information

Authors and Affiliations

Authors

Contributions

Qingqing Li and Xiaocen Liu designed the study. Fang Fang, Peipei Wang and Haoyu Huang performed the bioinformatics experiments. Qingqing Li and Fang Fang analyzed the results and drafted the manuscript. Xiaocen Liu and Peipei Wang discussed, contributed to the data analysis. Haoyu Huang designed the bioinformatics experiments. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Xiaocen Liu or Qingqing Li.

Ethics declarations

Ethics approval and consent to participate

The experimental protocol was approved by the Wannan Medical College Animal Experimental Ethics Committee and reporting of these experiments complied with the ARRIVE (Animal Research: Reporting of In Vivo Experiments) guidelines. The cells (2 × 106 Ishikawa -shCDKN2A cells and Ishikawa-shCtrl cells) were injected subcutaneously into the right dorsal flank.

Consent for publication

Not applicable.

Competing interests

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Supplementary Fig. 1

m6A gene expression between UCEC patients with or without mutations in m6A regulators. *, P < 0.05; **, P < 0.01.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fang, F., Wang, P., Huang, H. et al. m6A RNA methylation regulator-based signature for prognostic prediction and its potential immunological role in uterine corpus endometrial carcinoma. BMC Cancer 22, 1364 (2022). https://doi.org/10.1186/s12885-022-10490-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-022-10490-x

Keywords