Abstract
Background
   Glioblastoma is one of the most common brain cancers in adults, and is
   characterized by recurrence and little curative effect. An effective
   treatment for glioblastoma patients remains elusive worldwide.
   7-methylguanosine (m7G) is a common RNA modification, and its role in
   tumors has become a research hotspot.
Methods
   By searching for differentially expressed genes related to m7G, we
   generated a prognostic signature via cluster analysis and established
   classification criteria of high and low risk scores. The effectiveness
   of classification was validated using the Non-negative matrix
   factorization (NMF) algorithm, and repeatedly verified using training
   and test groups. The dimension reduction method was used to clearly
   show the difference and clinical significance of the data. All analyses
   were performed via R (version 4.1.2).
Results
   According to the signature that included four genes (TMOD2, CACNG2,
   PLOD3, and TMSB10), glioblastoma patients were divided into high and
   low risk score groups. The survival rates between the two groups were
   significantly different, and the predictive abilities for 1-, 3-, and
   5-year survivals were effective. We further established a Nomogram
   model to further examine the signature,as well as other clinical
   factors, with remaining significant results. Our signature can act as
   an independent prognostic factor related to immune-related processes in
   glioblastoma.
Conclusions
   Our research addresses the gap in knowledge in the m7G and glioblastoma
   research fields. The establishment of a prognostic signature and the
   extended analysis of the tumor microenvironment, immune correlation,
   and tumor mutation burden further suggest the important role of m7G in
   the development and development of this disease. This work will provide
   support for future research.
Supplementary Information
   The online version contains supplementary material available at
   10.1186/s12885-022-09791-y.
   Keywords: m7G methylation, Immune infiltration, Signature, Biomarkers,
   Cancer, Health informatics, Neuroscience, Risk factors
Introduction
   Glioblastoma is the most common adult primary brain cancer. It is the
   most malignant glioma of astrocytic tumors. About 50% of the primary
   central nervous system malignant tumors are categorized as
   glioblastoma, with an incidence rate of 3.2 cases per 100,000 people
   [[41]1]. Despite the unremitting efforts of researchers to design new
   treatment strategies, primary glioblastoma has always persisted in an
   invasive manner and the patient mortality rates have increased
   accordingly [[42]2]. Therefore, preventing the recurrence of
   glioblastoma is necessary to reduce its mortality. For patients with
   newly diagnosed glioblastoma, the standard treatment regimen consists
   of safe and feasible surgery, radiotherapy, and temozolomide
   chemotherapy for up to six treatment cycles [[43]3, [44]4]. Patients
   with recurrence are treated by reoperation and radiotherapy. However,
   these methods have little effect on improving the overall survival rate
   [[45]5]. Therefore, finding new and feasible treatments is a common
   goal among researchers.
   RNA modification is a reversible and dynamically regulated process that
   is involved in major biological processes [[46]6]. Transfer RNA (tRNA)
   modification is the most common RNA modification [[47]7]. tRNA is a
   classical non-coding RNA. It functions in the translation process by
   providing amino acids to ribosomes according to the specific codon in
   the mRNA. tRNA is modified to form a precise L-shaped structure to
   perform these functions [[48]8, [49]9]. In tRNA modification,
   7-methylguanosine (m7G) is one of the most conservative modified
   nucleosides, which widely exists in eubacteria, eukaryotes, and a few
   archaea [[50]10]. In most cases, m7G modification occurs at position 46
   of the variable region [[51]11]. m7G is installed at the 5′ cap in a
   co-transcriptional manner during transcription initiation. It can
   stabilize transcripts, prevent exonucleolysis and degradation, and
   regulate the mRNA life cycle [[52]12].
   For the relationship between RNA modification and glioblastoma, Cui et
   al. showed that mettl3 and mettl14 can inhibit tumor formation and
   demonstrated that m6A methylation can actually reduce the stability of
   key carcinogenic transcripts [[53]13]. However, Visvanathan et al.
   confirmed that mettl3 is a cancer-promoting gene that can promote the
   survival of glioma cancer cells by stabilizing SRY box 2 (Sox2,
   [[54]14]). These two contradictory experimental results make it
   difficult to clearly explain the effects of RNA modifications in
   glioblastoma. Some studies have shown that methyltransferase like 1
   (mettl1) is a tumor suppressor gene in colon cancer [[55]15]. Other
   reports have shown that eukaryotic translation initiation factor 4E
   (eIF4E) is very important for cancer cell transformation and has
   oncogenic potential in cancer development, as a eukaryotic translation
   initiation factor, it is significant to bind to the m7G cap existing at
   the 5 ‘- UTR of most eukaryotic mRNAs [[56]16]. However, the
   relationship between m7G and glioblastoma has rarely been explored,
   giving us a strong curiosity in this field.
   Thus, in this article, we cluster patients according to their
   expression of m7G-related genes and construct a scoring signature in
   which risk score is significantly related to clinical features and
   disease progression. We displayed the impact of m7G on glioblastoma as
   clearly as possible by using dimensionality reduction methods. Our
   results suggest a possible role of m7G-related genes that may indicate
   its potential as a therapeutic target in glioblastoma.
Methods
Glioblastoma dataset sources
   Gene expression, clinical features, and simple nucleotide variation of
   glioblastoma samples were obtained from the following public databases:
   The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and
   Chinese Glioma Genome Atlas (CGGA). Specifically, [57]GSE13041 of the
   GEO dataset was included in the analysis. Copy number variations (CNVs)
   of glioblastoma was downloaded through UCSC Xena
   ([58]http://xena.ucsc.edu/). Genes related to m7G were extracted from
   Gene Set Enrichment Analysis (GSEA,
   [59]http://www.gsea-msigdb.org/gsea/index.jsp), including
   GOBP_RNA_CAPPING (n = 34), GOMF_M7G_5_PPPN_DIPHOSPHATASE_ACTIVITY
   (n = 8), GOMF_RNA_7_METHYLGUANOSINE_CAP_BINDING (n = 12), and
   GOMF_RNA_CAP_BINDING (n = 19). After sorting and deleting duplicate
   genes, a total of 46 m7G-related and 11 immune checkpoint inhibitor
   (ICI) -related genes were included in the study. The specific genes
   were shown in Table S[60]1. Among all the 46 obtained m7G-related
   genes, 44 of them were analyzed here, the reason of which was that only
   these 44 of 46 m7G related genes were expressed in the merge samples of
   TCGA, GEO and CGGA.
Non-negative matrix factorization (NMF) clustering
   NMF is a novel way of clustering. Using the NMF R package, it can
   extricate sample classification from difficult positions where gene
   space is in high dimensionality and there are too few samples to
   further explore [[61]17]. With this method, 659 patient records were
   divided into groups A and B according to their expression levels of
   m7G-related genes. The classification of 659 patients into groups C1,
   C2, and C3 from the different genes of group A and B additionally
   utilized the NMF cluster method.
Gene set variation analysis (GSVA) and enrichment analysis
   GSVA is an updated algorithm to GSE, being the starting point of
   pathway-centric models of biology [[62]18]. It can detect minimal
   changes to biological pathways and calculate pathway activity scores.
   In this study, we chose c2.cp.kegg.v7.4.symbols for the gene set
   [[63]19], which was downloaded from the Molecular Signatures Database
   (MSigDB) database. The limma R package was then used to estimate the
   different biological processes that were enriched.
Signature construction and nomogram formation
   To verify m7G-related gene modifications in glioblastoma, the gene
   scoring system, named as the m7G score, was generated. After
   classification according to the expression levels of m7G-related genes,
   the differentially expressed genes (DEGs) of each group were found.
   Least absolute shrinkage and selection operator (LASSO) regression was
   cited to determine the optimal number of genes for the stability of the
   signature. Univariate Cox regression analysis was conducted to estimate
   the weight of each gene in the signature. The risk score formula is as
   follows:
   [MATH:
   Riskcor
   e=∑i=1
   NCoefi·<
   mrow>Expi
   :MATH]
   where Exp is the gene expression level and Coef is the weight
   coefficient of each gene.
Application of single-sample gene-set enrichment analysis (ssGSEA) in the
tumor microenvironment (TME)
   ssGSEA scoring was used to quantify the relative cell infiltration with
   respect to the TME in the glioblastoma patient cohort. A higher score
   indicates a greater abundance of cell infiltration. Specifically, the
   ESTIMATE score is positively related to the immune score and the
   stromal score, and the sum of the Estimation of STromal and Immune
   cells in MAlignant Tumours using Expression data (ESTIMATE) score and
   tumor score is unified. High tumor purity indicates the presence of
   more tumor cells and less immune and stromal cells within the TME
   [[64]20].
Dimensionality reduction of data
   Abstract data and ambiguous display methods can impede data
   interpretation and comprehension. Thus, we are committed to attempting
   various ways of exhibition of samples. Among the emerging algorithms,
   the dimensionality reduction method attracted our attention. We used
   decision curve analysis (DCA) and principal component analysis (PCA) to
   better display the significance of the analysis
Comparison of different signatures
   In order to further test the practicability of our new signature, we
   compared it with four other GBM signatures published in recent three
   years [[65]21–[66]24], and the results were displayed by C-index and
   Root Mean Square (RMS) value.
Cell culture and generation of lentiviral-transfected cell lines
   The human glioma cell lines (LN18 and T98G) were obtained from the Cell
   Bank of Shanghai Institutes of Biological Sciences, Chinese Academy of
   Sciences (Shanghai, China). The LN18 and T98G cells were cultured in
   RPMI 1640 medium (Gibco, CA, USA) with 10% fetal bovine serum (FBS,
   Gibco, USA) and 1% penicillin-streptomycin solution (Gibco, CA, USA) at
   37 °C in a humidified incubator containing 5% CO2. All of the cell
   lines tested negative for mycoplasma using a Mycoplasma Detection kit
   (Lonza). For generation of the inducible POLD3 knockdown cell lines was
   achieved using a pool of siRNA duplexe (lentiviral inducible human
   siRNA) using the following human PLOD3-specific siRNAs, synthesized by
   Genolution: #siRNA1, 5′- GGU UAA AGA AGG AAA UGG AUU − 3′; #siRNA2, 5′-
   GGA AGU ACA AGG AUG AUG AUG ACG ACG A - 3′. The siRNA duplexes were
   transfected into cells using Lipofectamine® RNAiMAX Reagent according
   to the manufacturer’s protocols.
Western blot
   Human gliomaLN18 and T98G cells (1 × 105) were seeded in 6-well plates.
   Western blot analyzes were performed as described previously [[67]25].
   Briefly, after 24 h, cells were treated for indicated times with DMSO
   (vehicle) or FHP01 or XAV939 (Merck). After transfected, cells were
   washed with cold PBS and total protein extracts obtained by adding RIPA
   Lysis buffer. After mechanicals detachment with cell scrapers, total
   lysates were collected in tubes, vortexed, and incubated for 15 min.
   For Western blot analysis, 10 μg of proteins derived from total lysates
   was loaded on 8% polyacrylamide gels with 1× of Laemmli buffer and
   resolved by SDS-PAGE, transferred to Immobilon-P PVDF membrane
   (Millipore, IPVH00010), probed with PLOD3 antibody (Themo Fisher,
   Product #PA5–106279) and GAPDH monoclonal antibody (Themo Fisher,
   Product #AM4300).
Cell proliferation assay
   The stably transfected LN18 and T98G cells were divided into negative
   control and siRNA1, siRNA2 transfected groups and seeded onto a 96-well
   plate at a density of 5 × 104 cells/ml. Next, the Cell Counting Kit-8
   (CCK-8 Kit; Dojindo, Japan), based on the manufacturer’s instructions,
   was added to determine the proliferative capacity of cells. Optical
   density (OD) values were obtained at 450 nm and was measured at 1, 2,
   3, 4 and 5 days after seeding using an automatic microplate reader
   (TEAN, Swiss). Three replicate analyses were performed for each sample.
Transwell assay
   A transwell cell migration assay was used to test the ability of cells
   to metastasize. The cell density of different groups was adjusted to
   1 × 105 cells/ml, and 100 μl cell suspension of different groups were
   added to the upper chamber with or without Matrigel (Corning, USA). The
   medium containing 20% FBS was added in the lower 24-well plate chamber.
   After 24 h, the bottom LN18 and T98G cells were treated with 4%
   polyoxymethylene for 15 min, deionized water, and 0.1% crystal violet
   for 30 min. Finally, the LN18 and T98G cells migrating to the lower
   surface of transwell chamber were counted using a microscope in six
   random fields utilizing a 200x microscope.
Statistical analysis
   Chi-square or Fisher’s exact tests were used to compare categorical
   variables. Two groups of continuous variables were compared using a t
   test, while three or more groups used one-way ANOVA and Kruskal-Wallis
   tests. Prognostic analysis utilized the survival R package and
   Kaplan-Meier method to examine the difference. In the methods mentioned
   above, P < 0.05 indicated statistical significance. A receiver
   operating characteristic (ROC) curve was employed to validate the
   effectiveness of prediction. An area under curve (AUC) > 0.7 was
   considered prominent. R (version 4.1.2) and R Bioconductor package were
   the foundation of the analysis.
Results
Variation of m7G-related genes in glioblastoma
   A total of 46 m7G-related genes were obtained from the GSEA dataset
   (Table S[68]1). Gene mutations in somatic cells are displayed in
   Fig. [69]1A. Overall, 27 of 390 samples experienced mutations with an
   incidence of 6.92%. From the CNV data from TCGA cohort, the mutations
   of m7G-related genes in glioblastoma were displayed in detail (Fig.
   [70]1B). It turned out that the main alteration was dominated by the
   deletion of copy number. The relationship between gene position on the
   chromosome and CNV mutation was visualized through a cycle graph (Fig.
   [71]1C). Furthermore, we examined the expression levels of each
   m7G-related gene in glioblastoma and normal samples. The results
   illustrate that all m7G-related genes are distinctly expressed in
   normal tissues and tumors with significant differences. Interestingly,
   all m7G-related genes other than NUDT3, EIF4E1B, and EIF4E3 were more
   highly expressed in normal samples compared with tumors (Fig. [72]1D).
   The prognostic analysis of each gene was also conducted, showing highly
   significant different survival rates according to gene expression (Fig.
   S[73]2). Consistent with the impact of genes on survival, the genes
   were defined as risk factors, whose expression levels were negatively
   correlated with survival. Favorable factors were genes with a positive
   relationship between expression levels and survival. The expression
   correlation network of m7G-related genes is depicted in Fig. [74]1E.
   The results represented above reveal the comprehensive landscape of
   m7G-related genes and glioblastoma.
Fig. 1.
   [75]Fig. 1
   [76]Open in a new tab
   Variation of m7G related genes in glioblastoma. A CNV of m7G related
   genes in somatic cells. The mutation frequency is listed on the right.
   B Copy number of each m7G related gene in detail. GAIN infers to
   amplification and LOSS indicate deletion. C Gene location on chromosome
   with mutation information. Blue dots are identical to deletion and red
   dots are amplification. D Expression level of m7G related genes in
   normal and Glioblastoma samples. E Expression modification of m7G
   related genes and their roles in regulation. F Cumulative distribution
   function curve proves the most effect way of clustering. G Grouping
   based on the expression of m7G related genes. Group 1 indicate group A
   and group 2 means group B. H Group A and B are separated, proving the
   significance of grouping. I Survival analysis between group A and B.
   P < 0.05 is witnessed as significant
Clustering by m7G expression level
   With the help of the sva R package, data from TCGA, CGGA, and
   [77]GSE13041 were merged to form a new sequence. When k = 2, the best
   clustering effect was obtained (Fig. [78]1F). Then, the new sequence of
   patients was divided into group A and B according to the expression
   levels of m7G-related genes (Fig. [79]1G). The PCA dimensionality
   reduction method validated the effectiveness of the grouping (Fig.
   [80]1H). The survival status is clearly significantly different between
   group A and group B (Fig. [81]1I). The relationship between the
   grouping, data source, and clinical manifestation is depicted in
   Fig. [82]2A. GSVA pathway enrichment analysis according to grouping
   shows that pathways in group A are downregulated, while upregulated in
   group B (Fig. [83]2B). The allocation is also significantly related to
   immune cells, implying an interaction between m7G and immune-related
   processes (Fig. [84]2C). To examine the genome wide changes between the
   two groups, 1253 genes with a prominent expression difference were
   listed (Table S[85]2). Notably, the different genes are related to
   brain disease in accordance with disease ontology analysis (Fig. [86]2D
   and Table S[87]3). Gene ontology (GO) enrichment analysis indicates a
   negative correlation between genes obtained above and synapse
   organization, and a positive correlation of leukocyte-mediated immunity
   and myeloid leukocyte activation (Fig. [88]2E and Table S[89]4). Kyoto
   Encyclopedia of Genes and Genomes (KEGG) enrichment analysis shows an
   accumulation of genes in synapse-related pathways, such as the
   dopaminergic synapse and synaptic vesicle cycle, and tumorigenesis,
   such as transcriptional misregulation in cancer and glioma (Fig. [90]2F
   and Table S[91]5). These results suggest that m7G participates in the
   development of glioblastoma through synapse-related pathways.
Fig. 2.
   [92]Fig. 2
   [93]Open in a new tab
   Conclusion and enrichment analysis of DEGs. A Heat map merging clinical
   information, m7G related gene expression and m7G cluster group. B Gene
   set variation analysis (GSVA) enrichment analysis of biological pathway
   between m7G cluster A and B. C Different content of immune cells in
   different group displayed respectively. D Disease Ontology analysis
   aiming at different expression genes (DEGs) of m7G cluster. E, F Gene
   Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment
   analysis on the basis of DEGs mentioned above
   Consistent with the DEGs, patients were grouped into three portions
   with the best effect, demonstrated by the NMF algorithm (Fig. [94]3A,
   B). The survival time and expression levels of m7G-related genes are
   remarkably different among these groups, demonstrating the conspicuous
   diversity and effectiveness of the division (Fig. [95]3C, D). The
   different gene expression levels of each patient and clinical features
   were distributed by group (Fig. [96]3E). Most of the genes had opposite
   expression patterns between groups C2 and C3, while group C1 showed
   random expression patterns.
Fig. 3.
   [97]Fig. 3
   [98]Open in a new tab
   Gene cluster of glioblastoma patients. A Non-negative matrix
   factorization (NMF) algorithm confirm that three group is the most
   suitable way of classification. B The effectiveness of different
   grouping methods according to NMF algorithm. C Survival analysis of
   different gene clusters with p < 0.05 as statistically significant
   borderline. D Expression of m7G related genes in gene clusters C1, C2
   and C3. E Heat map of clinical information, clusters and DEGs
Foundation of the m7G score signature
   To further investigate the predictive value of these genes, we aimed to
   establish a prognostic signature for analog computation. First, by
   merging the expression levels of the different genes and survival
   information, 53 prognosis-related differential genes were identified
   through univariate Cox regression with hazard ratio (HR) < 0.6 and
   HR > 1.6 as critical values (Table S[99]6). Then, the merged group of
   patients was separated to the training group (n = 330) and test group
   (n = 329) with the assistance of the caret R package. Next, using the
   data from the training group, four genes (TMOD2, CACNG2, PLOD3 and
   TMSB10) were selected to form the signature. The risk score calculation
   formula is as follows: risk score = (− 0.3206 × TMOD2
   expression) + (− 0.2556 × CACNG2 expression) + (0.3019 × PLOD3
   expression) + (0.2177 × TMSB10 expression) (Fig. [100]4A). Patients in
   the training group were marked as high risk or low risk relative to the
   median risk score value (1.1168). Patients in the test group and the
   entire samples were also considered as high- or low- risk group using
   the median risk score. The modeling process is displayed in Fig.
   [101]4B.
Fig. 4.
   [102]Fig. 4
   [103]Open in a new tab
   Construction of signature and statistically validation. A Column chart
   represents the weight of genes in the signature. B Sankey diagram to
   better state the modeling progress. C, D Phenotypic relationship
   between cluster and risk score. E Relationship of m7G related genes in
   high and low risk score group. F, G Forest maps illustrate uni and
   multi variant cox regression analysis. HR > 1 demonstrates that the
   element is risk factor. H C-index value estimates the probability that
   the predicted results are consistent with the actually observed
   results. In this diagram, our signature has the best performance
   After calculating the risk score of each patient, groups A and B have
   notably different risk scores, while the differences among C1, C2, and
   C3 are not that prominent (Fig. [104]4C, D). The low and high risk
   score groups have remarkably different m7G-related gene expression
   levels (Fig. [105]4E). Univariate Cox regression indicated that age and
   risk score are relevant to prognosis (Fig. [106]4F). Multivariate Cox
   regression further confirmed that these three elements are independent
   prognostic factors (Fig. [107]4G). The concordance index comparison of
   each element illustrates that our signature has the best predictive
   accuracy compared with age and gender (Fig. [108]4H). These results
   initially verify the accuracy and feasibility of the signature.
Validate of the signature at the clinical level
   To further identify the applicability of the signature in clinical
   characteristics, we tested and verified our signature using clinical
   indexes. First, we performed survival analysis between the high and low
   risk score groups with respect to gender, age, isocitrate dehydrogenase
   (IDH) status, RPS type, race, and tumor status using all patient data.
   The results confirm that patients with different gender, age, IDH
   status, and RPS type have varied survival times, while race and tumor
   status do not influence survival (Fig. S[109]2). Next, in the training
   group, the survival differences between the high and low risk score
   groups are significant (Fig. [110]5A). Additionally, patients with low
   risk scores are clearly more likely to survive and have higher
   expression levels of TMOD2 and CACNG2, but lower expression levels of
   PLOD3 and TMSB10 (Fig. [111]5B). These results are consistent with the
   former verification and risk score algorithm. The ROC curve
   demonstrated the predictive ability (Fig. [112]5C) of this approach.
   The same analysis was conducted in the test group (Fig. [113]5D-F) and
   the whole dataset (Fig. [114]5G-I), and the results were consistent
   with those of the training group.
Fig. 5.
   [115]Fig. 5
   [116]Open in a new tab
   Signature consolidation through clinical information. A Survival
   analysis of high and low risk score group in train session. B Risk
   curve, survival status and gene expression of each patient in train
   session. C Receiver operating characteristic (ROC) curve in order to
   testify the prediction ability. Area under curve (AUC) > 0.7 is
   considered as ideal state. D-F Same analysis in test session. G-I Same
   analysis utilizing all patients’ information
   After validating the m7G signature, another forecast pattern
   reconciling the risk score and clinical features, such as age, was
   generated. The nomogram vividly displays the weight of each factor and
   prognostic indication of the different scores (Fig. [117]6A). The
   calibration curve indicates the unity of the observed and predicted 1-,
   3-, and 5-year survival rates (Fig. [118]6B). According to 659 GBM
   samples, we divided the new risk value of the Nomogram into high and
   low groups, as is shown in Fig. [119]6C. Compared with the low-risk
   group (n = 329), the OS of the high-risk group (n = 330) was
   significantly lower, in which HR = 3.07 (2.54–3.70), p < 0.001. The ROC
   curve of 1-year prediction shows that the AUC of the risk signature,
   nomogram model, and age are all > 0.6, representing the effectiveness
   of prediction (Fig. [120]6D). Using the DCA dimension reduction method,
   the predictive efficiency is well exhibited (Fig. [121]6E). The same
   analysis was performed to analyze the 3-year (Fig. [122]6F, G) and
   5-year (Fig. [123]6H, I) prediction results. All of the results
   described above show the effectiveness of the nomogram model.
Fig. 6.
   [124]Fig. 6
   [125]Open in a new tab
   Establishment of nomogram and its forecast performance. A A putative
   displaying pattern of influence factors and weight of clinical model. B
   Calibration curves of nomogram to ascertain the prediction. C Overall
   survival of Nomogram model. D Receiver operating characteristic (ROC)
   curve shows the 1-year forecast probability through m7G score
   signature, nomogram, age and gender separately in detail. E Decision
   curve analysis (DCA) dimensionality reduction method to illustrate the
   accuracy of forecast. F, G Same analysis in 3-years prediction. H, I
   Same analysis in 5-years prediction
Broad relevance to immune cells, TME, tumor mutational burden (TMB), and stem
cells
   From the former analysis, we became interested in the relationship
   between m7G and the TME. The heat map displays that group C3 has higher
   stromal scores and immune scores, indicating that group C3 has an
   abundance of stromal and immune cells. There are relatively fewer tumor
   cells present, thus resulting in lower tumor purity scores
   (Fig. [126]7A). The definite value is demonstrated in Fig. [127]7B.
   Furthermore, we explored the intersection of immune cells. The
   correlations between the genes of the signature and immune cells were
   analyzed using the cibersort algorithm and is displayed in a heat map
   (Fig. [128]7C). The relevance of the risk score and immune cells via
   different calculation methods is exhibited in Fig. [129]7D. The risk
   score is positively related to immune checkpoint inhibitor genes, most
   notable of which are PDCD1 (PD1) and CD274 (PD-L1) (Fig. [130]7E).
Fig. 7.
   [131]Fig. 7
   [132]Open in a new tab
   The relationship of signature and TME. A Single-sample gene-set
   enrichment analysis (ssGSEA) measured tumor miccroenvironment (TME)
   score and content of immune cells in each sample. B TME score in high
   and low immune group displayed comparatively. C Relevance of genes and
   immune cells using cibersort algorithm. D The relationship of immune
   cells and high and low risk score through 7 different algorithms. E The
   expression relationship of risk score and immune checkpoint inhibitor
   (ICI)
   We also investigated the relationship between the tumor mutational
   burden (TMB) and risk score. The results indicate that the low risk
   score group has lower mutation percentages in many genes, especially
   TP53, TTN, and NF1, with 5, 11, and 8% respective decreases compared
   with the high risk score group (Fig. [133]8A, B). Then, patients were
   divided into high mutation and low mutation groups. Survival analysis
   suggested a meaningful difference between these two groups (Fig.
   [134]8C). However, when mutation risks were reconciled with risk score,
   the differences of survival became vague (Fig. [135]8D). In addition,
   we determined that the stem cell index is negatively related to risk
   score (Fig. [136]8E), suggesting a correlation between glioblastoma
   stem cells (GSCs) and m7G, and implying a research direction for future
   in-depth investigation.
Fig. 8.
   [137]Fig. 8
   [138]Open in a new tab
   The influence of risk score clustering on TMB. A, B TMB in low and high
   risk score group. C Survival forecast analysis compared high-TMB group
   with low-TMB group. D Merging comparison of risk score group and TMB
   group. (E) Relevance of steam cell index and risk score of the
   signature
Comparison of m7G and other GBM related sinatures
   Four signatures about GBM which were published within three years were
   selected to compare to our ones. The results were shown in
   Fig. [139]9A, B. The C-index value of the signature of GBM related
   genes in the manuscript is the highest, which is 0.65, also, the RMS
   value is the smallest (HR: 1.554, p < 0.001), representing low
   dispersion and high reliability.
Fig. 9.
   [140]Fig. 9
   [141]Open in a new tab
   Signature comparison. A Comparison of the C-index value between m7G and
   four individual signatures. B Root Mean Square (RMS) values among five
   signatures
Down-regulation of PLOD3 restrained proliferation and migration abilities of
glioma cells
   To reveal malignant behaviors of the hub gene modulating m7G
   modification patterns of glioma, we first validation biological
   behaviors of down-regulated expression of hub gene, PLOD3, in LN18 and
   T98G cells (Fig. [142]10A, S[143]3, S[144]4). According to the results
   of the CCK-8 assay, down-regulated level of PLOD3 expression
   significantly restrained the proliferation ability of glioma cells
   compared with control group (Fig. [145]10B). Transwell cell migration
   assay also indicated that the down-regulation level of PLOD3 expression
   significantly inhibited the metastasis ability of glioma cells compared
   with control group (Fig. [146]10C, D). Overall, the down-regulation of
   PLOD3, the hub gene modulating m7G modification patterns, significantly
   suppressed proliferation and migration capacity of glioma cells.
Fig. 10.
   [147]Fig. 10
   [148]Open in a new tab
   Experimental verification of PLOD3. A Biological behaviors of
   down-regulated expression of PLOD3 in LN18 and T98G cells. B Results of
   the CCK-8 assay of the influence of the PLOD3 expression on glioma
   cells. C, D Outcomes of the Transwell cell migration assay of PLOD3
Discussion
   Although emerging evidence has demonstrated the potential role of m7G
   in cancer and tumorigenesis, research on m7G in cancer is still
   relatively insufficient. In this study, we first displayed the overall
   mutation profile of m7G-related genes in glioblastoma samples using
   array data from the TCGA, CGGA, and GEO public datasets. Notably, a
   novel detection method called m7G Mutational Profiling sequencing
   (m7G-MaP-seq) has already been discovered to detect internal m7G
   modifications [[149]26]. Although the methodology is different, the
   purpose of this technique is also to further investigate the role of
   mutation modification patterns in disease progression. This emphasizes
   the goal of our research from another perspective. The grouping process
   is guided by the NMF algorithm, indicating the most meaningful and
   distinctive cluster method. The construction of a prognostic signature
   showed remarkable significance and can act as an independent prognostic
   factor with a strong predictive effect. The cut off value of the risk
   score was 1.1168, and proved to be significant, as the survival
   analysis, TMB, and TME analyses were all statistically different.
   Glioblastoma is a focus of many researchers because of its high
   recurrence rate and poor treatment effects. According to the WHO
   classification standard updated in 2016, which is based on the mutation
   status of the IDH 1 or 2 genes, wild-type genotypes accounted for more
   than 90% of cases [[150]27]. This typing method has strong prognostic
   significance in this disease. For treatment methods, Han [[151]28]
   summarized new molecular therapies that target mutant IDH, while
   therapies for wild-type genotypes need additional research. m7G has
   recently become a research hotspot, with many researchers becoming
   particularly interested in its role in tumorigenesis. m7G is a form of
   RNA methylation, in addition to N6-methyladenosine (m6A),
   2-O-dimethyladenosine (m6Am), N1-methyladenosine (m1A), and
   5-methylcytosine (m5C) [[152]29]. Numerous studies have confirmed that
   m7G cap is a unique molecular module that can recruit cellular proteins
   and mediates cap-related biological functions, such as pre-mRNA
   processing, nuclear output, and cap-dependent protein synthesis
   [[153]30]. Some articles have shown that RNA modifications are involved
   in cancer development and progression, including one by Barbieri
   [[154]31]. However, the specific role of m7G in tumorigenesis is
   remains understudied. Therefore, we conducted a detailed analysis on
   the role of m7G in glioblastoma to address this gap in knowledge.
   The scoring system composed of a m7G prognostic signature and grouping
   boundary value proved to be effective in many aspects. Tmod2 regulates
   the stability of F-actin and dendrite developing during dendrization
   and synaptic formation. These findings provide new insights into the
   actin regulatory mechanisms of neuronal development, revealing
   potential pathways that are disrupted in many neurological disorders
   [[155]32]. CACNG2 can affect the susceptibility to postoperative
   chronic pain [[156]33] or chronic pain caused by nerve injury
   [[157]34]. Existing research shows that elevated expression levels of
   PLOD3 can accelerate tumor progression and indicate poor prognosis
   [[158]35], which is consistent with our signature where the weight of
   PLOD3 was positive. Although the role of TMSB10 in glioblastoma has not
   been studied, existing publications have shown that TMSB10, as a
   cancer-promoting gene, can promote cell invasion and cancer
   progression. This has been demonstrated in gastric cancer [[159]36],
   renal clear cell carcinoma [[160]37], and lung cancer [[161]38]. The
   abovementioned results support the complex relationship between these
   fours genes and cancer, which is quantified by the risk score
   calculation formula in glioblastoma. Our correlation analysis of
   immunity lays a foundation for the exploration of immunotherapy in
   glioblastoma. Subsequent analysis also verified the relationship
   between m7G and the TME and TMB. Glioblastoma shows significant
   cellular heterogeneity, among which stem-like GSCs was the most
   significant [[162]39]. There is increasing evidence that GSCs play an
   important role in tumor growth and treatment responses [[163]40].
   Therefore, we studied the correlation between glioblastoma and stem
   cells.
   Based on the public database, four novel genes were discovered that are
   likely to be related to m7G. They are likely to affect the development
   and prognosis of GBM through corresponding pathways. We selected PLOD3
   with the largest risk coefficient for experimental verification. As we
   guessed, the down-regulation of PLOD3 significantly inhibited the
   proliferation and migration of glioma cells. Our research strongly
   addresses the current gap in the m7G and glioblastoma research fields,
   utilizes a macroanalysis of the phenotypes of m7G-related genes in
   glioblastoma, establishes a prognosis evaluation system, and quantifies
   the impact of m7G on glioblastoma at the micro level. Overall, our work
   lays a solid foundation for future research.
Supplementary Information
   [164]Additional file 1. ^(9.2KB, xlsx)
   [165]Additional file 2. ^(135.6KB, xlsx)
   [166]Additional file 3. ^(67.9KB, xlsx)
   [167]Additional file 4. ^(271.1KB, xlsx)
   [168]Additional file 5. ^(29.4KB, xlsx)
   [169]Additional file 6. ^(11.4KB, xlsx)
   [170]Additional file 7. ^(1.9MB, tif)
   [171]Additional file 8. ^(1.2MB, tif)
   [172]Additional file 9. ^(361.1KB, tif)
   [173]Additional file 10. ^(361.1KB, tif)
Acknowledgements