Skip to main content

ORIGINAL RESEARCH article

Front. Genet., 25 August 2022
Sec. Computational Genomics

m5C Regulator-mediated methylation modification clusters contribute to the immune microenvironment regulation of multiple myeloma

Hefei Ren&#x;Hefei Ren1Chang Liu&#x;Chang Liu1Hongkun WuHongkun Wu1Zhenhua WangZhenhua Wang1Sai ChenSai Chen1Xiaomin ZhangXiaomin Zhang1Jigang RenJigang Ren1Huiying Qiu
Huiying Qiu2*Lin Zhou
Lin Zhou1*
  • 1Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
  • 2Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China

Background: Multiple myeloma (MM) is a hematological malignancy in which plasma cells proliferate abnormally. 5-methylcytosine (m5C) methylation modification is the primary epigenetic modification and is involved in regulating the occurrence, development, invasion, and metastasis of various tumors; however, its immunological functions have not been systematically described in MM. Thus, this study aimed to clarify the significance of m5C modifications and how the immune microenvironment is linked to m5C methylation in MM.

Method: A total of 483 samples (60 healthy samples, 423 MM samples) from the Gene Expression Omnibus dataset were acquired to assess the expression of m5C regulators. A nomogram model was established to predict the occurrence of MM. We investigated the impact of m5C modification on immune microenvironment characteristics, such as the infiltration of immunocytes and immune response reactions. We then systematically evaluated three different m5C expression patterns to assess immune characteristics and metabolic functional pathways and established m5C-related differentially expressed genes (DEGs). In addition, biological process analysis was performed and an m5C score was constructed to identify potentially significant immunological functions in MM.

Result: Differential expressions of m5C regulators were identified between healthy and MM samples. The nomogram revealed that m5C regulators could predict higher disease occurrence of MM. We identified three distinct m5C clusters with unique immunological and metabolic characteristics. Among the three different m5C clusters, cluster C had more immune characteristics and more metabolism-related pathways than clusters A and B. We analyzed 256 m5C-related DEGs and classified the samples into three different m5C gene clusters. Based on the m5C and m5C gene clusters, we calculated m5C scores and classified each patient into high- and low-m5C score groups.

Conclusion: Our study demonstrated that m5C modification is involved in and contributes to the diversity and complexity of the immune microenvironment, which offers promise for the development of accurate therapeutic strategies.

1 Introduction

Multiple myeloma (MM) is a malignant proliferative hematological disease that originates from the hematopoietic system and is caused by the abnormal proliferation of plasma cells (Cowan et al., 2022). The disease can affect any organ of the human body, with the most frequently involved organs being the bone and bone marrow. MM accounts for 10% of the total number of patients with hematological tumors and is the second most common type of hematological malignancy globally (Allegra et al., 2022). Monoclonal immunoglobulins appear in the blood and urine of MM patients, which results in functional damage to related target organs, such as bone marrow hematopoiesis, kidneys, and bone. MM manifests clinically with anemia, bone pain and osteolytic destruction, renal failure, and repeated infections (Wang et al., 2021). Because MM tumor cells are derived from immune cells, they can affect the humoral and cellular immunity of MM patients (Jadoon and Siddiqui, 2021). In the early monoclonal gammopathy of undetermined significance (MGUS) stage of the disease, an average of 1.0–1.5% of MGUS patients per year progress to active myeloma, which indicates that the interaction between the immune system and tumor cells determines the final evolution of MM (Nakamura et al., 2020). Abnormal plasma cells in MGUS can affect various biological processes in the bone marrow microenvironment, such as cell homing, cytokine secretion, immune cell function, and angiogenesis. The homeostasis of the tumor immune microenvironment can also further promote the malignant proliferation of plasma cells. Therefore, a detailed understanding concerning the biological characteristics and immunoregulatory mechanisms of MM cells will be crucial for the discovery of new diagnostic biomarkers and possible drug targets as well as the exploration of new immunotherapy methods for MM.

As a groundbreaking and emerging area of epigenetic research, 5-methylcytosine (m5C) epigenetic modification has received increased attention in recent years. m5C methylation modification is a methylation of the fifth carbon atom on the DNA cytosine (C), in the context of cytosine-guanine dinucleotides (CpG) through the action of the DNMTs (Bestor, 1988). m5C modification occurs frequently in cells and plays an essential role in maintaining gene expression and genomic stability in cells. m5C is modified primarily by methyltransferases (“writers”), demethylases (“erasers”), and binding proteins (“readers”). A range of biological functions are exerted on determining cell differentiation state, selecting cell identity, mitochondrial activity, and immunoregulatory mechanisms. m5C methylation modification is a dynamic and reversible process; m5C “writers” catalyze the formation of m5C, “readers” function by recognizing and binding to m5C methylation sites, and “erasers” catalyze the demethylation of m5C (Hu et al., 2021). Several studies have suggested that m5C regulation contributes to underlying immune-regulating mechanisms (Wang et al., 2017; Guo et al., 2020). The abnormal expression of m5C regulators may represent a good candidate for modifying m5C in systemic lupus erythematosus CD4+ T cells. Moreover, in rat T lymphocytes, m5C writers promote the translation of interleukin 17A by methylating the C466 site.

Despite the growing evidence that m5C regulation plays a regulatory role in immune responses, no studies have been conducted to demonstrate the role m5C regulation plays in the pathogenesis of MM. Such in-depth studies of m5C regulators and immune characteristics between healthy and MM samples, as well as the possible influence of different m5C patterns, have important implications for understanding the immunoregulatory mechanisms of MM. Thus, in this study, m5C regulator expression analysis was systematically conducted in 60 healthy samples and 423 MM samples to examine correlations with immune characteristics. We identified m5C modification clusters and developed an m5C scoring system to predict and guide immunotherapy.

2 Materials and methods

2.1 Dataset collection and preparation

All gene expression profiles for samples were obtained from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo). Normative transformations and log2 transformations were applied to preprocess the raw data. A total of 483 samples (60 healthy samples and 423 MM samples) were obtained in the analysis, which included those from the GSE5900 (N = 22), GSE6477 (N = 81), GSE13591 (N = 138), GSE39754 (N = 176), GSE47552 (N = 46), and GSE80608 (N = 20). The array data of GSE5900 were obtained using the GPL570 platform (HG-U133_Plus_2 Affymetrix Human Genome U133 Plus 2.0 Array). GSE6477 and GSE13591 were obtained using the GPL96 platform (HG-U133A Affymetrix Human Genome U133A Array). GSE39754 was obtained using the GPL5175 platform (HuEx-1_0-st Affymetrix Human Exon 1.0 ST Array transcript (gene) version). GSE47552 was obtained using the GPL6244 platform (HuGene-1_0-st Affymetrix Human Gene 1.0 ST Array transcript [gene] version). Multiple probes were assigned to the same gene symbol, and median gene expression was calculated as the average expression level. “ComBat Batch correction” was applied to preprocess the value of the expression using the R package “sva” to remove batch effects (Leek et al., 2012).

2.2 m5C methylation regulator detection

The list of m5C regulators we used was derived from previously published data, and a total of 16 acknowledged m5C regulator genes were curated and analyzed to identify distinct m5C methylation modification patterns (Huang et al., 2021; Lv et al., 2021; Pan et al., 2021; Yu et al., 2021). The 16 m5C regulators included five writers (DNMT3A, DNMT3B, DNMT1, TRDMT1, and NSUN5), nine readers (MBD1, MBD2, MBD3, MBD4, MECP2, NTHL1, SMUG1, UNG, and YBX1), and two erasers (TET3 and TDG). The STRING (www.string-db.org) online database was used to build the protein–protein interaction (PPI) network among the m5C regulatory genes. Pearson’s correlation analysis was performed to examine the association among different m5C-related genes. Significant correlations were signified by a correlation coefficient r-value between −1 and +1. Visualization of the correlation matrix was performed using the “corrplot” package. We then performed expression analysis of m5C regulators between healthy and MM samples using the package “limma.” The boxplot and heatmap were generated using the “ggpubr” package and the “pheatmap” package, respectively.

2.3 Generation of the random forest algorithm for the m5C regulator and establishment of the nomogram

To establish an optimal training model that can predict the occurrence of MM, we constructed support vector machine (SVM) and random forest (RF) algorithm models using the “kernlab” and “DALEX” packages. An evaluation of the model was conducted using “reverse cumulative distribution of relative values,” alongside “boxplots of relative values”. We then used ten-fold cross-validation to validate the RF model. The RF algorithm was applied to rank the importance of m5C regulators using the “randomForest” package. A nomogram model was then built and developed by the “rmda” and “rms” packages to represent the selected m5C regulators. Furthermore, calibration curves were used to verify the consistency between actual and predicted occurrences of MM. We performed a decision curve analysis (DCA) to analyze the occurrence curve and evaluate whether the model offered beneficial decisions for MM patients (Iasonos et al., 2008). DCA is a method to evaluate clinical predictive models, diagnostic tests and molecular markers.

2.4 Correlation between m5C regulators and immune characteristics

Single-sample gene set enrichment analysis (ssGSEA) was performed to calculate an enrichment score, which estimates the relative abundance of each immune characteristic in each sample (Zhao et al., 2022). Immune characteristics consist of infiltrating immune cells (Zhang et al., 2020) and response reactions. The immune response reaction and related gene sets were accessed via ImmPort (http://www.immport.org). We derived enrichment correlations of differentially m5C regulators using the ssGSEA function to map the abundance of infiltrating immune cells and response reactions using the “GSEABase” package. Pearson’s correlation analysis was then used in order to calculate the correlation coefficient between m5C regulators and immune characteristics.

2.5 Consistent cluster analysis of m5C regulators

Based on the differential m5C regulators, unsupervised classification of the m5C dataset was performed for cluster analysis. Distinct m5C patterns were determined using “ConsensusClusterPlus” package in R, and the robustness of clustering was further systematically evaluated using a consensus clustering algorithm with 1,000 iterations (Wilkerson and Hayes, 2010). m5C regulator expression clusters were generated using principal component analysis (PCA). We subsequently evaluated the complex association between immune characteristics and different m5C clusters.

2.6 Pathway enrichment analysis of m5C clusters

Gene set variation analysis (GSVA) is a functional analysis to evaluate the enrichment of gene profiles through the expression dataset (Hänzelmann et al., 2013). The pathway gene sets, “h.all.v7.5.1 symbols” and “c2. cp.kegg.v7.5.1 symbols,” were obtained from the MSigDB database (http://www.gsea-msigdb.org/gsea/msigdb/index.jsp) and performed GSVA using the “GSVA” package to analyze pathway enrichment. Additionally, gene set enrichment analysis (GSEA) was applied to rank genes according to expression level, and we tested whether the gene set was enriched at the top or bottom of the ranking list (Subramanian et al., 2005). A normalized enrichment score (NES) was calculated as the primary statistic of GSEA. GSEA was conducted with the GSEA software (version 4.2.3) using the above official gene sets for enrichment.

2.7 Identification of m5C-related differentially expressed genes among three m5C clusters and biological function enrichment analysis

DEGs were screened among three distinct m5C clusters using the “limma” package, and m5C cluster-related genes were obtained by overlapping DEGs. Gene Ontology (GO) biological enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were visualized using a bar plot and bubble plot. To further investigate m5C methylation modification in MM patients, an m5C score was calculated using the following formula (Zhang et al., 2020):

m5Cscore=(PC1i+PC2i)(1)

Where PC represents the principal component, and i represents the DEG expression.

2.8 Statistical analysis

Statistical analyses were performed using R x64–4.1.3. Quantitative data and normally distributed variables were compared using t-tests, and non-normally distributed variables were compared using the Wilcoxon rank-sum test. Comparisons of more than two groups of variables were performed using a one-way analysis of variance or the Kruskal–Wallis test. Pearson’s correlation analysis was used for the correlation analysis. A p < 0.05 was considered statistically significant. To control for false discovery rate (FDR), Benjamini–Hochberg was employed for multiple hypothesis testing. All statistical tests were two-sided.

3 Results

3.1 Landscape of m5C regulators in healthy and multiple myeloma samples

We investigated 16 m5C methylation regulators, which included five writers (DNMT3A, DNMT3B, DNMT1, TRDMT1, and NSUN5), nine readers (MBD1, MBD2, MBD3, MBD4, MECP2, NTHL1, SMUG1, UNG, and YBX1), and two erasers (TET3 and TDG) (Figure 1A). The m5C regulators showed regular interactions via a PPI network (Figure 1B). The correlation analysis of the m5C regulators showed a close relationship between the m5C regulators, which suggested they function together (Figure 1C). The scatterplots between DNMT3A and MBD1 (r = −0.12), DNMT3A and MBD2 (r = −0.14), and DNMT3A and MBD4 (r = −0.14) showed significant negative correlations. Furthermore, the boxplot and the heatmap revealed that the expression of six significant m5C regulators in the MM samples compared to healthy samples (Figures 1D,E). Of all the 16 m5C regulators, we found that the expression of NSUN5, MBD3, MECP2, NTHL1, and TET3 was significantly upregulated, whereas DNMT3A was significantly downregulated in MM samples.

FIGURE 1
www.frontiersin.org

FIGURE 1. Landscape of m5C methylation regulators for healthy and MM samples. (A) The biological functions of m5C regulators, which are regulated by “writers,” “readers,” and “erasers” in the immune microenvironment. The diagram was drawn using Figdraw (https://www.figdraw.com). (B) Overview of the composition of m5C regulators and the PPI network among 16 m5C regulators in the STRING online database. (C) Correlation analysis of the m5C regulators. The size of each circle represents the correlation coefficient (negative values in blue and positive values in red). ***p < 0.001. Scatterplots show significant correlations between DNMT3A and MBD1, DNMT3A and MBD2, and DNMT3A and MBD4. (D,E) Boxplot and heatmap of the expression level of the m5C regulators for the healthy and MM samples.

3.2 Construction of a nomogram to predict the occurrence of multiple myeloma

Based on 16 m5C regulators adopted in this study, we constructed SVM and RF models to predict the risk of developing MM. According to the “reverse cumulative distribution of the residual” (Figure 2A) and the “boxplots of the residual” (Figure 2B), the RF model provided the minimal residuals. Most samples in RF model have relatively small residuals, indicating that the RF model is better. Ten-fold cross-validation was applied to evaluate the accuracy of the RF model (Figure 2C). The curve indicated that when more than two genes were included in the RF model, the accuracy remained stable above 0.85. This led to the RF model being considered an accurate predictor of MM. Based on the differentially expressed m5C regulators, we then built and developed a nomogram to evaluate the risk of developing MM (Figure 2D). The nomogram indicated that higher expression of NTHL1, MECP2, MBD3, NSUN5, and TET3 would result in a higher probability of the occurrence of MM. Calibration curves were used to determine the accuracy of the nomogram model (Figure 2E). The DCA curve also suggested that the nomogram was beneficial to MM patients (Figure 2F). The abscissa of DCA curve is Threshold Probability. When the probability of a patient being diagnosed with MM reaches a certain threshold, it is defined as positive and treatment measures are taken. At this time, there will be benefits for treatment patient and losses for untreated patients. The net benefit on the ordinate is the result of the benefits minus the losses. For example, in Figure 2F, assuming that we choose a diagnosis of MM with a predicted probability of 0.8, then for every 100 patients predicted via “m5C genes”, about 50 would benefit from it. For every 100 patients predicted using “all genes”, 40 patients benefit from it. We can see that in the threshold probability range (0–1), the net benefit of m5C genes is higher than all genes. Furthermore, the clinical impact curve indicated that the nomogram model had remarkably high predictive power (Figure 2G).

FIGURE 2
www.frontiersin.org

FIGURE 2. Construction of the nomogram to predict the occurrence of MM. (A) The RF model and SVM residual distributions were plotted using the reverse cumulative distribution of the residual. (B) Boxplots of the residual plot were obtained to test the residual distributions of the SVM and RF models. (C) Ten-fold cross-validation curve to assess the quality of MM prediction in the RF model. (D) Establishment of a nomogram model according to six differentially expressed m5C regulators. (E) The predictive ability of the nomogram model was evaluated and validated using a calibration curve. (F) Decision curves of the RF model of the m5C regulators. (G) Calibration curve of the nomogram for evaluating the occurrence of MM.

3.3 Relationship between the m5C regulators and immune characteristics of multiple myeloma

Correlations between differentially expressed m5C regulators and immune cells and immune response reactions provide additional insight into the relationships between differentially expressed m5C regulators and immune characteristics. Based on immune expression profiles, numerous differentially expressed immune characteristics were identified between healthy and MM samples (Figures 3A,B). The correlation analysis revealed that several immune cells were closely associated with differentially expressed m5C regulators (Figure 3C). Among all the immunocytes, CD56 (dim) natural killer cells were most positively correlated to MBD3 (r = 0.38), and immature B cells were most negatively correlated to NTHL1 (r = −0.38), which indicated that the number of immune cells in MM patients is affected by differentially expressed m5C regulators. The expression level of CD56 (dim) natural killer cells and MBD3, immature B cells and NTHL1 also differed between healthy and MM samples. Additionally, the most immune response reactions were closely related to differentially expressed m5C regulators according to the correlation (Figure 3D). The transforming growth factor-β (TGFβ) family member was most positively correlated to MBD3 (r = 0.43), and the TGFβ family member receptor was most negatively correlated to NTHL1 (r = −0.4), which indicated that the number of immune response reactions is affected by differentially expressed m5C regulators.

FIGURE 3
www.frontiersin.org

FIGURE 3. Relationship between m5C regulators and immune characteristics of MM. (A) Boxplot of the abundance of 22 immunocytes in the healthy and MM samples. (B) Boxplot of the abundance of 17 immune response reactions in the healthy and MM samples. (C) Heatmap showing correlations between the six m5C regulators and 22 immunocytes. The scatterplots showed the positive and negative correlations between m5C regulators and immunocytes. The expression data were presented in a violin box plot (right panel). (D) Heatmap showing correlations between the six m5C regulators and 17 immune response reactions. The scatterplots showed the positive and negative correlations between the m5C regulators and immune response reactions. The expression data were presented in a violin box plot (right panel).

3.4 Establishment of the m5C methylation modification clusters

To establish the methylation modification clusters, a non-negative matrix factorization was applied based on the MM samples (Figures 4A–C). Three distinct clusters were classified as having significant populations in the PCA; namely, m5C cluster A, m5C cluster B, and m5C cluster C (Figure 4D). The boxplot and heatmap showed the high NSUN5, MBD3, and NTHL1 expression in m5C cluster A, high DNMT3A expression in m5C cluster B, and high MECP2 expression in m5C cluster C (Figures 4E,F).

FIGURE 4
www.frontiersin.org

FIGURE 4. Establishment of m5C methylation modification patterns. (A) Consensus clustering of the cumulative distribution function (CDF) for k = 2–9. (B) Elbow plot showed the relative change in the area under the CDF curve. (C) The consensus heatmap defined three m5C clusters in 423 patients with MM. (D) PCA of the m5C clusters in MM patients. (E,F) Boxplot and heatmap showed the expression levels of m5C regulators among the three m5C clusters.

3.5 Immune biological functional characteristics of the three distinct m5C clusters

To deeply investigate the three m5C clusters, we compared the abundance of immune cells and immune response reactions. Results showed that various immunocytes differed in abundance in the three clusters (Figure 5A). Cluster A had a relatively higher number of immunocytes of CD56 (dim) natural killer cells and neutrophils than did clusters B and C. Cluster B had a higher level of eosinophil and T follicular helper cells, whereas active CD4+ T cells, CD56 (bright) natural killer cells, gamma delta T cells, immature B cells, natural killer T cells, natural killer cells, plasmacytoid dendritic cells, type17 T helper cells, and type 2 T helper cells were enriched in cluster C. As for immune response reactions, the TGFβ family member and tumor necrosis factor family member receptors were more active in cluster A than in clusters B and C. Antimicrobials and cytokines were more active in cluster B than in clusters A and C, and antigen processing and presentation, the B-cell receptor signaling pathway, interferon receptors, interleukins, natural killer cell cytotoxicity, the T-cell receptor signaling pathway, and TGFβ family member receptors were more active in cluster C than in clusters A and B (Figure 5B). These results suggested that cluster C played a more active role in immune characteristics. Taken together, our findings suggested that the m5C clusters possessed distinct immune characteristics and that differentially expressed m5C methylation regulators were critical for the regulation of the immune microenvironment in MM.

FIGURE 5
www.frontiersin.org

FIGURE 5. Immune characteristics of the three distinct m5C clusters. (A) Abundance of infiltrating immune cells in the three m5C modification clusters. (B) Differences in the 17 immune response reactions between the three m5C modification clusters. *p < 0.05; **p < 0.01; ***p < 0.001.

To further explore the biological functional characteristics of the three m5C clusters, we performed GSVA and GSEA to determine the enrichment of the biological pathways. The HALLMARK and KEGG pathways were compared between the clusters. Among the three m5C clusters, cluster C had more abundant HALLMARK and KEGG pathways, whereas cluster A had the fewest pathways (Figures 6A,C,E,G,I,K). Furthermore, the GSEA revealed that cluster A had the most significantly enriched terms related to the MYC targets V2 (NES = −1.705, p = 0.035, FDR = 0.198), and cluster B had the most enriched terms related to TGFβ signaling (NES = 1.925, p < 0.001, FDR = 0.024) in the HALLMARK pathway (Figure 6B). In the KEGG pathways, alanine aspartate and glutamate metabolism (NES = −1.746, p = 0.004, FDR = 0.447) were most enriched in cluster A, and pancreatic cancer (NES = 0.871, p = 0.002, FDR = 0.168) was most enriched in cluster B (Figure 6D). Similar enrichment pathways were observed in cluster C versus cluster A (Figures 6F,H) and cluster C versus cluster B (Figures 6J,L). After comprehensive GSVA and GSEA, we found that both methods enriched numerous similar HALLMARK and KEGG pathways.

FIGURE 6
www.frontiersin.org

FIGURE 6. Biological functional characteristics of the three distinct m5C clusters. (A–D) HALLMARK pathway and KEGG pathway between m5C clusters A and B ((A,B) for HALLMARK pathway and (C,D) for KEGG pathway). (E–H) HALLMARK pathway and KEGG pathway between m5C modification clusters A and C ((E,F) for HALLMARKS pathway and (G,H) for KEGG pathway). (I–L) HALLMARK pathway and KEGG pathway between m5C modification clusters B and C ((I,J) for HALLMARKS pathway and (K,L) for KEGG pathway).

3.6 Development of the m5C score and its biological functions

We performed the analysis of the DEGs of the three m5C clusters to confirm their potential biological functions and overlapped these DEGs to obtain the m5C cluster-related genes (Figure 7A). We identified 256 genes as m5C cluster-related genes, and we subsequently performed enrichment analysis. GO and KEGG enrichment results demonstrated that the tumor metabolism and immune-related pathways were enriched, which suggested that the m5C regulators exert a non-negligible role in MM (Figures 7B,C). To further examine the mechanism underlying the relationship between m5C and MM, we performed another consensus clustering analysis based on the 256 m5C cluster-related genes, and the samples were assigned to three distinct genomic patterns, which were named m5C gene clusters A, B, and C (Figure 7D). The boxplot showed statistically significant expression differences in the six significant m5C regulators among the three m5C gene clusters (Figure 7E). We then compared the abundance of immune cells and immune response reactions among the three m5C gene clusters. Results showed that most immunocytes and immune response reactions differed among the three clusters (Figures 7F,G). To predict the patterns of m5C gene clusters in the MM samples and assess the risk level of these patients, we implemented an m5C scoring scheme to assess m5C regulator expression patterns in MM samples and a Sankey plot to depict the score distribution (Figures 7H,I). Most samples of m5C cluster A are high risk, and most samples of gene cluster A are low risk; most samples of m5C cluster C are low risk, and most samples of gene cluster C are high risk. We then analyzed the differences in m5C scores among the m5C and gene clusters. Results revealed notable differences in m5C scores among the m5C clusters. The median of m5C cluster C was the lowest, and that of m5C cluster A was the highest, which indicated that a low m5C score was closely related to immune characteristics (Figure 7J). The m5C gene cluster C had the highest median m5C score, and gene cluster A had the lowest (Figure 7K).

FIGURE 7
www.frontiersin.org

FIGURE 7. Development of the m5C score and its biological functions. (A) Venn diagram showing 256 m5C-related DEGs for the three clusters. (B) GO enrichment analysis of m5C-related DEGs. (C) KEGG enrichment analysis of m5C-related DEGs. (D) The consensus matrix heatmap defined three m5C gene clusters. (E) Expression levels of the m5C regulators for each of the three m5C gene clusters. (F) Abundance of infiltrating immune cells in each of the three gene clusters. (G) Differences in the activity of 17 immune response reactions among the three gene clusters. (H) A Sankey diagram of the different m5C modification clusters and m5C scores. (I) A Sankey diagram of the different m5C gene clusters and m5C scores. (J) Differences in the m5C scores among the three m5C modification clusters (p < 0.001). (K) Differences in the m5C scores among the three m5C gene clusters (p < 0.001). *p < 0.05; **p < 0.01; ***p < 0.001.

4 Discussion

MM is a malignant tumor derived from terminally differentiated B lymphocytes and is characterized by the clonal proliferation of a large number of plasma cells in MM patients’ bone marrow (Das et al., 2022). MM tumor cells can remodel the tumor microenvironment and establish an immunosuppressive tumor microenvironment, which promotes the occurrence and development of MM (Minnie and Hill, 2020; Casey and Nakamura, 2021). The development of next-generation sequencing technology has demonstrated that m5C modification affects various disease processes, such as genomic stability, cancer cell differentiation, and inflammatory responses (Xue et al., 2020). Therefore, we believed that similar results of m5C modification could be observed in the MM immune microenvironment. To test this hypothesis, we used six GEO databases to obtain 60 healthy samples and 423 MM samples. Our study is the first study focused on m5C regulators in MM and shedding light on the links between m5C regulation and immune characteristics.

Firstly, we assessed the expressed gene profile of m5C regulators in the healthy and MM samples, and the differences indicated that m5C regulators were involved in MM progression. We also identified an RF model and selected six candidate m5C regulators (NTHL1, MECP2, MBD3, NSUN5, DNMT3A, and TET3) from the 16 m5C regulators to predict the occurrence of MM. A nomogram model to accurately predict the occurrence of MM was constructed, and the DCA curve demonstrated favorable calibration and benefit to MM patients when the nomogram was applied. DNMT3A plays a special role in the initiation of chromatin remodeling and gene expression regulation and is responsible for methylation pattern acquisition (Chen and Zhang, 2020). DNMT3A mutation carriers are characterized by increased expression of the T-cell alpha receptor constant chain and may be involved in monocyte-T-cell interactions (Abplanalp et al., 2021). NTHL1, MECP2, and MBD3 all have a binding domain that specifically maintain stability (Yano et al., 2006; Senarisoy et al., 2020). TET3 can catalyze the demethylation of m5C methyl groups under the synergistic effect of α-ketoglutarate and Fe2+ (Shen et al., 2021). It has not been reported whether any of the six differential m5C regulators are associated with MM. In m5C modifications, a methyl group attached to the fifth carbon of the cytosine ring in DNA and RNA molecules. This modification was first identified on DNA and later on RNA in the 1970s (Dubin and Stollar, 1975; Zin’kovskaia et al., 1978). For instance, MBDs could bind m5C and convert the methylation pattern information into appropriate functional cellular states. Chromosomal binding experiments indicate MBD-R2 and MECP2 associate on shared genomic loci (Gupta et al., 2016). All readers that “read” the information contained in these modifications to maintain stability and participate in translation and splicing.

We also investigated the correlation between m5C regulators and immune characteristics. For immunocytes, CD56 (dim) natural killer cells were most positively correlated with m5C regulators. The prognosis of MM patients has been shown to be influenced by CD56 (ElMenshawy et al., 2021), and DNMT3A-mutated AML patients have higher expression of CD56 (Hájková et al., 2012). The link between m5C regulation and the immune system suggests a close relationship between CD56 natural killer cells and MM. A set of immune reactions was correlated with m5C regulators, specifically the TGFβ family member and its receptor. The TGFβ family is a multifunctional cytokine and has long been recognized as an immune-suppressive factor (Tschernia and Gulley, 2022). It plays a major role in the beneficial immunosuppressive microenvironment of MM patients’ bone marrow niches (Alabanza et al., 2022), which indicates that m5C regulators are crucial to the progress of MM. However, there is no clear correlation between immune characteristics and m5C regulators. This may be due to the previous detection techniques limitations. For RNA sequencing, samples contain extremely few immune cells, so their abundance may not accurately reflect infiltration of immune cells (Stewart et al., 2020).

Our results further identified three m5C clusters with different immune characteristics and metabolism-related pathways. This immune pattern clustering approach can help us understand the underlying mechanism of immune regulation, allowing for the application of more precise molecular therapeutic approaches by clustering MM at the immune level, rather than simply at the phenotypic level. Various studies have confirmed that this strategy is feasible. For non-tumor diseases, several studies have applied this clustering method to study the impact of the immune microenvironment on disease (Zhang et al., 2021; Zhao et al., 2021). For tumors, one study divided gastric cancer into three patterns and demonstrated the impact of the tumor microenvironment on the disease, which offered new ideas for immunotherapy (Zhang et al., 2020). In our study, among the three different m5C clusters, cluster C had more immune characteristics and metabolism-related pathways than did clusters A and B. For example, cluster C was characterized by a higher number of immunocytes and immune response reactions, which included a greater abundance of plasmacytoid dendritic cells (pDCs) and interferon receptors, the former of which are increased in MM patients’ bone marrow and play a key role in the progression of MM (Ray et al., 2015). Moreover, pDCs are observed in m5C regulators, including the DNMT3A and NSUN family, and are regulated by interferon receptors (Bi et al., 2018; Fang et al., 2022). All three m5C clusters may have abundant metabolism-related pathways, which can help guide the identification of key m5C regulators and immune characteristics of MM. It may prove that MM can be classified as an alternate pathobiology-based classification based on the three distinct m5C clusters, which is related to clinical symptoms of the disease.

Finally, the m5C cluster-related genes and m5C gene clusters were identified. Given the need for individualized immunotherapy strategies for MM patients and improved understanding of m5C clustering patterns, developing a new m5C scoring system is urgently needed. We developed a scoring system to assess m5C regulator expression patterns in MM patients according to a previous study (Zhang et al., 2020). Based on the results of the m5C cluster-related genes, MM patients were divided into three m5C gene clusters. These genes’ expression may be influenced by m5C modification, and uncovering their biological functions helps to explain MM pathogenesis from the perspective of m5C modification. The m5C gene cluster C had the highest median m5C score. It has more activation in cytokines, and at the same time more CD56dim natural killer cells are seen in m5C gene cluster C. Most differential expressed m5C regulators are also up-regulated in m5C gene cluster C and MM. These results may suggest m5C regulators, cytokines, CDR56dim natural killer cells and MM have correlations. Our study reveals many similar correlations due to the abundance of results and a major scientific benefit of our study is helping other researchers identify key immune and m5C regulator features in MM quickly. Our study has this significance as one of its most important scientific implications.

However, there are some limitations in our study. More clinical data for each patient was not available, such as treatment and prognosis, for the longitudinal analysis. Correlation analysis of m5C clusters, pathological stages, and other clinical characteristics could not be performed on all samples. Besides, our findings were obtained mainly through bioinformatics analysis and require verification in subsequent single-cell sequencing and other experiments. Nevertheless, our findings provide new insights into the relationship between m5C regulators and the immune microenvironment of MM.

In conclusion, our study identified six differential m5C regulators and then built a nomogram to precisely predict the occurrence of MM. The three m5C clusters were shown to significantly impact the immune microenvironment and biological functional pathways, and these findings will be valuable for further progress in immunotherapy. Comprehensive analyses of m5C regulators in MM may offer a promising future for effective therapeutic strategies.

Data availability statement

The datasets presented in this study can be found in online repositories. The names of the repository/repositories and accession number(s) can be found in the article/Supplementary Material.

Author contributions

HR, CL, HW, and LZ participated in the design of the study. ZW, SC, XZ, and JR performed statistical analysis. HR wrote the manuscript. HQ and LZ revised the manuscript. Contributors all approved the submitted version of the article.

Funding

This work was supported by National Natural Science Foundation of China (82102482, 82072371), Leading Talent Project of Shanghai Huangpu District (2020-1-28), Shanghai Health and Medical Development Foundation under Grant (201972), Shanghai Science and Technology Committee under Grant (21ZR1478200).

Acknowledgments

We would like to thank the authors who provided the Gene Expression Omnibus Database public datasets.

Conflict of interest

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.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abplanalp, W. T., Cremer, S., John, D., Hoffmann, J., Schuhmacher, B., Merten, M., et al. (2021). Clonal hematopoiesis-driver DNMT3A mutations alter immune cells in heart failure. Circ. Res. 128, 216–228. doi:10.1161/CIRCRESAHA.120.317104

PubMed Abstract | CrossRef Full Text | Google Scholar

Alabanza, L. M., Xiong, Y., Vu, B., Webster, B., Wu, D., Hu, P., et al. (2022). Armored BCMA CAR T cells eliminate multiple myeloma and are resistant to the suppressive effects of TGF-β. Front. Immunol. 13, 832645. doi:10.3389/fimmu.2022.832645

PubMed Abstract | CrossRef Full Text | Google Scholar

Allegra, A., Petrarca, C., Di Gioacchino, M., Casciaro, M., Musolino, C., and Gangemi, S. (2022). Modulation of cellular redox parameters for improving therapeutic responses in multiple myeloma. Antioxidants 11, 455. doi:10.3390/antiox11030455

PubMed Abstract | CrossRef Full Text | Google Scholar

Bestor, T. H. (1988). Cloning of a mammalian DNA methyltransferase. Gene 74 (1), 9–12. doi:10.1016/0378-1119(88)90238-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Bi, E., Li, R., Bover, L. C., Li, H., Su, P., Ma, X., et al. (2018). E-cadherin expression on multiple myeloma cells activates tumor-promoting properties in plasmacytoid DCs. J. Clin. Invest. 128, 4821–4831. doi:10.1172/JCI121421

PubMed Abstract | CrossRef Full Text | Google Scholar

Casey, M., and Nakamura, K. (2021). The Cancer-immunity cycle in multiple myeloma. Immunotargets. Ther. 10, 247–260. doi:10.2147/ITT.S305432

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Z., and Zhang, Y. (2020). Role of mammalian DNA methyltransferases in development. Annu. Rev. Biochem. 89, 135–158. doi:10.1146/annurev-biochem-103019-102815

PubMed Abstract | CrossRef Full Text | Google Scholar

Cowan, A. J., Green, D. J., Kwok, M., Lee, S., Coffey, D. G., Holmberg, L. A., et al. (2022). Diagnosis and management of multiple myeloma: a review. JAMA 327, 464–477. doi:10.1001/jama.2022.0003

PubMed Abstract | CrossRef Full Text | Google Scholar

Das, S., Juliana, N., Yazit, N., Azmani, S., and Abu, I. F. (2022). Multiple myeloma: challenges encountered and future options for better treatment. Int. J. Mol. Sci. 23, 1649. doi:10.3390/ijms23031649

PubMed Abstract | CrossRef Full Text | Google Scholar

Dubin, D. T., and Stollar, V. (1975). Methylation of Sindbis virus "26S" messenger RNA. Biochem. Biophys. Res. Commun. 66 (4), 1373–1379. doi:10.1016/0006-291x(75)90511-2

PubMed Abstract | CrossRef Full Text | Google Scholar

ElMenshawy, N., Farag, N. A., Atia, D. M., Abousamra, N., Shahin, D., Fawzi, E., et al. (2021). Prognostic relevance of concordant expression CD69 and CD56 in response to bortezomib combination therapy in multiple myeloma patients. Cancer Invest. 39, 777–782. doi:10.1080/07357907.2021.1964521

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, X., Miao, C., Zeng, T., Chu, W., Zheng, Y., Sun, X., et al. (2022). Role of m(5) C RNA methylation regulators in colorectal cancer prognosis and immune microenvironment. J. Clin. Lab. Anal. 36, e24303. doi:10.1002/jcla.24303

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, G., Wang, H., Shi, X., Ye, L., Yan, K., Chen, Z., et al. (2020). Disease activity-associated alteration of mRNA m(5) C methylation in CD4(+) T Cells of systemic lupus erythematosus. Front. Cell Dev. Biol. 8, 430. doi:10.3389/fcell.2020.00430

PubMed Abstract | CrossRef Full Text | Google Scholar

Gupta, T., Morgan, H. R., Bailey, J. A., and Certel, S. J. (2016). Functional conservation of MBD proteins: MeCP2 and Drosophila MBD proteins alter sleep. Genes Brain Behav. 15 (8), 757–774. doi:10.1111/gbb.12314

PubMed Abstract | CrossRef Full Text | Google Scholar

Hájková, H., Marková, J., Haškovec, C., Sárová, I., Fuchs, O., Kostečka, A., et al. (2012). Decreased DNA methylation in acute myeloid leukemia patients with DNMT3A mutations and prognostic implications of DNA methylation. Leuk. Res. 36, 1128–1133. doi:10.1016/j.leukres.2012.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Hänzelmann, S., Castelo, R., and Guinney, J. (2013). GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinforma. 14, 7. doi:10.1186/1471-2105-14-7

CrossRef Full Text | Google Scholar

Hu, J., Othmane, B., Yu, A., Li, H., Cai, Z., Chen, X., et al. (2021). 5mC regulator-mediated molecular subtypes depict the hallmarks of the tumor microenvironment and guide precision medicine in bladder cancer. BMC Med. 19 (1), 289. doi:10.1186/s12916-021-02163-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, Z., Pan, J., Wang, H., Du, X., Xu, Y., Wang, Z., et al. (2021). Prognostic significance and tumor immune microenvironment heterogenicity of m5C RNA methylation regulators in triple-negative breast cancer. Front. Cell Dev. Biol. 9, 657547. doi:10.3389/fcell.2021.657547

PubMed Abstract | CrossRef Full Text | Google Scholar

Iasonos, A., Schrag, D., Raj, G. V., and Panageas, K. S. (2008). How to build and interpret a nomogram for cancer prognosis. J. Clin. Oncol. 26, 1364–1370. doi:10.1200/JCO.2007.12.9791

PubMed Abstract | CrossRef Full Text | Google Scholar

Jadoon, Y., and Siddiqui, M. A. (2021). Immunotherapy in multiple myeloma. Cancer Treat. Res. Commun. 29, 100468. doi:10.1016/j.ctarc.2021.100468

PubMed Abstract | CrossRef Full Text | Google Scholar

Leek, J. T., Johnson, W. E., Parker, H. S., Jaffe, A. E., and Storey, J. D. (2012). The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics 28, 882–883. doi:10.1093/bioinformatics/bts034

PubMed Abstract | CrossRef Full Text | Google Scholar

Lv, X., Liu, X., Zhao, M., Wu, H., Zhang, W., Lu, Q., et al. (2021). RNA methylation in systemic lupus erythematosus. Front. Cell Dev. Biol. 9, 696559. doi:10.3389/fcell.2021.696559

PubMed Abstract | CrossRef Full Text | Google Scholar

Minnie, S. A., and Hill, G. R. (2020). Immunotherapy of multiple myeloma. J. Clin. Invest. 130, 1565–1575. doi:10.1172/JCI129205

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakamura, K., Smyth, M. J., and Martinet, L. (2020). Cancer immunoediting and immune dysregulation in multiple myeloma. Blood 136, 2731–2740. doi:10.1182/blood.2020006540

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, J., Huang, Z., and Xu, Y. (2021). m5C RNA methylation regulators predict prognosis and regulate the immune microenvironment in lung squamous cell carcinoma. Front. Oncol. 11, 657466. doi:10.3389/fonc.2021.657466

PubMed Abstract | CrossRef Full Text | Google Scholar

Ray, A., Das, D. S., Song, Y., Richardson, P., Munshi, N. C., Chauhan, D., et al. (2015). Targeting PD1-PDL1 immune checkpoint in plasmacytoid dendritic cell interactions with T cells, natural killer cells and multiple myeloma cells. Leukemia 29, 1441–1444. doi:10.1038/leu.2015.11

PubMed Abstract | CrossRef Full Text | Google Scholar

Senarisoy, M., Barette, C., Lacroix, F., De Bonis, S., Stelter, M., Hans, F., et al. (2020). Förster resonance energy transfer based biosensor for targeting the hNTH1-YB1 interface as a potential anticancer drug target. ACS Chem. Biol. 15, 990–1003. doi:10.1021/acschembio.9b01023

PubMed Abstract | CrossRef Full Text | Google Scholar

Shen, H., Ontiveros, R. J., Owens, M. C., Liu, M. Y., Ghanty, U., Kohli, R. M., et al. (2021). TET-mediated 5-methylcytosine oxidation in tRNA promotes translation. J. Biol. Chem. 296, 100087. doi:10.1074/jbc.RA120.014226

PubMed Abstract | CrossRef Full Text | Google Scholar

Stewart, B. J., Ferdinand, J. R., and Clatworthy, M. R. (2020). Using single-cell technologies to map the human immune system - implications for nephrology. Nat. Rev. Nephrol. 16, 112–128. doi:10.1038/s41581-019-0227-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Subramanian, A., Tamayo, P., Mootha, V. K., Mukherjee, S., Ebert, B. L., Gillette, M. A., et al. (2005). Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. U. S. A. 102, 15545–15550. doi:10.1073/pnas.0506580102

PubMed Abstract | CrossRef Full Text | Google Scholar

Tschernia, N. P., and Gulley, J. L. (2022). Tumor in the crossfire: inhibiting TGF-β to enhance cancer immunotherapy. BioDrugs 36, 153–180. doi:10.1007/s40259-022-00521-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, N., Tang, H., Wang, X., Wang, W., and Feng, J. (2017). Homocysteine upregulates interleukin-17A expression via NSun2-mediated RNA methylation in T lymphocytes. Biochem. Biophys. Res. Commun. 493, 94–99. doi:10.1016/j.bbrc.2017.09.069

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, X., He, L., Huang, X., Zhang, S., Cao, W., Che, F., et al. (2021). Recent progress of exosomes in multiple myeloma: pathogenesis, diagnosis, prognosis and therapeutic strategies. Cancers 13, 1635. doi:10.3390/cancers13071635

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilkerson, M. D., and Hayes, D. N. (2010). ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics 26, 1572–1573. doi:10.1093/bioinformatics/btq170

PubMed Abstract | CrossRef Full Text | Google Scholar

Xue, M., Shi, Q., Zheng, L., Li, Q., Yang, L., and Zhang, Y. (2020). Gene signatures of m5C regulators may predict prognoses of patients with head and neck squamous cell carcinoma. Am. J. Transl. Res. 12, 6841–6852.

PubMed Abstract | Google Scholar

Yano, A., Kodama, Y., Koike, A., Shinya, T., Kim, H. J., Matsumoto, M., et al. (2006). Interaction between methyl CpG-binding protein and ran GTPase during cell division in tobacco cultured cells. Ann. Bot. 98, 1179–1187. doi:10.1093/aob/mcl211

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, X., Zhang, Q., Gao, F., Zhang, M., Zheng, Q., He, Y., et al. (2021). Predictive value of m5C regulatory gene expression in pancreatic adenocarcinoma. Sci. Rep. 11, 17529. doi:10.1038/s41598-021-96470-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, B., Wu, Q., Li, B., Wang, D., Wang, L., and Zhou, Y. L. (2020). m(6)A regulator-mediated methylation modification patterns and tumor microenvironment infiltration characterization in gastric cancer. Mol. Cancer 19, 53. doi:10.1186/s12943-020-01170-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X., Zhang, S., Yan, X., Shan, Y., Liu, L., Zhou, J., et al. (2021). m6A regulator-mediated RNA methylation modification patterns are involved in immune microenvironment regulation of periodontitis. J. Cell. Mol. Med. 25, 3634–3645. doi:10.1111/jcmm.16469

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, H., Pan, S., Duan, J., Liu, F., Li, G., Liu, D., et al. (2021). Integrative analysis of m(6)A regulator-mediated RNA methylation modification patterns and immune characteristics in lupus nephritis. Front. Cell Dev. Biol. 9, 724837. doi:10.3389/fcell.2021.724837

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, R., Pan, Z., Li, B., Zhao, S., Zhang, S., Qi, Y., et al. (2022). Comprehensive analysis of the tumor immune microenvironment landscape in glioblastoma reveals tumor heterogeneity and implications for prognosis and immunotherapy. Front. Immunol. 13, 820673. doi:10.3389/fimmu.2022.820673

PubMed Abstract | CrossRef Full Text | Google Scholar

Zin'kovskaia, G. G., Berdyshev, G. D., and Vaniushin, B. F. (1978). Tissue-specific decrease and change in the character of DNA methylation in cattle with aging. Biokhimiia 43 (10), 1883–1892.

PubMed Abstract | Google Scholar

Keywords: multiple myeloma, 5-methylcytosine, immune microenvironment, immune therapy, biological function

Citation: Ren H, Liu C, Wu H, Wang Z, Chen S, Zhang X, Ren J, Qiu H and Zhou L (2022) m5C Regulator-mediated methylation modification clusters contribute to the immune microenvironment regulation of multiple myeloma. Front. Genet. 13:920164. doi: 10.3389/fgene.2022.920164

Received: 14 April 2022; Accepted: 19 July 2022;
Published: 25 August 2022.

Edited by:

Nguyen Quoc Khanh Le, Taipei Medical University, Taiwan

Reviewed by:

Abhijit Chakraborty, La Jolla Institute for Immunology, United States
Valentina S. Caputo, London South Bank University, United Kingdom
Ken Mills, Queen’s University Belfast, United Kingdom

Copyright © 2022 Ren, Liu, Wu, Wang, Chen, Zhang, Ren, Qiu and Zhou. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Lin Zhou, lynnzhou36@126.com; Huiying Qiu, qiuhy5@126.com

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.