Next Article in Journal
RETRACTED: Sudha et al. Triazole Modified Tetraiodothyroacetic Acid Conjugated to Polyethylene Glycol, a Thyrointegrin αvβ3 Antagonist as a Radio- and Chemo-Sensitizer in Pancreatic Cancer. Biomedicines 2022, 10, 795
Previous Article in Journal
Endothelial Dysfunction: From Pathophysiology to Novel Therapeutic Approaches 2.0
Previous Article in Special Issue
Could let-7f, miR-10b, miR-34a, miR-181b, and miR-181d Be Useful Tools as a Target Therapy for Uterine Leiomyosarcoma?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Prognostic Neuromodulation-Related Gene Signature Identifies Immunomodulation and Tumour-Associated Hallmarks in Glioblastoma

by
Min Yee Chow
1,†,
Sylvia Sue Xian Liew
1,†,
Mastura Monif
2,3,
Muhamad Noor Alfarizal Kamarudin
1,* and
Brandon Wee Siang Phon
1,*,‡
1
Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
2
Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
3
Department of MS and Neuroimmunology, Alfred Health, Melbourne, VIC 3004, Australia
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Current address: Department of Urology, New York University Langone Health, 550 1st Ave, New York, NY 10016, USA.
Biomedicines 2025, 13(11), 2640; https://doi.org/10.3390/biomedicines13112640
Submission received: 24 July 2025 / Revised: 8 October 2025 / Accepted: 14 October 2025 / Published: 28 October 2025
(This article belongs to the Special Issue Epigenetic Regulation in Cancer Progression)

Abstract

Background and Objective: Neuromodulators such as neuropeptide, neurotrophic factors and neurotransmitters are increasingly reported to be involved in glioblastoma (GBM) progression. Nonetheless, the association between neuromodulation-related genes (NMRGs) and GBM prognosis remains elusive. Hence, this study aims to identify clinically significant NMRGs that can form a prognostic gene signature for GBM patients. Methods and Results: Differential expression analysis of transcriptomic profiles extracted from GSE147352, GSE165595, TCGA and CGGA determined 272 differentially expressed NMRGs (deNMRGs) in GBM compared to normal brain tissue. The subsequent Kaplan–Meier survival analysis and Cox proportional hazard model further identified ten common deNMRGs (IGF2, RETN, EDNRB, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7), forming a 10-NMRG signature. This signature stratifies GBM patients and consistently predicts poorer survival outcomes for the high-risk score group compared to the low-risk score group in the TCGA and CGGA cohorts. The gene set enrichment analysis and active-subnetwork-oriented enrichment analysis identified a connection between immunomodulation and tumour-associated hallmarks with the high-risk GBM patient group. Next, the correlation proportionality analysis identified a positive association between the signature genes with immune activators, immune suppressors and pro-motility genes. Additionally, high expressions of the 10-NMRGs were noted in the mesenchymal GBM subtype. Conclusions: Collectively, our analysis highlights the potential use of the 10-NMRG signature to stratify the high-risk GBM group with a strong association of immunomodulation and tumour-associated hallmarks that can contribute to the poor survival outcomes.

1. Introduction

The glioblastoma IDH-wildtype (GBM) is the most common and aggressive form of brain malignancy, accounting for about 50.9% of all malignant brain tumours [1]. Despite advances in molecular diagnostics and therapeutic efforts for GBM, the median survival of GBM patients has barely improved over the last decade, with only a relative estimate of 6.8% of patients surviving five years post-diagnosis [2]. This dismal prognosis underscores the critical need for continued research to develop novel prognostic tools for improved treatment stratification and patient outcomes.
A growing body of evidence has demonstrated that regulators of neuronal functions and synaptic transmission (neuropeptides, neurotrophic factors, neurotransmitters and their associated receptors) play vital roles in GBM tumorigenesis and progression. Notably, the activation of tachykinin NK1 receptors by the neuropeptide substance P induces cytokine production and promotes tumour growth [3]. Similarly, the specific activation of neurotrophic factors, receptors TrkB and TrkC by their ligands neurotrophin 3 (NT3) and brain-derived neurotrophic factor (BDNF) via Akt and ERK pathways, promotes the survival of brain tumour-initiating cells [4]. Furthermore, signalling by neurotransmitters such as acetylcholine [5,6] and glutamate [7,8] has been shown to regulate GBM tumour growth, proliferation, survival, migration, and invasion. Collectively, these findings highlight the critical roles of neuromodulator signalling in GBM pathogenesis, prompting the urgent need to elucidate its clinical potential to enhance diagnostic accuracy and treatment outcomes in GBM patients.
While previous studies have shown that neuromodulators contribute to GBM aggressiveness, their potentials as prognostic markers and therapeutic targets are still not well understood. Recently, the construction of a predictive prognostic assessment model with combined multiple related genes has provided a significant improvement in prediction accuracy, compared to a single gene prognostic marker. In this study, we sought to identify clinically significant neuromodulation-related genes (NMRGs) that could serve as a prognostic signature for GBM patients. The differentially expressed NMRGs (deNMRGs) were systematically screened from The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA), and Gene Expression Omnibus (GEO) databases. The common prognostic deNMRGs were integrated to form a predictive model consisting of a 10-neuromodulation-related gene (10-NMRG) signature. Findings from this study showed strong associations of these genes with immunomodulation and tumour-associated hallmarks, contributing to poor survival outcomes in GBM patients.

2. Materials and Methods

The study workflow is summarised in Figure 1. The fifth edition of the WHO Classification of the Tumour of the Central Nervous System (CNS) released in 2021 (WHO CNS5) has refined GBM as a Grade 4 astrocytic glioma with wildtype IDH [9], while diffuse gliomas with IDH mutation are defined as lower grade glioma. Hence, for accuracy, only GBM patients with IDH-wildtype were included in this study. Their clinicopathological data are summarised in Table S1, Supplementary File S1.

2.1. Human Neuromodulation-Related Gene Set

A human neuromodulation-related gene set (n = 420) comprising (1) neuropeptides (n = 97), (2) neuropeptide receptors (n = 97) (Table S2, Supplementary File S1), (3) neurotrophic factors (n = 26), (4) neurotrophic factor receptors (n = 22) (Table S3, Supplementary File S1), (5) neurotransmitter receptors (n = 131) (Table S4, Supplementary File S1) and (6) neurotransmitter system-related genes (n = 47) (Table S5, Supplementary File S1) was established.

2.2. RNA-Seq Data Acquisition

Four RNA-Seq expression datasets containing IDH1R132-wildtype GBM patients and normal brain samples were obtained. Two RNA-Seq expression datasets with their respective datasets’ clinical information were downloaded from GEO (https://www.ncbi.nlm.nih.gov/geo/) (accessed on 2 July 2023). Huang et al. graciously provided the raw expression count for the dataset GSE147352, which was directly obtained from GEO [10]. For GSE165595, that encompasses matched GBM and brain samples [11], RNA-Seq fastq files were obtained using SRA Explorer instead, where paired-end alignments and transcript quantifications were performed in-house. STAR [12] was leveraged to align the transcripts with Ensembl’s reference human genome (GRCh38 primary assembly), while gene quantifications were performed with the help of RSEM [13], using ENCODE3’s STAR-RSEM pipeline parameters. Exact linux scripts and parameters used are provided.
Additionally, level 3 gene expression profiles of GBM patients from TCGA and healthy brain samples from TCGA (5 samples) and GTEx (206 samples from the cortex and frontal cortex region), specifically Illumina Hiseq2000 RNASeq log2 transformed HTSeq counts, were obtained from the UCSC Xena data portal (https://xenabrowser.net/datapages/) [14] (accessed on 2 July 2023). Clinical data of the GBM patients containing information about the IDH1R132 mutation status, molecular subtypes, survival times and overall survival outcomes were obtained from cBioPortal (https://www.cbioportal.org) [15,16,17] (accessed on 2 July 2023). The utilisation of the Toil pipeline allowed for a unified processing workflow between the TCGA and GTEx datasets, with STAR being used to generate alignments and quantifications being performed using RSEM [18]. The recomputation of the raw RNA-Seq data from TCGA and GTEx by the UCSC Xena project makes the two datasets compatible, allowing for the direct expression analyses. Following the acquisition of expression data from the TCGA/GTEx cohort, RNA-Seq expression counts were also obtained from CGGA (http://www.cgga.org.cn/download.jsp) [19,20,21,22] (accessed on 3 July 2023). The Illumina HiSeq expression counts and the corresponding clinical information for batch mRNAseq_325 were obtained. Table 1 summarises the number of GBM patients and normal brain samples in each RNA-Seq cohort.

2.3. Differential Expression Analysis

Differential expression analysis was performed in R v4.3.2, using the package ‘DESeq2’ (v1.48.2) [23]. |Log2 fold change (LFC)| > 0.6 and Benjamini–Hochberg adjusted p-values (padj) < 0.05 were set as the cut-offs for the screening of differentially expressed genes (DEGs). To reduce the problem of exaggerated LFCs for genes with low counts, LFC shrinkage was performed using the estimator ‘apeglm’ [24]. Additionally, gene expression between different GBM subtypes was also analysed and plotted using the ‘plotCounts’ function from DESeq2. All R scripts used for differential expression analysis and subsequent analyses are provided.

2.4. Survival Analysis

Survival analysis was performed on both the TCGA and CGGA cohorts using a Kaplan–Meier curve and the Cox proportional hazard model. The analyses were executed in R v4.3.2 using the ‘survival’ (v3.8-3) [25] and ‘survminer’ (v0.5.1) [26] packages. Patients were dichotomised into low- and high-expression groups, using an optimal cut-off value computed by the ‘maxstat’ (v0.7-26) package [27]. This method evaluates all possible thresholds and selects the one that provides the greatest separation of survival outcomes based on log-rank statistics, ensuring that the chosen cut-off is both statistically and clinically meaningful. The cut-off value for each gene can be found in Tables S6–S9, Supplementary File S1.
Transcript per million (TPM) values were used for the TCGA dataset, while variance-stabilised transformed (VST) values were used for the CGGA dataset. The overall survival probabilities of the query genes were evaluated using Kaplan–Meier curves (log-rank p-values ≤ 0.05 denotes significance), while their prognostic values were assessed using the Cox proportional hazard model by calculating the hazard ratio (HR). Hazard ratios (HR) < 1 indicated favourable prognostic markers and HR > 1 indicated adverse prognostic markers (p < 0.05). Genes meeting these significance thresholds in both analyses were considered prognostic. The prognostic value of each NMRG was evaluated independently in the TCGA and CGGA cohorts, and only those showing consistent associations across both datasets were retained to construct the 10-NMRG signature.

2.5. Computation of the 10-NMRG Signature Prognostic Index

A total of 10 NMRGs were obtained from the survival analysis to compute the prognostic index, using the formula: i E x p r e s s i o n G e n e i   G e n e   C o e f f i c i e n t i . The TCGA cohort was randomly divided into a training set (n = 83) and a validation set (n = 57) to enable independent evaluation of the risk model’s performance prior to validation in the complete TCGA and CGGA cohorts. The prognostic index was used to stratify GBM patients into high- and low-risk score groups based on the log-rank test. The survival probability between the two groups was then estimated using Kaplan–Meier plots for both the training and validation sets. The predictive accuracy of the gene signature was assessed using time-dependent ROC curve analyses, via the survivalROC (v1.0.3.1) R package. The prognostic performance of the 10-NMRG signature was assessed using the multivariate Cox regression analysis in the complete TCGA and CGGA cohorts. To account for potential clinical confounders, the available covariates, including age, gender, molecular subtype, chemotherapy, radiotherapy, and MGMT methylation status, were incorporated into the analysis.

2.6. Gene Set Enrichment Analysis (GSEA)

GSEA was performed among TCGA GBM patients that were stratified into high- and low-risk score groups by the 10-NMRG prognostic index [28]. The median of ratios normalised counts from DESeq2 were input into the GSEA v4.3.2 software, with the Molecular Signatures Database (MSigDB), hallmark gene sets (H) and curated gene sets (C2) KEGG_MEDICUS and Reactome subset of canonical pathways, being used for the analysis [29]. The permutation type was based on phenotype, with a total of 1000 iterations. A gene set was considered significantly enriched when the nominal (NOM) p-value < 0.01 and the false discovery rate (FDR) q-value < 0.05.

2.7. Active-Subnetwork-Oriented Enrichment Analysis (ASOEA)

ASOEA was performed using DEGs from the high- and low-risk score groups of the TCGA GBM cohort, as stratified by the 10-NMRG signature prognostic index. This was made possible using the R package ‘pathfindR’ (v2.6.0) (https://github.com/egeulgen/pathfindR) (accessed on 4 September 2023) [30]. Enrichment analysis performed through ‘pathfindR’ integrates the differential expression gene information to produce interaction information through protein–protein interaction networks via active subnetwork identification, which are subsequently fed into pathway/gene set annotation enrichment over 10 iterations.

2.8. Compositional Proportionality Analysis

Proportionality analyses between the ten genes of interest and various genes involved in GBM progression were carried out using the ‘propr’ package (v5.1.7) in R v4.3.2 (https://github.com/tpq/propr) (accessed on 11 September 2023) [31] on the raw expression count for GBM patients from TCGA. To overcome compositional data analyses’ inability to compute for zeros, all zeros were replaced by a Bayesian multiplicative replacement strategy that preserves the ratios between the non-zero components [32] using the zCompositions package (v1.5.0-5; cmultRepl function) [33]. Feature coordination between the various genes was calculated using the “ρ” measure. p-values are not calculated for proportionality; instead, the FDR was permuted for a given cut-off. To achieve an FDR of below 5%, we selected a cut-off of Ρρ > 0.25. The proportionality matrices were illustrated using the ‘corrplot’ (v0.95) package.

3. Results

3.1. Identification of deNMRGs in GBM Patient Samples

To identify neuromodulation-related genes (NMRGs) potentially implicated in GBM pathogenesis, differential expression analyses between GBM and normal brain tissues were independently performed across four RNA-Seq datasets (TCGA, CGGA, GSE147352 and GSE165595), encompassing a total of 300 IDH-wildtype GBM samples. To ensure consistent dysregulation of the identified differentially expressed NMRGs (deNMRGs), only genes exhibiting concordant expression trends in at least two datasets were retained. This selection approach accounted for variations in sample size (GBM vs. normal brain) and sequencing platforms across datasets, ensuring analytical robustness without excluding biologically relevant deNMRGs associated with GBM. Among the 420 screened NMRGs, 272 deNMRGs were identified, with 86 upregulated and 186 downregulated in the GBM samples. The distributions of deNMRGs by their neuromodulation category is detailed in Table 2, with the complete deNMRGs lists provided in Tables S10 and S11, Supplementary File S1.

3.2. IGF2, RETN, EDNRB, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7 Are the Common Prognostic Genes in TCGA and CGGA GBM Cohorts

The prognostic values of identified deNMRGs were evaluated using Kaplan–Meier survival and univariate Cox regression analyses in the TCGA and CGGA GBM cohorts. The analysis revealed that among the 86 upregulated and 186 downregulated NMRGs, 18 upregulated and 42 downregulated genes in TCGA, along with 32 upregulated and 51 downregulated genes in CGGA, exhibited significant prognostic value for overall survival (OS) (Tables S6–S9, Supplementary File S1). In addition, comparison of prognostic deNMRGs between the TCGA and CGGA cohorts identified ten deNMRGs (IGF2, RETN, EDNRB, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3, HTR7) that consistently predicted for similar survival outcomes in both cohorts (Figure 2). In the TCGA and CGGA cohorts, GBM with high expressions of IGF2 (HR = 1.57; 1.85), RETN (2.24; 2.13), C3AR1 (1.83; 2.16), CLCF1 (1.76; 2.44), NTRK1 (1.58; 2.50), OSMR (3.72; 2.37), KCNN4 (2.36; 2.62), SLC18A3 (1.88; 1.87) or HTR7 (1.74; 1.79) had significant shorter OS, indicating predictions for a worse prognosis. Meanwhile, high EDNRB expression was associated with longer OS (HR = 0.579; 0.420) and better outcomes in the TCCGA and CGGA cohorts (Figure 3 and Figure 4).

3.3. A 10-NMRG Signature Forms a Risk Score Model That Predicts Prognosis in TCGA and CGGA GBM Cohorts

A 10-NMRG signature comprising IGF2, RETN, EDNRB, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7 was constructed. By splitting the TCGA dataset into a training (n = 83) and a validation set (n = 57), the predictive performance of this ten-gene signature in TCGA dataset was evaluated using Kaplan–Meier survival analysis. Both sets showed similar results, where high-risk score GBM patients showed decreased OS (Figure 5A,C). The prognostic performance of the 10-NMRG signature in evaluating 1-, 2- and 3-year OS rates were also favourable, where the time-dependent ROC analysis (Figure 5B,D) returned AUC values above 0.7 across both datasets.
The observation was similar when the 10-NMRG signature was evaluated on the entire TCGA GBM dataset, where high-risk score GBM had approximately 4.5 months shorter OS (Figure 6A). The accuracy of the gene signature in predicting OS was further validated in the CGGA cohort, which consisted of a different population. Similarly to the TCGA cohort, the CGGA high-risk score GBM had approximately 16 months shorter OS (Figure 6C). Lastly, multivariate Cox regression analysis (Table 3) showed that the gene signature can act as an independent prognostic factor among common GBM prognostic factors (age, gender, subtypes, chemo- and radio-therapy statuses and MGMT methylation status) in both cohorts.

3.4. GBM with High 10-NMRG Signature Risk Score Is Associated with Immunomodulation and Tumour-Associated Hallmarks

To understand the molecular features of high-risk score GBM stratified by the 10-NMRG signature, GSEA was conducted to analyse the transcriptomic profiles of a GBM tumour in the TCGA cohort. GSEA using the MSigDB hallmark gene sets revealed significant gene enrichment associated with immunomodulation (TNFα signalling via NF-κB, IL6-JAK-STAT3 signalling, inflammatory response, complement, IL2-STAT5 signalling) and tumour-associated (epithelial–mesenchymal transition, hypoxia, KRAS signalling activation, apical junction, early response to oestrogen, angiogenesis) hallmarks in high-risk score GBM. Furthermore, GSEA using KEGG MEDICUS and Reactome pathway gene sets also identified enrichment of the pro-inflammatory immune response (IL6, IL2 and hormone-like cytokines-family-mediated JAK-STAT signalling pathways, TNF receptor superfamily TNFSF members mediating non-canonical NF-κB pathway and complement cascade), anti-inflammatory immune response (IL4 and IL13 signalling), integrins-mediated cell–matrix interaction and cytoskeleton remodelling (ITGA/B-RHOGAP or RHOGEF- modulated RHOA signalling, ITGA/B-FAK-modulated RAC and CDC42 signalling and ITGA/B-talin-vinculin signalling) and extracellular matrix (ECM) remodelling (degradation of ECM, activation of matrix metalloproteinase (MMPs) and collagen degradation) gene sets in high-risk score GBM (Table 4; Figures S1–S3, Supplementary File S2).
Next, DEGs were identified in high-risk against low-risk score GBM from TCGA cohort. Using pathfindR, ASOEA was conducted to identify gene subsets, which form functional protein interactions in mediating immunomodulation and tumour-associated pathways in high-risk score GBM. Significant upregulation of interleukin, C-C motif chemokine ligand (CCL), C-X-C motif chemokine ligand (CXCL) and complement system genes were observed in the high-risk group (Table S12, Supplementary File S1). These genes were significantly enriched in immunomodulation-related GO biological process terms, including positive regulation of cytokine, IL-6, IL-2 and TNF production, positive regulation of NF-kB signalling and neutrophil chemotaxis (Table 5). In addition, the high-risk score GBM also showed significantly higher expressions of integrins, EMT transcription factors, ECM components and matrix proteases genes, which regulate GO biological processes related to cell adhesion, cell migration, cell polarity, cytoskeletal dynamics, ECM remodelling and angiogenesis (Table 5; Table S12, Supplementary File S1).
Moreover, KEGG and Reactome pathway enrichment analyses further supported that upregulated genes in the high-risk GBM formed functional networks modulating three key categories: (i) immunomodulatory pathways (e.g., JAK-STAT, chemokine and TNF signalling pathway), (ii) oncogenic signalling pathways (e.g., oestrogen, Rap1, Ras signalling pathways) and (iii) cell–matrix interactions-related processes (e.g., ECM-receptor interaction, cell surface, laminin interactions, focal adhesion, regulation of actin cytoskeleton, degradation of ECM) (Table 5; Table S12, Supplementary File S1).

3.5. C3AR1, CLCF1, OSMR, KCNN4 and HTR7 Are Positively Correlated with Immune Activator and Immune Suppressor Genes in GBM

Next, compositional proportionality analysis revealed relationships between the 10-NMRGs and genes involved in immune responses, EMT, cell–ECM interactions, ECM dynamics, and angiogenesis in GBM. Among the 10-NMRGs, C3AR1, CLCF1, OSMR, KCNN4 and HTR7 showed strong positive proportionality with many immune activator (Figure 7A) and suppressor genes (Figure 7B), while moderate positive correlations were observed with the cell–ECM adhesion genes, especially integrins (Figure 7C). In contrast, C3AR1, CLCF1, OSMR and KCNN4 showed weak positive proportionality with EMT regulators, matrix proteases and angiogenesis genes (Figure 7D–F).

3.6. GBM Mesenchymal Subtype Showed High Expressions of RETN, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, and HTR7 Compared to Classical and Proneural Subtypes

Comparison of the 10-NMRGs expression patterns across GBM subtypes revealed significant differences in their expression levels among the proneural, classical and mesenchymal subtypes. Notably, the mesenchymal subtype showed significantly higher expressions of RETN, C3AR1, CLCF1, NTRK1, KCNN4 and HTR7 compared to proneural and classical subtypes (all p < 0.0001, except RETN mesenchymal vs. proneural p < 0.001), while OSMR is significantly elevated in mesenchymal compared to the proneural subtype (p < 0.0001). Meanwhile, EDNRB was significantly downregulated in the mesenchymal versus the proneural (p < 0.01) and classical (p < 0.0001) subtypes (Figure 8).

4. Discussion

GBM tumours co-opt normal neuromodulatory pathways by aberrantly expressing neuromodulators, such as neuropeptides, neurotrophic factors, neurotransmitters and their cognate receptors, to drive progression [34,35]. While several neuromodulators are known to regulate GBM behaviour, the roles of many other neuromodulators remain unclear. In this study, systematic analysis of GBM gene expression profiles and clinical data identified a 10-NMRG signature that stratifies high-risk tumours and predicts poorer survival among GBM patients, through associations with immunomodulation and tumour-associated hallmarks.
In both the TCGA and CGGA GBM cohorts, high expressions of IGF2, RETN, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7 predicted poorer prognoses, while EDNRB was correlated with a better outcome (Figure 3 and Figure 4). The resulting 10-NMRG signature effectively stratified the TCGA GBM patients into low- and high-risk score groups with distinct survival outcomes. This predictive model was validated in the CGGA cohort, demonstrating its robustness across Caucasian and Asian populations (Figure 6). When analysed in relation to different ages, sex, subtypes, treatment received and MGMT methylation status, the 10-NMRG signature was able to predict survival independently in both cohorts, signifying its potential as a feature-agnostic signature (Table 3).
Among the ten genes, RETN [36,37], C3AR1 [38,39], CLCF1 [40], NTRK1 [41,42], OSMR [43], KCNN4 [44,45] and HTR7 [46] have established roles in cancer pathogenesis through regulating tumour growth, motility, angiogenesis, chemoresistance and immune evasion. In GBM, preclinical studies have demonstrated the oncogenic roles of IGF2 [47], EDNRB [48,49], C3AR1 [50,51], CLCF1 [52], NTRK1 [42,53], OSMR [54,55,56,57], KCNN4 [58,59] and SLC18A3 [6] in promoting proliferation, migration, invasion, EMT, chemoresistance, stemness and survival (Table 6). The roles of the remaining genes in GBM pathogenesis remain unreported. GSEA revealed that GBM tumours in the high-risk score group are associated with immunomodulation (TNFα signalling via NF-kB, IL6-JAK-STAT3 signalling, inflammatory response, complement, IL2-STAT5 signalling) and tumour-associated hallmarks (EMT, KRAS signalling activation, early response to oestrogen, angiogenesis) (Table 4; Figure S1, Supplementary File S2). These findings suggested the plausible molecular mechanisms linking the 10-NMRG high-risk score group to poorer prognosis in GBM.
The GBM tumour microenvironment is shaped by dynamic tumour–immune interactions that promote tumour progression through the complex interplay of inflammation and immune suppression [61,62,63]. The release of pro-inflammatory chemokines and cytokines, such as IL-1, IL-2, IL-6, CCL-2, CCL-8, TNF-α and TGF-β, facilitate the recruitment of brain-resident immune cells to the tumour site [64]. These factors not only promote GBM growth but also activate JAK-STAT and NF-κB pathways, which upregulate immune checkpoint molecules and immunosuppressive cytokines within the tumour microenvironment [64,65]. Cytokines including IL-6, IL-10, IL-11 and IL-24 can suppress immune effector cells, such as T cells, natural killer cells and dendritic cells, impairing the host’s anti-tumour response while facilitating the immune evasion of GBM cells [61,62,63]. Consistent with the existing literature, our analysis revealed that the high-risk score group expressed significantly higher levels of interleukins, chemokines and complement system genes involved in modulating pro-inflammatory (IL-2, IL-6, TNFα signalling pathways) and anti-inflammatory (IL-4, IL-6 and IL-13 signalling pathways) immune response, complement cascades activation, JAK-STAT and NF-κB pathways (Table 4 and Table 5; Table S12, Supplementary File S1).
Moreover, a strong positive correlation was observed between the expression of C3AR1, CLCF1, OSMR, KCNN4 and HTR7 genes with the immune activator (e.g., IL1A, IL15, CCL2) and suppressor (e.g., TGFB1, IL6, IL10) genes (Figure 7A,B), suggesting a possible co-expression and/or co-regulation of these five genes with the immune-related genes in GBM. To date, the understanding of the roles of NMRGs in immunomodulation remains limited, with preliminary preclinical findings suggesting the involvement of EDNRB [49] and C3AR1 [51] in modulating immune escape, and immune cells infiltration to tumour microenvironment in GBM. The strong association of heightened immune and inflammatory responses within the tumour site in the high-risk score GBM, shown in our analysis, suggests that dysregulation in host anti-tumour immune response might contribute to the poor prognosis predicted by the 10-NMRG signature.
In addition, high-risk score GBM also expressed significantly higher levels of integrins, EMT transcription factors, ECM components and matrix protease genes. ASOEA revealed that these genes are involved in regulating integrins-mediated cell–matrix interaction, RHO family GTPases- and FAK-signalling-induced cytoskeleton reorganisation, ECM remodelling and angiogenesis (Table 5; Table S12, Supplementary File S1), all of which are crucial for tumour cell migration and invasion. Among them, C3AR1, CLCF1, OSMR, KCNN4 and HTR7 were also positively correlated with several genes related to cell–matrix adhesion (e.g., FN1, ITGA3, ITGA5, ITGB1), EMT (e.g., TGFB1, TGFBR2, SNAI1, SNAI2, VIM) and matrix remodelling (e.g., ADAM8, ADAM9, MMP14, MMP19) in GBM (Figure 7C–E). Altogether, the current findings suggest a possible connection between high expressions of the 10-NMRGs with increased modulation of the cell–matrix interaction, cytoskeleton reorganisation and ECM degradation in the high-risk score GBM, through the modulation of the listed genes. This may further enhance the invasion potential and aggressiveness of high-risk score GBM, leading to shorter OS in the patients.
The poor prognosis of the mesenchymal subtype GBM is known to be associated with their mesenchymal differentiation, angiogenesis, pro-inflammatory and immune suppression gene signatures [66,67]. While most of the genes in the 10-NMRG signature (RETN, C3AR1, CLCF1, NTRK1, OSMR, KCNN4 and HTR7) are individually linked with poor prognosis, they also exhibited a relatively higher expression in the mesenchymal subtype compared to the classical and proneural subtypes (Figure 8). It is noteworthy that 50% of the high-risk score patients were classified as a mesenchymal subtype. Furthermore, three-quarters of these mesenchymal subtype GBM patients were also stratified as high-risk, based on the 10-NMRG signature. While the current findings suggest a link between 10-NMRG signature and mesenchymal subtype, a strong association between increased inflammatory, immune response and tumour-promoting hallmarks are observed in the high-risk score GBM. Notably, hyperactivation of immune-related pathways, including JAK-STAT signalling, NF-kB signalling and complement system, are all implicated in mediating EMT, tumour angiogenesis and production of MMPs in creating a proteolytic environment that favours GBM invasion [64,65,68]. This dual interplay of immunomodulation suggests a multifaceted microenvironment in the high-risk score GBM that facilitates tumour immune evasion and tumour invasion. Together, these factors collectively contribute to a more aggressive phenotype in GBM, which results in poor survival in the high-risk score group.
The strong association between high-risk score GBM with immunomodulation and tumour-associated hallmarks has provided insight into the clinically relevant mechanisms that contribute to the poor prognosis predicted by the 10-NMRG signature. Beyond their established neuromodulatory roles in neurons and glial cells [69,70,71], the current findings indicated that neuromodulators, including neuropeptides, neurotrophic factors, and neurotransmitters, could potentially drive GBM progression by altering immune response and activating oncogenic pathways. These findings also grant future preclinical research with elucidating the biological functions of the clinically significant neuromodulators in GBM pathogenesis, especially their roles in modulating tumour–immune dynamics in facilitating GBM progression and invasion.

5. Conclusions

This study established a clinically significant 10-NMRG signature comprising neuropeptides (IGF2, RETN), neuropeptide receptors (EDNRB, C3AR1), neurotrophic factor (CLCF1), neurotrophic factor receptor (NTRK1, OSMR), neurotransmitter receptor (KCNN4, HTR7) and neurotransmitter system-related (SLC18A3) genes that identifies high-risk GBM with immunomodulation and tumour-associated hallmarks. The risk-scoring model effectively stratifies high-risk patients across Caucasian and Asian populations, suggesting potential broader clinical application for treatment stratification and survival prediction. Moving forward, further research is needed to clarify their clinical significance and develop targeted therapies, especially for high-risk patients who may benefit from intensified monitoring or treatment.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines13112640/s1. Supplementary File S1—Table S1: Demographic and clinical characteristics of GBM patients from TCGA and CGGA database; Table S2: List of neuropeptide and neuropeptide receptor genes included in differential expression analysis; Table S3: List of neurotrophic factor and neurotrophic factor receptor genes included in differential expression analysis; Table S4: List of neurotransmitter receptor genes included in differential expression analysis; Table S5: List of neurotransmitter system-related genes included in differential expression analysis; Table S6: List of upregulated NMRGs acting as a prognostic marker for GBM patients from TCGA cohort; Table S7: List of downregulated NMRGs acting as a prognostic marker for GBM patients from TCGA cohort; Table S8: List of upregulated NMRGs acting as a prognostic marker for GBM patients from CGGA cohort; Table S9: List of downregulated NMRGs acting as a prognostic marker for GBM patients from CGGA cohort; Table S10: List of upregulated NMRGs identified in GBM samples from TCGA, CGGA, GSE147352 and GSE165595 database; Table S11: List of downregulated NMRGs identified in GBM samples from TCGA, CGGA, GSE147352 and GSE165595 database; Table S12: Differentially expressed genes (DEGs) enriched in GO Biological Process, KEGG Pathway and Reactome Pathway terms in GBM with high 10-NMRGs risk score; Supplementary File S2—Figure S1: GSEA (MSigDB hallmark) of the 10-NMRG high-risk score group in GBM TCGA cohort; Figure S2: GSEA (KEGG_MEDICUS pathway) of the 10-NMRG high-risk score group in GBM TCGA cohort; Figure S3: GSEA (Reactome pathway) of the 10-NMRG high-risk score in GBM TCGA cohort.

Author Contributions

M.Y.C.: conceptualization, formal analysis (bioinformatics), validation, visualisation, writing—original draft, review and editing; S.S.X.L.: conceptualization, formal analysis (bioinformatics), validation, writing—original draft, review and editing; M.M.: writing—review and editing; B.W.S.P.: conceptualization, software, data curation, supervision (bioinformatics analysis), visualisation, writing—review and editing; M.N.A.K.: conceptualization, supervision, project administration, funding acquisition, writing—review and editing. M.Y.C. and S.S.X.L. contributed equally as first authors. B.W.S.P. and M.N.A.K. contributed equally as corresponding authors. All authors have read and agreed to the published version of the manuscript.

Funding

This project was funded by the Fundamental Research Grant Scheme (FRGS) from the Ministry of Higher Education (FRGS/1/2023/STG03/MUSM/03/1) and supported by the Seed Grant from the Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia (SED-000231 and SED-000381).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All alignment scripts and R scripts used in this paper can be accessed through the following link: https://github.com/drgellerbing/nmrg (accessed on 11 November 2023).

Acknowledgments

M.Y.C., S.S.X.L. and B.W.S.P. were supported by the Monash University Malaysia Ph.D. Merit Scholarship. The authors would like to thank the Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Malaysia for the support provided, and also acknowledge Monash University Malaysia’s High-Performance Computing for the provision of computational resources. The results shown here are in part based upon data generated by The Cancer Genome Atlas (TCGA) Research Network (https://www.cancer.gov/tcga) (accessed on 2 July 2023). We acknowledge the TCGA Research Network for generating and making publicly available the data that supported this work. Data from the Genotype-Tissue Expression (GTEx) Project were obtained from the GTEx Portal (https://xenabrowser.net/datapages/) (accessed on 2 July 2023) and supported by the Common Fund of the Office of the Director of the National Institutes of Health, and by NCI, NHGRI, NHLBI, NIDA, NIMH, and NINDS. Additionally, RNA-Seq expression datasets and the respective dataset’s clinical information were retrieved from the GEO database (GSE147352, GSE165595) (https://www.ncbi.nlm.nih.gov/geo/) (accessed on 2 July 2023), and we thank the authors of the original studies for making these data publicly available. We also acknowledge the Chinese Glioma Genome Atlas (CGGA) database (http://www.cgga.org.cn) (accessed on 2 July 2023) for providing data used in this study.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
NMRGsNeuromodulation-Related Genes
deNMRGsDifferentially Expressed NMRG
GBMGlioblastoma, IDH Wildtype
TCGAThe Cancer Genome Atlas
CGGAChinese Glioma Genome Atlas
GEOGene Expression Omnibus
10-NMRG10-Neuromodulation-Related Gene
GSEAGene Set Enrichment Analysis
ASOEAActive Subnetwork-Oriented Enrichment Analysis
ECMExtracellular Matrix
CCLC-C Motif Chemokine Ligand
CXCLC-X-C Motif Chemokine Ligand
GOGene Ontology
EMTEpithelial-to-Mesenchymal Transition

References

  1. Ostrom, Q.T.; Price, M.; Neff, C.; Cioffi, G.; Waite, K.A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2016—2020. Neuro-Oncology 2023, 25, iv1–iv99. [Google Scholar] [CrossRef] [PubMed]
  2. Ostrom, Q.T.; Price, M.; Neff, C.; Cioffi, G.; Waite, K.A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2015–2019. Neuro-Oncology 2022, 24, v1–v95. [Google Scholar] [CrossRef]
  3. Palma, C.; Nardelli, F.; Manzini, S.; Maggi, C. Substance P activates responses correlated with tumour growth in human glioma cell lines bearing tachykinin NK1 receptors. Br. J. Cancer 1999, 79, 236–243. [Google Scholar] [CrossRef]
  4. Lawn, S.; Krishna, N.; Pisklakova, A.; Qu, X.; Fenstermacher, D.A.; Fournier, M.; Vrionis, F.D.; Tran, N.; Chan, J.A.; Kenchappa, R.S. Neurotrophin signaling via TrkB and TrkC receptors promotes the growth of brain tumor-initiating cells. J. Biol. Chem. 2015, 290, 3814–3824. [Google Scholar] [CrossRef] [PubMed]
  5. Pucci, S.; Fasoli, F.; Moretti, M.; Benfante, R.; Di Lascio, S.; Viani, P.; Daga, A.; Gordon, T.J.; McIntosh, M.; Zoli, M. Choline and nicotine increase glioblastoma cell proliferation by binding and activating α7-and α9-containing nicotinic receptors. Pharmacol. Res. 2021, 163, 105336. [Google Scholar] [CrossRef]
  6. Thompson, E.G.; Sontheimer, H. Acetylcholine receptor activation as a modulator of glioblastoma invasion. Cells 2019, 8, 1203. [Google Scholar] [CrossRef]
  7. Piao, Y.; Lu, L.; De Groot, J. AMPA receptors promote perivascular glioma invasion via β1 integrin–dependent adhesion to the extracellular matrix. Neuro-Oncology 2009, 11, 260–273. [Google Scholar] [CrossRef]
  8. Venkatesh, H.S.; Morishita, W.; Geraghty, A.C.; Silverbush, D.; Gillespie, S.M.; Arzt, M.; Tam, L.T.; Espenel, C.; Ponnuswami, A.; Ni, L. Electrical and synaptic integration of glioma into neural circuits. Nature 2019, 573, 539–545. [Google Scholar] [CrossRef]
  9. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.; Pfister, S.M.; Reifenberger, G. The 2021 WHO classification of tumors of the central nervous system: A summary. Neuro-Oncology 2021, 23, 1231–1251. [Google Scholar] [CrossRef]
  10. Huang, T.; Yang, Y.; Song, X.; Wan, X.; Wu, B.; Sastry, N.; Horbinski, C.M.; Zeng, C.; Tiek, D.; Goenka, A. PRMT6 methylation of RCC1 regulates mitosis, tumorigenicity, and radiation response of glioblastoma stem cells. Mol. Cell 2021, 81, 1276–1291.e9. [Google Scholar] [CrossRef] [PubMed]
  11. Xu, L.; Chen, Y.; Huang, Y.; Sandanaraj, E.; Yu, J.S.; Lin, R.Y.-T.; Dakle, P.; Ke, X.-Y.; Chong, Y.K.; Koh, L. Topography of transcriptionally active chromatin in glioblastoma. Sci. Adv. 2021, 7, eabd4676. [Google Scholar] [CrossRef]
  12. Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef]
  13. Li, B.; Dewey, C.N. RSEM: Accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 2011, 12, 323. [Google Scholar] [CrossRef]
  14. Goldman, M.J.; Craft, B.; Hastie, M.; Repečka, K.; McDade, F.; Kamath, A.; Banerjee, A.; Luo, Y.; Rogers, D.; Brooks, A.N. Visualizing and interpreting cancer genomics data via the Xena platform. Nat. Biotechnol. 2020, 38, 675–678. [Google Scholar] [CrossRef]
  15. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef]
  16. Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013, 6, pl1. [Google Scholar] [CrossRef]
  17. de Bruijn, I.; Kundra, R.; Mastrogiacomo, B.; Tran, T.N.; Sikina, L.; Mazor, T.; Li, X.; Ochoa, A.; Zhao, G.; Lai, B.; et al. Analysis and Visualization of Longitudinal Genomic and Clinical Data from the AACR Project GENIE Biopharma Collaborative in cBioPortal. Cancer Res. 2023, 83, 3861–3867. [Google Scholar] [CrossRef]
  18. Vivian, J.; Rao, A.A.; Nothaft, F.A.; Ketchum, C.; Armstrong, J.; Novak, A.; Pfeil, J.; Narkizian, J.; Deran, A.D.; Musselman-Brown, A. Toil enables reproducible, open source, big biomedical data analyses. Nat. Biotechnol. 2017, 35, 314–316. [Google Scholar] [CrossRef] [PubMed]
  19. Liu, X.; Li, Y.; Qian, Z.; Sun, Z.; Xu, K.; Wang, K.; Liu, S.; Fan, X.; Li, S.; Zhang, Z. A radiomic signature as a non-invasive predictor of progression-free survival in patients with lower-grade gliomas. NeuroImage Clin. 2018, 20, 1070–1077. [Google Scholar] [CrossRef] [PubMed]
  20. Wang, Y.; Qian, T.; You, G.; Peng, X.; Chen, C.; You, Y.; Yao, K.; Wu, C.; Ma, J.; Sha, Z. Localizing seizure-susceptible brain regions associated with low-grade gliomas using voxel-based lesion-symptom mapping. Neuro-Oncology 2015, 17, 282–288. [Google Scholar] [CrossRef] [PubMed]
  21. Zhang, K.; Liu, X.; Li, G.; Chang, X.; Li, S.; Chen, J.; Zhao, Z.; Wang, J.; Jiang, T.; Chai, R. Clinical management and survival outcomes of patients with different molecular subtypes of diffuse gliomas in China (2011–2017): A multicenter retrospective study from CGGA. Cancer Biol. Med. 2022, 19, 1460. [Google Scholar] [CrossRef] [PubMed]
  22. Zhao, Z.; Zhang, K.-N.; Wang, Q.; Li, G.; Zeng, F.; Zhang, Y.; Wu, F.; Chai, R.; Wang, Z.; Zhang, C. Chinese Glioma Genome Atlas (CGGA): A comprehensive resource with functional genomic data from Chinese glioma patients. Genom. Proteom. Bioinform. 2021, 19, 1–12. [Google Scholar] [CrossRef]
  23. Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef] [PubMed]
  24. Zhu, A.; Ibrahim, J.G.; Love, M.I. Heavy-tailed prior distributions for sequence count data: Removing the noise and preserving large differences. Bioinformatics 2019, 35, 2084–2092. [Google Scholar] [CrossRef]
  25. Therneau, T.M.; Grambsch, P.M.; Therneau, T.M.; Grambsch, P.M. The Cox Model; Springer: New York, NY, USA, 2000. [Google Scholar]
  26. Kassambara, A.; Kosinski, M.; Biecek, P.; Fabian, S. Survminer: Drawing Survival Curves Using’ggplot2′, R Package Version 0.4.9.2021; 2021. Available online: https://rpkgs.datanovia.com/survminer/index.html (accessed on 13 October 2025).
  27. Lausen, B.; Hothorn, T.; Bretz, F.; Schumacher, M. Assessment of optimal selected prognostic factors. Biom. J. J. Math. Methods Biosci. 2004, 46, 364–374. [Google Scholar] [CrossRef]
  28. Subramanian, A.; Tamayo, P.; Mootha, V.K.; Mukherjee, S.; Ebert, B.L.; Gillette, M.A.; Paulovich, A.; Pomeroy, S.L.; Golub, T.R.; Lander, E.S. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 2005, 102, 15545–15550. [Google Scholar] [CrossRef]
  29. Liberzon, A.; Birger, C.; Thorvaldsdóttir, H.; Ghandi, M.; Mesirov, J.P.; Tamayo, P. The molecular signatures database hallmark gene set collection. Cell Syst. 2015, 1, 417–425. [Google Scholar] [CrossRef]
  30. Ulgen, E.; Ozisik, O.; Sezerman, O.U. pathfindR: An R package for comprehensive identification of enriched pathways in omics data through active subnetworks. Front. Genet. 2019, 10, 425394. [Google Scholar] [CrossRef]
  31. Quinn, T.P.; Richardson, M.F.; Lovell, D.; Crowley, T.M. propr: An R-package for identifying proportionally abundant features using compositional data analysis. Sci. Rep. 2017, 7, 16252. [Google Scholar] [CrossRef]
  32. Martín-Fernández, J.A.; Palarea-Albaladejo, J.; Olea, R.A. Dealing with zeros. Compos. Data Anal. Theory Appl. 2011, 43–58. [Google Scholar]
  33. Palarea-Albaladejo, J.; Martín-Fernández, J.A. zCompositions—R package for multivariate imputation of left-censored data under a compositional approach. Chemom. Intell. Lab. Syst. 2015, 143, 85–96. [Google Scholar] [CrossRef]
  34. Monje, M. The neuroscience of brain cancers. Neuron 2025, 113, 2734–2739. [Google Scholar] [CrossRef] [PubMed]
  35. Winkler, F.; Venkatesh, H.S.; Amit, M.; Batchelor, T.; Demir, I.E.; Deneen, B.; Gutmann, D.H.; Hervey-Jumper, S.; Kuner, T.; Mabbott, D. Cancer neuroscience: State of the field, emerging directions. Cell 2023, 186, 1689–1707. [Google Scholar] [CrossRef] [PubMed]
  36. Deb, A.; Deshmukh, B.; Ramteke, P.; Bhati, F.K.; Bhat, M.K. Resistin: A journey from metabolism to cancer. Transl. Oncol. 2021, 14, 101178. [Google Scholar] [CrossRef]
  37. Sudan, S.K.; Deshmukh, S.K.; Poosarla, T.; Holliday, N.P.; Dyess, D.L.; Singh, A.P.; Singh, S. Resistin: An inflammatory cytokine with multi-faceted roles in cancer. Biochim. Et Biophys. Acta (BBA)-Rev. Cancer 2020, 1874, 188419. [Google Scholar] [CrossRef]
  38. Huang, J.; Zhou, L.; Deng, K. Prognostic marker C3AR1 is associated with ovarian cancer cell proliferation and immunosuppression in the tumor microenvironment. J. Ovarian Res. 2023, 16, 64. [Google Scholar] [CrossRef]
  39. Shu, C.; Zha, H.; Long, H.; Wang, X.; Yang, F.; Gao, J.; Hu, C.; Zhou, L.; Guo, B.; Zhu, B. C3a-C3aR signaling promotes breast cancer lung metastasis via modulating carcinoma associated fibroblasts. J. Exp. Clin. Cancer Res. 2020, 39, 11. [Google Scholar] [CrossRef]
  40. Sims, N.A. Cardiotrophin-like cytokine factor 1 (CLCF1) and neuropoietin (NP) signalling and their roles in development, adulthood, cancer and degenerative disorders. Cytokine Growth Factor Rev. 2015, 26, 517–522. [Google Scholar] [CrossRef]
  41. Cocco, E.; Scaltriti, M.; Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol. 2018, 15, 731–747. [Google Scholar] [CrossRef]
  42. Kim, J.; Lee, Y.; Cho, H.-J.; Lee, Y.-E.; An, J.; Cho, G.-H.; Ko, Y.-H.; Joo, K.M.; Nam, D.-H. NTRK1 fusion in glioblastoma multiforme. PLoS ONE 2014, 9, e91940. [Google Scholar] [CrossRef] [PubMed]
  43. Masjedi, A.; Hajizadeh, F.; Dargani, F.B.; Beyzai, B.; Aksoun, M.; Hojjat-Farsangi, M.; Zekiy, A.; Jadidi-Niaragh, F. Oncostatin M: A mysterious cytokine in cancers. Int. Immunopharmacol. 2021, 90, 107158. [Google Scholar] [CrossRef] [PubMed]
  44. Li, Q.-T.; Feng, Y.-M.; Ke, Z.-H.; Qiu, M.-J.; He, X.-X.; Wang, M.-M.; Li, Y.-N.; Xu, J.; Shi, L.-L.; Xiong, Z.-F. KCNN4 promotes invasion and metastasis through the MAPK/ERK pathway in hepatocellular carcinoma. J. Investig. Med. 2020, 68, 68–74. [Google Scholar] [CrossRef] [PubMed]
  45. Xu, P.; Mo, X.; Xia, R.; Jiang, L.; Zhang, C.; Xu, H.; Sun, Q.; Zhou, G.; Zhang, Y.; Wang, Y. KCNN4 promotes the progression of lung adenocarcinoma by activating the AKT and ERK signaling pathways. Cancer Biomark. 2021, 31, 187–201. [Google Scholar] [CrossRef] [PubMed]
  46. Karmakar, S.; Lal, G. Role of serotonin receptor signaling in cancer cells and anti-tumor immunity. Theranostics 2021, 11, 5296. [Google Scholar] [CrossRef]
  47. Soroceanu, L.; Kharbanda, S.; Chen, R.; Soriano, R.H.; Aldape, K.; Misra, A.; Zha, J.; Forrest, W.F.; Nigro, J.M.; Modrusan, Z. Identification of IGF2 signaling through phosphoinositide-3-kinase regulatory subunit 3 as a growth-promoting axis in glioblastoma. Proc. Natl. Acad. Sci. USA 2007, 104, 3466–3471. [Google Scholar] [CrossRef]
  48. Liu, Y.; Ye, F.; Yamada, K.; Tso, J.L.; Zhang, Y.; Nguyen, D.H.; Dong, Q.; Soto, H.; Choe, J.; Dembo, A. Autocrine endothelin-3/endothelin receptor B signaling maintains cellular and molecular properties of glioblastoma stem cells. Mol. Cancer Res. 2011, 9, 1668–1685. [Google Scholar] [CrossRef]
  49. Pan, D.-s.; Feng, S.-z.; Cao, P.; Li, J.-j. Endothelin B receptor promotes the proliferation and immune escape of malignant gliomas. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1230–1235. [Google Scholar] [CrossRef]
  50. Rosberg, R.; Smolag, K.I.; Sjolund, J.; Johansson, E.; Bergelin, C.; Wahlden, J.; Pantazopoulou, V.; Ceberg, C.; Pietras, K.; Blom, A.M. Hypoxia-induced Complement Component 3 Promotes Aggressive Tumor Growth in the Glioblastoma Microenvironment. JCI Insight 2024, 9, e179854. [Google Scholar] [CrossRef]
  51. Ah-Pine, F.; Malaterre-Septembre, A.; Bedoui, Y.; Khettab, M.; Neal, J.W.; Freppel, S.; Gasque, P. Complement activation and up-regulated expression of anaphylatoxin C3a/C3aR in glioblastoma: Deciphering the links with TGF-β and VEGF. Cancers 2023, 15, 2647. [Google Scholar] [CrossRef]
  52. Shen, S.-H.; Guo, J.-F.; Huang, J.; Zhang, Q.; Cui, Y. Bromodomain-containing protein 4 activates cardiotrophin-like cytokine factor 1, an unfavorable prognostic biomarker, and promotes glioblastoma in vitro. Ann. Transl. Med. 2022, 10, 475. [Google Scholar] [CrossRef]
  53. Torre, M.; Vasudevaraja, V.; Serrano, J.; DeLorenzo, M.; Malinowski, S.; Blandin, A.-F.; Pages, M.; Ligon, A.H.; Dong, F.; Meredith, D.M.; et al. Molecular and clinicopathologic features of gliomas harboring NTRK fusions. Acta Neuropathol. Commun. 2020, 8, 107. [Google Scholar] [CrossRef]
  54. Matsumoto, Y.; Ichikawa, T.; Kurozumi, K.; Otani, Y.; Fujimura, A.; Fujii, K.; Tomita, Y.; Hattori, Y.; Uneda, A.; Tsuboi, N. Annexin A2–STAT3–Oncostatin M receptor axis drives phenotypic and mesenchymal changes in glioblastoma. Acta Neuropathol. Commun. 2020, 8, 42. [Google Scholar] [CrossRef]
  55. Natesh, K.; Bhosale, D.; Desai, A.; Chandrika, G.; Pujari, R.; Jagtap, J.; Chugh, A.; Ranade, D.; Shastry, P. Oncostatin-M differentially regulates mesenchymal and proneural signature genes in gliomas via STAT3 signaling. Neoplasia 2015, 17, 225–237. [Google Scholar] [CrossRef]
  56. Sharanek, A.; Burban, A.; Laaper, M.; Heckel, E.; Joyal, J.-S.; Soleimani, V.D.; Jahani-Asl, A. OSMR controls glioma stem cell respiration and confers resistance of glioblastoma to ionizing radiation. Nat. Commun. 2020, 11, 4116. [Google Scholar] [CrossRef]
  57. Jahani-Asl, A.; Yin, H.; Soleimani, V.D.; Haque, T.; Luchman, H.A.; Chang, N.C.; Sincennes, M.-C.; Puram, S.V.; Scott, A.M.; Lorimer, I.A.J.; et al. Control of glioblastoma tumorigenesis by feed-forward cytokine signaling. Nat. Neurosci. 2016, 19, 798–806. [Google Scholar] [CrossRef] [PubMed]
  58. D’Alessandro, G.; Catalano, M.; Sciaccaluga, M.; Chece, G.; Cipriani, R.; Rosito, M.; Grimaldi, A.; Lauro, C.; Cantore, G.; Santoro, A.; et al. KCa3.1 channels are involved in the infiltrative behavior of glioblastoma in vivo. Cell Death Dis. 2013, 4, e773. [Google Scholar] [CrossRef]
  59. Alessandro, G.; Grimaldi, A.; Chece, G.; Porzia, A.; Esposito, V.; Santoro, A.; Salvati, M.; Mainiero, F.; Ragozzino, D.; Di Angelantonio, S.; et al. KCa3.1 channel inhibition sensitizes malignant gliomas to temozolomide treatment. Oncotarget 2016, 7, 30781–30796. [Google Scholar] [CrossRef]
  60. Mahé, C.; Bernhard, M.; Bobirnac, I.; Keser, C.; Loetscher, E.; Feuerbach, D.; Dev, K.K.; Schoeffter, P. Functional expression of the serotonin 5-HT7 receptor in human glioblastoma cell lines. Br. J. Pharmacol. 2004, 143, 404–410. [Google Scholar] [CrossRef] [PubMed]
  61. Bikfalvi, A.; da Costa, C.A.; Avril, T.; Barnier, J.-V.; Bauchet, L.; Brisson, L.; Cartron, P.F.; Castel, H.; Chevet, E.; Chneiweiss, H. Challenges in glioblastoma research: Focus on the tumor microenvironment. Trends Cancer 2023, 9, 9–27. [Google Scholar] [CrossRef] [PubMed]
  62. DeCordova, S.; Shastri, A.; Tsolaki, A.G.; Yasmin, H.; Klein, L.; Singh, S.K.; Kishore, U. Molecular heterogeneity and immunosuppressive microenvironment in glioblastoma. Front. Immunol. 2020, 11, 1402. [Google Scholar] [CrossRef]
  63. Gieryng, A.; Pszczolkowska, D.; Walentynowicz, K.A.; Rajan, W.D.; Kaminska, B. Immune microenvironment of gliomas. Lab. Investig. 2017, 97, 498–518. [Google Scholar] [CrossRef]
  64. Piperi, C.; Papavassiliou, K.A.; Papavassiliou, A.G. Pivotal role of STAT3 in shaping glioblastoma immune microenvironment. Cells 2019, 8, 1398. [Google Scholar] [CrossRef]
  65. Atkinson, G.P.; Nozell, S.E.; Benveniste, E.N. NF-κB and STAT3 signaling in glioma: Targets for future therapies. Expert Rev. Neurother. 2010, 10, 575–586. [Google Scholar] [CrossRef]
  66. Verhaak, R.G.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010, 17, 98–110. [Google Scholar] [CrossRef]
  67. Wang, Q.; Hu, B.; Hu, X.; Kim, H.; Squatrito, M.; Scarpace, L.; DeCarvalho, A.C.; Lyu, S.; Li, P.; Li, Y. Tumor evolution of glioma-intrinsic gene expression subtypes associates with immunological changes in the microenvironment. Cancer Cell 2017, 32, 42–56.e46. [Google Scholar] [CrossRef]
  68. Bouwens Van Der Vlis, T.; Kros, J.; Mustafa, D.; van Wijck, R.; Ackermans, L.; van Hagen, P.; van der Spek, P. The complement system in glioblastoma multiforme. Acta Neuropathol. Commun. 2018, 6, 91. [Google Scholar] [CrossRef]
  69. Ayala-Lopez, N.; Watts, S.W. Physiology and pharmacology of neurotransmitter transporters. Compr. Physiol. 2021, 11, 2279–2295. [Google Scholar] [CrossRef]
  70. Colucci-D’Amato, L.; Speranza, L.; Volpicelli, F. Neurotrophic factor BDNF, physiological functions and therapeutic potential in depression, neurodegeneration and brain cancer. Int. J. Mol. Sci. 2020, 21, 7777. [Google Scholar] [CrossRef] [PubMed]
  71. Van Den Pol, A.N. Neuropeptide transmission in brain circuits. Neuron 2012, 76, 98–115. [Google Scholar] [CrossRef]
Figure 1. Workflow of the study. Differential expression analysis was performed across four RNA-Seq datasets (TCGA, CGGA, GSE147532, and GSE165595) to identify neuromodulation-related genes (NMRGs) dysregulated in GBM compared to normal brain tissues. NMRGs with concordant expression trends in at least two datasets were evaluated for prognostic value in TCGA and CGGA cohorts. Genes consistently associated with similar survival outcomes were used to construct the 10-NMRG signature. The prognostic performance of this signature was evaluated and validated in both cohorts. GSEA was conducted to characterise high-risk score GBM stratified by the gene signature. Compositional proportionality analysis was performed to understand the relationship between signature genes with immune-related genes and pro-motility genes, while expression patterns of the signature genes were examined across different GBM subtypes. GBM—glioblastoma; DEGs—differentially expressed genes; TCGA—The Cancer Genome Atlas; CGGA—Chinese Glioma Genome Atlas; GTEX—genotype-tissue expression; GSEA—gene set enrichment analysis.
Figure 1. Workflow of the study. Differential expression analysis was performed across four RNA-Seq datasets (TCGA, CGGA, GSE147532, and GSE165595) to identify neuromodulation-related genes (NMRGs) dysregulated in GBM compared to normal brain tissues. NMRGs with concordant expression trends in at least two datasets were evaluated for prognostic value in TCGA and CGGA cohorts. Genes consistently associated with similar survival outcomes were used to construct the 10-NMRG signature. The prognostic performance of this signature was evaluated and validated in both cohorts. GSEA was conducted to characterise high-risk score GBM stratified by the gene signature. Compositional proportionality analysis was performed to understand the relationship between signature genes with immune-related genes and pro-motility genes, while expression patterns of the signature genes were examined across different GBM subtypes. GBM—glioblastoma; DEGs—differentially expressed genes; TCGA—The Cancer Genome Atlas; CGGA—Chinese Glioma Genome Atlas; GTEX—genotype-tissue expression; GSEA—gene set enrichment analysis.
Biomedicines 13 02640 g001
Figure 2. Venn Diagram showing common prognostic NMRGs between TCGA and CGGA cohorts. IGF2, RETN, EDNRB, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7 predicted similar survival outcomes for GBM patients from the two cohorts. Genes predicting poorer prognoses are indicated in red, whereas those predicting better prognoses are denoted in green. The deNMRGs predicting for similar survival outcomes in GBM patients from both cohorts are presented in the intersection region.
Figure 2. Venn Diagram showing common prognostic NMRGs between TCGA and CGGA cohorts. IGF2, RETN, EDNRB, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7 predicted similar survival outcomes for GBM patients from the two cohorts. Genes predicting poorer prognoses are indicated in red, whereas those predicting better prognoses are denoted in green. The deNMRGs predicting for similar survival outcomes in GBM patients from both cohorts are presented in the intersection region.
Biomedicines 13 02640 g002
Figure 3. Kaplan–Meier survival curves for the 10 NMRGs from GBM (A) TCGA (n = 140) and (B) CGGA (n = 89) cohorts. GBM patients from the respective cohorts were stratified into low- (blue) and high-expression (red) groups, based on optimal cut-offs (log-rank p ≤ 0.05). The Kaplan–Meier survival curves showed that IGF2, RETN, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7 consistently predict poorer survival, while EDNRB predicts a more favourable survival outcome in both cohorts. The patient group sizes and log-rank p-value for each curve are as shown.
Figure 3. Kaplan–Meier survival curves for the 10 NMRGs from GBM (A) TCGA (n = 140) and (B) CGGA (n = 89) cohorts. GBM patients from the respective cohorts were stratified into low- (blue) and high-expression (red) groups, based on optimal cut-offs (log-rank p ≤ 0.05). The Kaplan–Meier survival curves showed that IGF2, RETN, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7 consistently predict poorer survival, while EDNRB predicts a more favourable survival outcome in both cohorts. The patient group sizes and log-rank p-value for each curve are as shown.
Biomedicines 13 02640 g003
Figure 4. Forest plot of the Cox proportional hazard model for the 10-NMRG in GBM patients from TCGA and CGGA cohorts. The prognostic values of the 10 genes were assessed using the Cox proportional hazard model. Hazard ratio (HR) > 1 indicates a worse prognosis in GBM patients with higher gene expression, while HR < 1 implies a better prognosis in GBM patients with higher gene expression.
Figure 4. Forest plot of the Cox proportional hazard model for the 10-NMRG in GBM patients from TCGA and CGGA cohorts. The prognostic values of the 10 genes were assessed using the Cox proportional hazard model. Hazard ratio (HR) > 1 indicates a worse prognosis in GBM patients with higher gene expression, while HR < 1 implies a better prognosis in GBM patients with higher gene expression.
Biomedicines 13 02640 g004
Figure 5. GBM patients with high 10-NMRG signature risk scores were associated with worse survival outcomes. GBM patients in the training (A,B; n = 83) and validation sets (C,D; n = 57) were stratified into low-risk score (blue) and high-risk score (red) groups based on log-rank tests. The overall survival probabilities of the 10-NMRG signature were evaluated using Kaplan–Meier curves, with the prognostic capacity evaluation of the gene signature being evaluated using time-dependent ROC curves.
Figure 5. GBM patients with high 10-NMRG signature risk scores were associated with worse survival outcomes. GBM patients in the training (A,B; n = 83) and validation sets (C,D; n = 57) were stratified into low-risk score (blue) and high-risk score (red) groups based on log-rank tests. The overall survival probabilities of the 10-NMRG signature were evaluated using Kaplan–Meier curves, with the prognostic capacity evaluation of the gene signature being evaluated using time-dependent ROC curves.
Biomedicines 13 02640 g005
Figure 6. 10-NMRG signature risk scores are consistently associated with worse survival outcomes. GBM patients in the full TCGA and CGGA cohorts were similarly stratified into low-risk score (blue) and high-risk score (red) groups based on the log-rank tests. Kaplan–Meier survival analysis was used to evaluate the prognosis of the patients stratified by the gene signature in TCGA (A) and CGGA (C) cohorts. (B,D) Heatmaps of the expression of the 10-NMRG in the GBM samples.
Figure 6. 10-NMRG signature risk scores are consistently associated with worse survival outcomes. GBM patients in the full TCGA and CGGA cohorts were similarly stratified into low-risk score (blue) and high-risk score (red) groups based on the log-rank tests. Kaplan–Meier survival analysis was used to evaluate the prognosis of the patients stratified by the gene signature in TCGA (A) and CGGA (C) cohorts. (B,D) Heatmaps of the expression of the 10-NMRG in the GBM samples.
Biomedicines 13 02640 g006
Figure 7. Compositional proportionality analysis reveals close association between the 10-NMRGs and cancer hallmark-related genes. The heatmap displays proportional relationships between the 10-NMRGs and immune response, EMT, cell–matrix adhesion, EC remodelling and angiogenesis genes. Dot size and colour intensity reflect proportionality strength (blue = direct; red = inverse), while blanks indicate non-significant relationships.
Figure 7. Compositional proportionality analysis reveals close association between the 10-NMRGs and cancer hallmark-related genes. The heatmap displays proportional relationships between the 10-NMRGs and immune response, EMT, cell–matrix adhesion, EC remodelling and angiogenesis genes. Dot size and colour intensity reflect proportionality strength (blue = direct; red = inverse), while blanks indicate non-significant relationships.
Biomedicines 13 02640 g007
Figure 8. Expression of 10-NMRG in different GBM subtypes. The violin plots showed the different expression levels of IGF2, RETN, C3AR1, EDNRB, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7 in classical, mesenchymal and proneural subtypes of GBM from TCGA cohort. * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001.
Figure 8. Expression of 10-NMRG in different GBM subtypes. The violin plots showed the different expression levels of IGF2, RETN, C3AR1, EDNRB, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7 in classical, mesenchymal and proneural subtypes of GBM from TCGA cohort. * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001.
Biomedicines 13 02640 g008
Table 1. Number of samples in each RNA-Seq cohort.
Table 1. Number of samples in each RNA-Seq cohort.
DatasetNormal BrainGBMPaired/Unpaired
TCGA-GTEx211140Unpaired
CGGA2089Unpaired
GSE1473521556Unpaired
GSE1655951515Paired
Table 2. Summary of differentially expressed NMRGs identified in GBM patient samples vs. normal brain tissues.
Table 2. Summary of differentially expressed NMRGs identified in GBM patient samples vs. normal brain tissues.
GenesInput
Gene List
Number
of DEGs
Number of
Upregulated
Genes
Number of
Downregulated Genes
Neuropeptides97502525
Neuropeptide receptors97602238
Neurotrophic factors261495
Neurotrophic factor receptors221697
Neurotransmitter receptors131991287
Neurotransmitter system-related 4733924
Table 3. Multivariate Cox Regression Analysis of 10-NMRG signature and other common GBM prognostic clinical factors.
Table 3. Multivariate Cox Regression Analysis of 10-NMRG signature and other common GBM prognostic clinical factors.
VariableCharacteristicsUnivariable CoxMultivariable Cox
HR95% CIp-ValueHR95% CIp-Value
TCGA
Risk ScoreHigh vs. Low2.151.41–3.27<0.0012.861.36–5.980.005
AgeContinuous1.031.01–1.050.0031.020.99–1.050.196
GenderMale vs. Female0.850.57–1.280.4441.140.55–2.360.716
RadiationYes vs. No0.210.13–0.34<0.0010.140.06–0.36<0.001
ChemotherapyYes vs. No0.400.26–0.62<0.0011.880.72–4.870.196
MGMTMethylated vs. Unmethylated0.670.41–1.100.1120.630.30–1.310.219
SubtypeMesenchymal vs. Classical1.120.66–1.920.6750.920.44–1.920.280
Proneural vs. Classical1.370.76–2.460.2921.020.4–2.60.96
CGGA
Risk ScoreHigh vs. Low2.771.54–4.99<0.0012.411.27–4.560.007
AgeContinuous1.021.00–1.040.0691.021.00–1.040.047
GenderMale vs. Female1.270.79–2.060.3211.480.85–2.560.164
RadiationYes vs. No0.860.47–1.580.6340.830.49–1.410.490
ChemotherapyYes vs. No0.330.19–0.57<0.0010.570.29–1.120.100
MGMTMethylated vs. Unmethylated0.800.50–1.280.3470.440.25–0.780.005
Table 4. Enriched GSEA terms identified in high 10-NMRG risk score GBM using MSigDB hallmark, KEGG_MEDICUS and Reactome gene sets.
Table 4. Enriched GSEA terms identified in high 10-NMRG risk score GBM using MSigDB hallmark, KEGG_MEDICUS and Reactome gene sets.
Hallmarks/PathwaysNESNOM
p-Val
FDR
q-Val
MSigDB Hallmark
TNFA_SIGNALING_VIA_NFKB2.03<0.00010.0053
EPITHELIAL_MESENCHYMAL_TRANSITION2.050.00200.0076
COAGULATION1.99<0.00010.0076
HYPOXIA1.970.00200.0077
IL6_JAK_STAT3_SIGNALING1.910.00790.0093
INFLAMMATORY_RESPONSE1.940.00390.0095
KRAS_SIGNALING_UP1.860.00190.0128
APICAL_JUNCTION1.87<0.00010.0138
ESTROGEN_RESPONSE_EARLY1.81<0.00010.0186
COMPLEMENT1.810.00390.0203
IL2_STAT5_SIGNALING1.790.00980.0213
ANGIOGENESIS1.710.01540.0435
KEGG_MEDICUS
IL6_FAMILY_TO_JAK_STAT_SIGNALING_PATHWAY1.970.00200.0177
ITGA_B_RHOGAP_RHOA_SIGNALING_PATHWAY1.950.00200.0182
ITGA_B_RHOGEF_RHOA_SIGNALING_PATHWAY1.970.00200.0231
ITGA_B_FAK_RAC_SIGNALING_PATHWAY1.900.00400.0281
IL2_FAMILY_TO_JAK_STAT_SIGNALING_PATHWAY1.870.00380.0299
ITGA_B_FAK_CDC42_SIGNALING_PATHWAY1.910.00400.0309
HORMONE_LIKE_CYTOKINE_TO_JAK_STAT_SIGNALING_PATHWAY1.870.00600.0334
ITGA_B_RHOG_RAC_SIGNALING_PATHWAY1.870.00790.0338
ITGA_B_TALIN_VINCULIN_SIGNALING_PATHWAY1.980.00400.0461
Reactome
CELL_SURFACE_INTERACTIONS_AT_THE_VASCULAR_WALL1.980.00190.0343
TNF_RECEPTOR_SUPERFAMILY_TNFSF_MEMBERS_
MEDIATING_NON_CANONICAL_NF_KB_PATHWAY
1.980.00200.0379
DEGRADATION_OF_THE_EXTRACELLULAR_MATRIX1.96<0.00010.0388
ACTIVATION_OF_MATRIX_METALLOPROTEINASES1.99<0.00010.0391
COMPLEMENT_CASCADE1.950.00200.0415
COLLAGEN_DEGRADATION1.94<0.00010.0450
NEGATIVE_REGULATION_OF_TCF_DEPENDENT_
SIGNALING_BY_WNT_LIGAND_ANTAGONISTS
1.92<0.00010.0467
ANTIMICROBIAL_PEPTIDES1.93<0.00010.0472
INTERLEUKIN_4_AND_INTERLEUKIN_13_SIGNALING1.990.00200.0473
Abbreviations: NES, normalised enrichment score; NOM p-val, normalised p value; FDR q-val, false discovery rate (corrected by Benjamini–Hochberg procedure) q value.
Table 5. Enriched ASOEA terms mapped by the differentially expressed genes identified in high 10-NMRG risk score GBMs.
Table 5. Enriched ASOEA terms mapped by the differentially expressed genes identified in high 10-NMRG risk score GBMs.
Term IDGO Biological ProcessHighest
p Value
Immunomodulation-related
GO:0042110T cell activation3.16 × 10−2
GO:0050853B cell receptor signalling pathway2.02 × 10−5
GO:0007159Leukocyte cell–cell adhesion1.44 × 10−2
GO:0030217T cell differentiation6.30 × 10−5
GO:0001819Positive regulation of cytokine production7.93 × 10−5
GO:0002250Adaptive immune response9.31 × 10−6
GO:0043123Positive regulation of I-kappaB kinase/NF-kappaB signalling2.99 × 10−5
GO:0050901Leukocyte tethering or rolling2.10 × 10−2
GO:0032755Positive regulation of interleukin-6 production2.77 × 10−3
GO:0032760Positive regulation of tumour necrosis factor production2.18 × 10−4
GO:0019221Cytokine-mediated signalling pathway9.43 × 10−4
GO:0051092Positive regulation of NF-kappaB transcription factor activity2.55 × 10−3
GO:0031295T cell costimulation1.54 × 10−2
GO:0030593Neutrophil chemotaxis1.55 × 10−2
GO:0032743Positive regulation of interleukin-2 production4.76 × 10−2
GO:0045087Innate immune response2.19 × 10−2
EMT/cell-ECM adhesion/matrix remodelling/angiogenesis-related
GO:0007229Integrin-mediated signalling pathway1.98 × 10−9
GO:0034113Heterotypic cell–cell adhesion4.90 × 10−6
GO:0071260Cellular response to mechanical stimulus7.10 × 10−5
GO:0030335Positive regulation of cell migration5.27 × 10−6
GO:0030199Collagen fibril organisation5.02 × 10−4
GO:0034446Substrate adhesion-dependent cell spreading2.28 × 10−6
GO:0007155Cell adhesion1.32 × 10−4
GO:0030574Collagen catabolic process3.62 × 10−5
GO:0045109Intermediate filament organisation1.61 × 10−5
GO:0070372Regulation of ERK1 and ERK2 cascade2.67 × 10−5
GO:0070374Positive regulation of ERK1 and ERK2 cascade2.81 × 10−4
GO:0022617Extracellular matrix disassembly1.81 × 10−4
GO:0014065Phosphatidylinositol 3-kinase signalling4.16 × 10−4
GO:0014068Positive regulation of phosphatidylinositol 3-kinase signalling1.20 × 10−3
GO:0045766Positive regulation of angiogenesis1.41 × 10−5
GO:0010575Positive regulation of vascular endothelial growth factor production1.20 × 10−3
Term IDKEGG PathwayHighest
p Value
Immunomodulation-related
hsa04630JAK-STAT signalling pathway9.61 × 10−9
hsa04613Neutrophil extracellular trap formation1.50 × 10−15
hsa04060Cytokine–cytokine receptor interaction1.93 × 10−10
hsa04659Th17 cell differentiation2.68 × 10−4
hsa04610Complement and coagulation cascades8.87 × 10−10
hsa04064NF-kappa B signalling pathway2.37 × 10−10
hsa04062Chemokine signalling pathway4.68 × 10−3
hsa04750Inflammatory mediator regulation of TRP channels1.99 × 10−5
hsa04670Leukocyte transendothelial migration7.53 × 10−5
hsa04668TNF signalling pathway1.01 × 10−6
hsa04662B cell receptor signalling pathway3.62 × 10−6
hsa04658Th1 and Th2 cell differentiation8.28 × 10−3
hsa04660T cell receptor signalling pathway6.01 × 10−6
EMT/cell-ECM adhesion/matrix remodelling/angiogenesis-related
hsa04512ECM-receptor interaction1.18 × 10−14
hsa04510Focal adhesion3.01 × 10−13
hsa04915Oestrogen signalling pathway1.88 × 10−9
hsa04014Ras signalling pathway7.27 × 10−6
hsa05205Proteoglycans in cancer2.79 × 10−8
hsa04810Regulation of actin cytoskeleton5.78 × 10−8
hsa04010MAPK signalling pathway5.05 × 10−7
hsa04015Rap1 signalling pathway1.17 × 10−3
hsa04520Adherens junction2.77 × 10−5
Term IDReactome PathwayHighest
p Value
Immunomodulation-related
R-HSA-173623Classical antibody-mediated complement activation1.14 × 10−21
R-HSA-166663Initial triggering of complement5.71 × 10−22
R-HSA-977606Regulation of complement cascade1.06 × 10−21
R-HSA-166786Creation of C4 and C2 activators1.22 × 10−20
R-HSA-166658Complement cascade7.87 × 10−21
R-HSA-5690714CD22-mediated BCR regulation1.51 × 10−15
R-HSA-983695Antigen activates B cell receptor (BCR) leading to generation of second messengers3.11 × 10−15
R-HSA-983705Signalling by the B cell receptor (BCR)3.17 × 10−11
R-HSA-5668541TNFR2 non-canonical NF-kB pathway5.49 × 10−12
R-HSA-451927Interleukin-2 family signalling4.84 × 10−7
R-HSA-512988Interleukin-3, Interleukin-5 and GM-CSF signalling4.84 × 10−7
R-HSA-6785807Interleukin-4 and Interleukin-13 signalling2.83 × 10−8
R-HSA-5676594TNF receptor superfamily (TNFSF) members mediating non-canonical NF-kB pathway1.16 × 10−9
R-HSA-1266695Interleukin-7 signalling2.04 × 10−2
EMT/cell-ECM adhesion/matrix remodelling/angiogenesis-related
R-HSA-1474244Extracellular matrix organisation8.58 × 10−21
R-HSA-1474290Collagen formation6.02 × 10−17
R-HSA-202733Cell surface interactions at the vascular wall1.58 × 10−13
R-HSA-2219530Constitutive signalling by aberrant PI3K in cancer4.06 × 10−11
R-HSA-2022090Assembly of collagen fibrils and other multimeric structures5.64 × 10−12
R-HSA-2219528PI3K/AKT signalling in cancer8.74 × 10−10
R-HSA-6811558PI5P, PP2A and IER3 regulate PI3K/AKT signalling9.63 × 10−10
R-HSA-3000157Laminin interactions1.56 × 10−14
R-HSA-199418Negative regulation of the PI3K/AKT network1.85 × 10−9
R-HSA-9013149RAC1 GTPase cycle3.79 × 10−5
R-HSA-8874081MET activates PTK2 signalling8.36 × 10−11
R-HSA-216083Integrin cell surface interactions1.19 × 10−10
R-HSA-1474228Degradation of the extracellular matrix8.44 × 10−10
R-HSA-1257604PIP3 activates AKT signalling1.67 × 10−5
Table 6. Known oncogenic roles of prognostic genes in GBM.
Table 6. Known oncogenic roles of prognostic genes in GBM.
Prognostic GenesOncogenic Roles in GBMReferences
IGF2
  • ↑ mRNA expression in GBM tumours → associated with shorter survival.
  • Promote growth in GBM-derived neurosphere and tumour lacking EGFR amplification via IGF2-PIK3R3 signalling axis.
[47]
RETN
  • No known oncogenic functions in GBM.
-
EDNRB
  • ↑ expression in glioma tissues and cells (C6, U251, U81).
  • Maintain GSCs migration, undifferentiation and survival via EDN3-EDNRB signalling.
  • Promotes proliferation and immune escape in glioma cells (U87).
[48,49]
C3AR1
  • ↑ mRNA expression in IDH-wildtype GBM → associated with shorter survival.
  • Hypoxia-induced C3a/C3AR1 signalling drives tumour growth and supports glioma cells (U3082MG) self-renewal → ↑ aggressiveness.
[50,51]
CLCF1
  • BRD4-activated CLCF1 plays multiple roles in cell proliferation, cell cycle progression, survival, migration and invasion capabilities of GBM cells (U87 and U251).
[52]
NTRK1
  • NTRK1 fusion genes (e.g., NFASC-NTRK1 and BCAN-NTRK1) are highly expressed in GBM → ↑ NTRK1-pathway activity.
  • NFASC-NTRK1 fusion gene confers tumorigenic functions → ↑ cell proliferation in vitro and tumour formation in vivo.
  • Possible roles in GBM tumour initiation or maintenance in fusion-positive GBMs.
[42,53]
OSMR
  • OSMR-mediated feed forward signalling mechanism with EGFRvIII and STAT3 to drive cell proliferation and tumour growth in BTSCs and GBM cells in vitro and in vivo.
  • Induced migration and invasion via OSM-STAT3 signalling → ↑ mesenchymal markers such as fibronectin and YKL-4 in GBM cells (LN-18, LN229, GBM primary culture G1)—associated with mesenchymal subtype GBM.
  • ↑ regulation of cell invasion, angiogenesis, proliferation, growth and mesenchymal transition via ANXA2-OSMR signalling axis in GBM cell lines (MGG23, U87MG) and tumour-bearing mice.
  • Confers resistance to IR via regulation of oxidative phosphorylation in GBM BTSCs.
[54,55,56,57]
KCNN4
  • KCNN4 (KCa3.1) channel activity → responsible for GBM tumour cell migration, invasion and infiltration in vivo.
  • Contributes to TMZ resistance.
[58,59]
SLC18A3
  • Co-expression of functional choline transporters, choline acetyltransferase, and vesicular acetylcholine transporters (SLC18A3) required for ACh synthesis and release in GBM cells and xenograft cell lines (U251 and PDX14).
  • ↑ invasive capability of GBM cells via AChR signalling-induced MMP-9 activity.
[6]
HTR7
  • Expression of functional 5-HT7 receptors in human glioblastoma cell lines.
  • Oncogenic roles remain elusive.
[60]
Abbreviations: EGFR—Epidermal growth factor receptor; PIK3R3—Phosphoinositide-3-kinase regulatory subunit 3; GSCs—GBM stem cells; EDN3—Endothelin 3; BRD4—Bromodomain-containing protein 4; NFASC—Neurofascin; BCAN—Brevican; STAT3—Signal transducer and activator of transcription 3; OSM—Oncostatin M; YKL-40—Chitinase-3-like-1; ANXA2—Annexin A2; IR—Ionising radiation; BTSCs—Brain tumour-initiating stem cells; TMZ—Temozolomide, ACh—Acetylcholine; MMP-9—Matrix Metallopeptidase 9; ↑—Increase or increased.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chow, M.Y.; Liew, S.S.X.; Monif, M.; Kamarudin, M.N.A.; Phon, B.W.S. A Prognostic Neuromodulation-Related Gene Signature Identifies Immunomodulation and Tumour-Associated Hallmarks in Glioblastoma. Biomedicines 2025, 13, 2640. https://doi.org/10.3390/biomedicines13112640

AMA Style

Chow MY, Liew SSX, Monif M, Kamarudin MNA, Phon BWS. A Prognostic Neuromodulation-Related Gene Signature Identifies Immunomodulation and Tumour-Associated Hallmarks in Glioblastoma. Biomedicines. 2025; 13(11):2640. https://doi.org/10.3390/biomedicines13112640

Chicago/Turabian Style

Chow, Min Yee, Sylvia Sue Xian Liew, Mastura Monif, Muhamad Noor Alfarizal Kamarudin, and Brandon Wee Siang Phon. 2025. "A Prognostic Neuromodulation-Related Gene Signature Identifies Immunomodulation and Tumour-Associated Hallmarks in Glioblastoma" Biomedicines 13, no. 11: 2640. https://doi.org/10.3390/biomedicines13112640

APA Style

Chow, M. Y., Liew, S. S. X., Monif, M., Kamarudin, M. N. A., & Phon, B. W. S. (2025). A Prognostic Neuromodulation-Related Gene Signature Identifies Immunomodulation and Tumour-Associated Hallmarks in Glioblastoma. Biomedicines, 13(11), 2640. https://doi.org/10.3390/biomedicines13112640

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop