Abstract Background Low-grade gliomas (LGG) are a heterogeneous category of brain tumors characterized by a variable clinical course, frequently associated with unfavorable prognosis and therapeutic challenges. Understanding the molecular mechanisms underlying LGG progression is crucial for improving prognosis and therapeutic strategies. This study integrates single-cell RNA sequencing and bioinformatics to explore the role of METCGs (mitochondrial electron transport chain genes) in LGG and construct a predictive model for prognosis, and through in vitro experiments, the feasibility of this model was validated. Methods We analyzed 5,691 cells and 22,947 genes from the [34]GSE117891 dataset. Using cell marker genes from the CellMarker 2.0 database and classical markers, we identified four distinct cell types: oligodendrocytes, T cells, astrocytes, and microglial cells. The METCGs profiles were calculated using various algorithms, including AUCell, UCell, ssGSEA, and others. Differentially expressed genes (DEGs) were identified and enriched for relevant pathways. Machine learning algorithms were employed to construct a prognostic risk model based on five selected METCGs. The model was validated using independent LGG cohorts. Biological pathway analyses, immune infiltration profiles, and potential drug targets were also explored. To validate the reliability of this model through experiments, functional experiments, including Blue native Page (BN-Page), western blotting, immunofluorescence, and cell viability assays, were conducted to validate SDHB expression and its role in LGG progression. Results Astrocytes exhibited the highest METCG scores, indicating their central role in mitochondrial energy regulation. The prognostic model, constructed using the StepCox[forward] + plsRcox approach, included five genes: SDHB, SDHC, SLC25A27, UQCRB, and NDUFA13. The model demonstrated high prognostic accuracy with an average C-index of 0.67 and successfully stratified LGG patients into low- and high-risk groups. High-risk patients had worse survival outcomes, with significant differences observed in KEGG pathways, immune infiltration, and metabolic processes. The low-risk group exhibited higher immune cell infiltration, including follicular helper T and monocyte cells. AZD1208_1449 was identified as a potential drug targeting high-risk patients. Additionally, SDHB expression was significantly higher in LGG cells, and knockdown of SDHB inhibited cell proliferation and invasion, supporting its role in tumor progression. Conclusion This study provides a comprehensive analysis of METCGs in LGG and develops a robust prognostic model for patient stratification. SDHB, a key subunit of Complex II, plays a crucial role in mitochondrial function and tumor progression. Our findings suggest that he high expression of SDHB in LGG contributes to maintaining elevated SDH and Complex II activity, ensuring the structural and functional integrity of mitochondrial ETC complexes. This supports the high ROS production and MMP required for the rapid growth of LGG, thereby promoting its proliferation and invasion. Thus, targeting SDHB and its associated pathways could offer new therapeutic avenues for LGG treatment. Keywords: Low-grade glioma, METCGs, SDHB, Prognostic model, Single-cell RNA sequencing Introduction Low-grade gliomas (LGGs), originating from glial cells, represent a relatively rare subset of primary intracranial tumors, accounting for approximately 15% of central nervous system (CNS) neoplasms [[35]1]. These tumors encompass diffuse oligodendrogliomas, astrocytomas, and oligoastrocytomas, as classified by the 2021 WHO Classification of Tumors of the Central Nervous System (5th Edition) [[36]2–[37]4]. While LGGs exhibit slower growth and less aggressive invasion compared to their high-grade counterparts, they carry a significant risk of malignant transformation over time, progressing to higher-grade gliomas with increased potential for neurological deterioration and mortality. The indolent nature of LGGs often results in asymptomatic early stages, delaying clinical detection until the disease has advanced to a more aggressive state [[38]5]. Although surgical resection, chemotherapy, and radiotherapy remain the mainstay of treatment for LGGs, current strategies primarily focus on managing the disease as a chronic condition and delaying its progression rather than achieving a definitive cure [[39]6]. The prognosis for patients with LGG is relatively unfavorable, with median survival typically ranging from 3 to 15 years [[40]7]. With the advancement of research and the rapid progress in molecular biology, it has become evident that distinct molecular mechanisms underlie different types of LGGs. These molecular alterations not only contribute to more accurate diagnosis and classification but also serve as valuable prognostic indicators and hold potential as novel therapeutic targets in the future. Accordingly, this study aims to comprehensively investigate the mitochondrial mechanisms contributing to the pathogenesis of LGG, providing deeper insights into its molecular underpinnings. The mitochondrial electron transport chain (ETC) consists of five large protein complexes [[41]8]. These complexes function as electron donors and acceptors, playing a crucial role in oxidative phosphorylation (OXPHOS). During this process, ADP is phosphorylated to ATP utilizing the electrochemical gradient, and mitochondrial ROS (mtROS) is generated as a by-product [[42]9]. OXPHOS is also essential for maintaining mitochondrial membrane potential (MMP) and serves as an initiator of various metabolic pathways that are important for tumor progression and invasion [[43]10]. Recent studies have revealed the mutation or dysregulation of subunits, structure alteration and dysfunction of ETC protein complexes is correlated with the activation, progression and radioresistance of glioma [[44]11–[45]15].For example, NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 4-like 2(NDUFA4L2) is a subunit of Complex I and significantly upregulated in GBM, knockdown of NDUFA4L2 GBM proliferation by protective mitophagy [[46]14]. Fractionated radiation enhances Complex IV activity through a switch in the cytochrome c oxidase subunit 4 (COX4), reducing ROS production and apoptosis while lowering labile iron and lipid peroxidation levels, ultimately promoting radioresistance [[47]13]. Mutations in complexes III and IV impair mitochondrial respiratory chain activity, inhibiting glioblastoma proliferation [[48]12]. ATP synthase subunits alpha and beta (ATP5A1 and ATP5B), part of Complex V, are significantly upregulated in glioblastoma and correlate with microvascular proliferation [[49]16]. However, the role of Complex II and its subunits in the development, proliferation, and invasion of gliomas, particularly low-grade gliomas (LGG), remains elucidated. In this study, we first found Complex II and its subunit succinate dehydrogenase complex iron sulfur subunit B (SDHB) was upregulated in LGG. Mitochondrial complex II, also known as succinate dehydrogenase (SDH), consists of four subunits encoded by the SDHx gene family: SDHA, SDHB, SDHC, and SDHD [[50]17]. Among these, SDHA and SDHB serve as catalytic subunits, whereas SDHC and SDHD function as anchoring subunits, stabilizing the complex within the mitochondrial inner membrane [[51]17, [52]18]. Complex II plays a pivotal role in cellular energy metabolism by catalyzing the oxidation of succinate to fumarate in the tricarboxylic acid (TCA) cycle, a key step in cellular respiration and ATP production [[53]18]. Dysfunction of complex II disrupts normal mitochondrial metabolism, leading to the accumulation of succinate—a metabolite that influences cellular signaling, gene expression, and oxidative stress [[54]19]. Among the causes of complex II dysfunction, mutations and dysregulation of SDHB subunit is the most prevalent. These alterations impair complex II assembly, disrupt mitochondrial oxidative phosphorylation, and rewire metabolic pathways. Consequently, they elevate mitochondrial reactive oxygen species (mtROS) production, enhance genetic instability, and contribute to tumorigenesis. SDHB mutation and dysregulation have been implicated in the initiation, progression, and invasion of several tumor types, including pheochromocytomas, paragangliomas (PPGLs), and gastrointestinal stromal tumors (GISTs) [[55]20–[56]22]. For instance, in PPGLs, SDHB mutations are associated with a higher risk of metastasis and shorter overall survival [[57]23]. Similarly, the presence of SDHB mutations in GISTs and PPGLs correlates with increased tumor aggressiveness and poor prognosis [[58]22]. Despite the established role of complex II and SDHB in these tumors, their contributions to glioma, particularly low-grade gliomas (LGG), remain poorly understood and warrant further investigation. In this study, we found SDHB and complex II was upregulated in LGG and correlated with the poor prognosis of patients with LGG. Moreover, we showed that the elevated expression of SDHB in LGG sustains high SDH and Complex II activity, preserving the structural and functional integrity of mitochondrial ETC complexes. This, in turn, facilitates increased ROS production and MMP, which are essential for the rapid growth of LGG, thereby driving its proliferation and invasion. Materials and methods Data collection W Single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq data (counts and TPM), along with baseline clinical characteristics and prognostic outcomes, were retrospectively collected from publicly available databases for patients diagnosed with low-grade gliomas (LGG). The scRNA-seq dataset [59]GSE117891 included 73 tissue samples from 14 LGG patients. For model construction and validation, data from 1,490 LGG patients were sourced from TCGA (n = 498), CGGA325 (n = 172), CGGA693 (n = 420), CGGA301 (n = 159), [60]GSE16011 (n = 98), and M-TAB-3982 (n = 142). Analysis of single-cell RNA sequencing data The scRNA-seq data were processed using the Seurat R package [[61]24]. Quality control involved excluding genes expressed in fewer than three cells, cells with fewer than 300 detected genes, and cells with > 15% mitochondrial gene content or > 0.1% hemoglobin gene content. Mitochondrial genes were removed, and doublets were excluded. Normalization was performed using the NormalizeData function, followed by centering and scaling via the ScaleData function. Highly variable genes (n = 2000) were identified for principal component analysis (PCA). Sample integration was achieved using the Harmony R package, and the first 20 principal components were utilized for clustering analysis. Cell clusters were visualized using UMAP, with clustering performed using the FindClusters function (resolution = 0.5). Cell type annotation relied on marker genes from the CellMarker 2.0 database and prior literature [[62]25]. Differentially expressed genes (DEGs) were identified using the FindAllMarkers function, with significance thresholds set at p-adjusted < 0.05 and absolute log2 fold change (log2FC) > 0.25. Enrichment analysis of METCGs in cell clusters Enrichment scores for Mitochondrial Electron Transport Chain genes (METCGs) in each cell cluster were calculated using the irGSEA.score function from the irGSEA R package [[63]26]. The wiki pathway METCGs, “WP_ELECTRON_TRANSPORT_CHAIN_OXPHOS_SYSTEM_IN_MITOCHONDRIA.v2024.1 “, were retrieved from the MSigDB database ([64]https://www.gsea-msigdb.org/gsea/msigdb/human/geneset/WP_ELECTRON_ TRANSPORT_CHAIN_OXPHOS_SYSTEM_IN_MITOCHONDRIA.html). Enrichment algorithms (AUCell, UCell, ssGSEA, Singscore, JASMINE, and viper) were applied to normalized RNA-seq data. The GSVA R package [[65]27] was employed to assess pathway activity in astrocyte clusters based on METCG-associated DEGs. Identification of prognosis-associated genes Univariate Cox regression analysis within the TCGA-LGG cohort identified prognostic genes. Upregulated METCGs in the astrocyte cluster were extracted using the FindAllMarkers function. Prognostic METCGs were determined through intersection analysis. Model construction and validation To develop the optimal prognostic model, 101 machine learning algorithm combinations were evaluated using the Mime package (https://github.com/l-magnificence/Mime) [[66]28]. These combinations included both single algorithms (e.g., StepCox[forward] and partial least squares regression for Cox [plsRcox]) and hybrid models that combined two distinct algorithms for feature selection and modeling (e.g., StepCox[forward] + plsRcox), as previously described [[67]29]. First, univariate Cox regression (p < 0.05) identified prognostic genes in the the training set. Then, 101 machine learning algorithm combinations were trained using 10-fold cross-validation and tested in the other independent LGG cohorts. Final, the model with the highest average C-index was selected, identifying a five-gene signature (SDHB, SDHC, SLC25A27, UQCRB, NDUFA13). We compared our optimal model against 95 published signatures (33 for glioma/22 for LGG) derived from Liu’s study [[68]28] and clinical characteristics using C-index. Kaplan-Meier curve analysis was used to evaluate the overall survival difference between high-risk and low-risk groups based on median cutoff of each of cohorts. Model-related plot was generated via the Mime package. Biological pattern evaluation Differential gene expression analysis was performed using the DESeq2 algorithm [[69]30] to identify DEGs between high-risk and low-risk groups in the TCGA-LGG cohort, with thresholds of log2 fold change (log2FC) > 1 and adjusted p-value < 0.05. Pathway enrichment analysis utilized KEGG, HALLMARK, and GO gene sets from MSigDB ([70]https://www.gsea-msigdb.org/gsea/msigdb/index) via Gene Set Enrichment Analysis (GSEA), with significance set at adjusted p-value < 0.05 and absolute normalized enrichment score (NES) > 1. GO analysis focused on biological processes (BP), cellular components (CC), and molecular functions (MF). Gene mutation analysis identified somatic and chromosomal region-associated mutations to uncover potential therapeutic targets. Immune cell infiltration was assessed using the Cibersort algorithm and IOBR R package [[71]31], analyzing 22 immune cell types and comparing immune activation, suppression factors, and MHC-related molecule expression between risk groups. Results were visualized with the BEST tool ([72]https://rookieutopia.hiplot.com.cn/app_direct/BEST/). Identification of potential therapeutic agents based on risk model stratification Drug sensitivity was evaluated using IC50 values of 198 compounds across five LGG cohorts, predicted via the OncoPredict R package [[73]32] and GDSC2 database [[74]33]. IC50 comparisons between risk groups were complemented by ROC curve analysis to assess predictive accuracy. Potential therapeutic agents were identified through intersection analysis of drugs with significant distribution differences. Molecular Docking analysis PDB files were obtained from the Protein Data Bank (RCSB PDB, [75]https://www.rcsb.org/structure/). Compound structures were downloaded in sdf format from the PubChem database ([76]https://pubchem.ncbi.nlm.nih.gov/). The cavity-detection guided blind docking algorithm [[77]34] (CB-Dock2) website ([78]https://cadd.labshare.cn/cb-dock2/index.php) was used to perform molecular docking analysis between SDHB and AZD1208. Nomogram scoring system Significant factors from univariate Cox regression (p < 0.05) were incorporated into a multivariate model to construct a nomogram. Predictive performance was evaluated using AUC, time-dependent C-index, and calibration plots comparing predicted versus actual overall survival outcomes. Cell culture HEB and LGG cell lines (HS683 and SHG-44) were cultured in DMEM (Gibco, C11330500BT) supplemented with 10% fetal bovine serum (FBS, Excell Bio, FSP500) and 1% penicillin-streptomycin (Gibco) at 37 °C in a humidified atmosphere with 5% CO₂. Upon reaching 70–80% confluence, cells were washed with DPBS (calcium- and magnesium-free) and detached with 500–1000 µL of trypsin for 1–2 min at 37 °C. Trypsinization was halted with DMEM containing 10% FBS, followed by centrifugation at 1000 rpm for 5 min. The cell pellet was resuspended in fresh medium, transferred to new culture dishes, and incubated for further growth. SiRNA transfection Cells were seeded in 6-well plates and grown to 40–50% confluence. siRNA (Sangon Biotech, China) was prepared in DEPC-treated water (20 µM) and complexed with RNATransMate (Sangon Biotech, E607402) in serum- and antibiotic-free DMEM. After a 10-minute incubation at room temperature, the complexes were added to cells and distributed by gentle rocking. Following 8 h of incubation at 37 °C with 5% CO₂, the medium was replaced with complete medium. Transfected cells were cultured for 24–48 h before further analysis. siRNA sequences were as follows: siRNA#2: F-UCUUGAUCUCUGCAAUAGC, R-GCUAUUGCAGAGAUCAAGA; siRNA#3: F-UCAAAUCGGGAACAAGAUC, R-GAUCUUGUUCCCGAUUUGA. Cell migration and proliferation assays For migration assays, cells were trypsinized, centrifuged at 1000 rpm for 5 min, and resuspended in serum-free medium. A total of 5 × 10⁴ cells in 300 µL serum-free medium were seeded into the upper chamber of Transwell inserts (Corning, 3422), with 500 µL DMEM containing 20% FBS in the lower chamber. After 24–48 h, migrated cells were fixed with paraformaldehyde, stained with 0.1% crystal violet (Abiowell, AWI0364a), and counted in five random microscopic fields. For proliferation assays, 5 × 10³ cells were seeded into 96-well plates and treated with CCK-8 reagent (Beyotime, C0046) according to the manufacturer’s protocol. Absorbance was measured at 450 nm using a microplate reader 2 h post-treatment. Proliferation was assessed over time by repeating the CCK-8 assay at 24-hour intervals. SDS-PAGE and Western blotting Cells were lysed with 4× SDS sample buffer containing 1 mM phosphatase inhibitor (Absin, abs9162) and 1 mM PMSF (Beyotime, ST507-10 ml). Protein lysates were separated by SDS-PAGE at 150–200 V for 60 min and transferred to methanol-activated PVDF membranes (Vazyme, E801-01). Membranes were blocked with 5% non-fat milk in TBST for 1 h and incubated overnight at 4 °C with primary antibodies diluted in 1% BSA. After washing with TBST, membranes were incubated with HRP-conjugated secondary antibodies (Sangon, D123504) for 3 h at room temperature. Protein bands were visualized using ECL substrate (Abbkine, BMU102-CN) and imaged using a chemiluminescence detection system. Primary antibodies included anti-SDHB (Proteintech, 10620-1-AP), SDHA (Promab, 32308), anti-GAPDH (Proteintech, 60004-1-Ig). Flow cytometry Cells were incubated with 5 µM MitoSox Red (Thermo, [79]M36008) at 37 °C for 30 min, protected from light. After washing twice with PBS, cells were trypsinized, resuspended in PBS, and analyzed using a flow cytometer. Data were processed with FlowJo software to quantify mitochondrial ROS levels. Controls included untreated cells and cells treated with Tempo (KKLMED, KM17362). Immunohistochemistry Tumor and adjacent tissues were formalin-fixed, paraffin-embedded, and sectioned (4–5 μm). Sections were deparaffinized, rehydrated, and subjected to antigen retrieval in citrate buffer. After blocking with 5% goat serum, sections were incubated overnight at 4 °C with primary antibodies. Following washing, biotinylated secondary antibodies were applied, and sections were developed using DAB substrate (Sigma, D4418). Hematoxylin counterstaining was performed, and protein expression was assessed microscopically. Immunofluorescence staining HEB and LGG cells were incubated with 50 nM MitoTracker Red (ThermoFisher, M7512) at 37 °C for 30 min. After fixation with 4% paraformaldehyde and permeabilization with 0.1% Triton X-100, cells were blocked with 1% BSA in PBST and incubated overnight at 4 °C with primary antibodies. Following washing, secondary antibodies were applied for 3–4 h at room temperature in the dark. Nuclei were counterstained with Hoechst 33,258, and slides were mounted for imaging. ATP level detection ATP levels were measured using the Enhanced ATP Detection Kit (Beyotime, S0027). Lysates were prepared, centrifuged, and supernatants were mixed with ATP detection reagent. Luminescence was measured using a luminometer, and relative light units (RLU) were recorded to quantify ATP concentrations. SDH activity assay SDH activity was assessed using the Succinate Dehydrogenase Activity Assay Kit (Boxbio, AKAC011M). Cell lysates were prepared, and enzyme activity was measured by recording absorbance changes at 600 nm over time. Final SDH activity was calculated after adjusting for background values. Mitochondrial respiratory chain complex II activity assay Complex II activity was measured using the Mitochondrial Respiratory Chain Complex II Activity Detection Kit (Boxbio, AKOP006M). Mitochondria were isolated, and enzyme activity was determined by monitoring absorbance changes at 605 nm. Results were adjusted for background to calculate activity. Blue-native PAGE Cells were harvested from 10 cm dishes, trypsinized, and centrifuged at room temperature for 5 min. The pellet was resuspended in 1 mL cold Digitonin buffer (1% Digitonin, 20 mM Tris-HCl pH 7.4, 0.1 mM EDTA, 50 mM NaCl, 10% glycerol, 1 mM PMSF, 1 mM PI) and incubated on ice for 30 min with gentle pipetting (10 repetitions). The lysate was centrifuged at maximum speed for 15 min at 4 °C, and the supernatant was collected, discarding the insoluble pellet. Blue-Native PAGE was performed at 600 V with the apparatus connected to a cooling pump to maintain low temperatures. Once the samples entered the separating gel, the buffer was replaced with 1× Cathode buffer (without Coomassie G-250), and electrophoresis continued for 3 h. Proteins were transferred to a PVDF membrane pre-activated with methanol for 1 min and rinsed in PBS. The transfer was carried out at 300 mA for 3 h using transfer buffer. The membrane was blocked with 5% non-fat milk in TBST for 60 min and incubated overnight at 4 °C with primary antibodies, including NDUFS1 (Sangon, D122747-0025), UQCRC1 (Sangon, D123504-0025), ATP5A1 (Proteintech, 14676-1-AP), MTCO2 (Proteintech, 55070-1-AP), and SDHB (Proteintech, 10620-1-AP). The following day, the membrane was incubated with the corresponding secondary antibody (Sangon, D123504), and protein bands were detected using chemiluminescence. Results Cell type annotation and METCG score calculation The workflow of this study is outlined in Fig. [80]1A. From the [81]GSE117891 dataset, we analyzed 5,691 cells and 22,947 genes. Using common brain tissue cell marker genes from the CellMarker 2.0 database and classical markers, we annotated four distinct cell types: oligodendrocytes (MBP, MOP, PLP1), T cells (CD3D, CD3E, CD8A), astrocytes (GFAP, AQP4, SOX9), and microglial cells (TMEM119, CX3CR1, P2RY12). The distribution of these cells was as follows: oligodendrocytes (858 cells, 15.1%), T cells (405 cells, 7.1%), astrocytes (2,876 cells, 50.5%), and microglial cells (1,552 cells, 27.3%). The cell type distribution was visualized using UMAP and DotPlot analyses (Figs. [82]1B-C). Fig. 1. [83]Fig. 1 [84]Open in a new tab Analysis of METCGs in Glioma Single-Cell RNA Sequencing and Their Biological Significance.(A) Study flowchart. LGG = Low-grade glioma. (B) UMAP plot illustrating the distinct clustering patterns of four predominant cell lineages. (C) Dot plot depicting the expression levels of marker genes across the four major cell lineages. (D) Distribution of METCG activity scores across different clusters, calculated using six distinct algorithms. (E) Results of Gene Ontology (GO) analysis, with the horizontal axis representing -Log10 (adjusted p-value). (F) Bar plot showing KEGG pathway enrichment and the top five cluster-related differentially expressed genes To examine the METCG profiles of each cell type, glycosylation scores were calculated based on the expression of genes involved in the “WP_ELECTRON_TRANSPORT_CHAIN_OXPHOS_SYSTEM_IN_MITOCHONDRIA” pathway. By employing multiple algorithms (AUCell, UCell, ssGSEA, Singscore, JASMINE, and viper), we found that astrocytes exhibited the highest METCG scores compared to other cell types (Fig. [85]1D; Table 1, all adjust p value < 0.001). These findings suggest that METCGs play a critical role in astrocyte function. Furthermore, KEGG pathway enrichment analysis (Fig. [86]1E) revealed that these DEGs were significantly associated with nucleocytoplasmic transport and neurodegenerative disease pathways, emphasizing the role of astrocytes in neurodegenerative processes. Identification of prognosis-associated genes To construct an effective prognostic model, we evaluated 101 combinations of machine learning algorithms using the Mime package ([87]https://github.com/l-magnificence/Mime). These combinations included single algorithms (e.g., StepCox[forward] and partial least squares regression for Cox [plsRcox]) and hybrid models combining two distinct algorithms for feature selection and modeling (e.g., StepCox[forward] + plsRcox) [[88]29]. The TCGA cohort served as the training set, while additional independent cohorts were used for validation. For each cohort, the C-index was computed, and the model with the highest average C-index was selected. Ultimately, the StepCox[forward] + plsRcox model emerged as the optimal prognostic risk score model, identifying five key genes: SDHB, SDHC, SLC25A27, UQCRB, and NDUFA13. Kaplan-Meier survival analysis, univariate and multivariate Cox regression analyses, time-dependent receiver operating characteristic (ROC) curves, and the C-index were employed to evaluate the model’s prognostic accuracy. Comparative analysis with previously published glioma prognostic models across six independent LGG cohorts confirmed the model’s robustness and generalizability, as evidenced by superior hazard ratios (HRs) and C-index values. Construction of the prognostic risk model The StepCox[forward] + plsRcox model demonstrated strong predictive power and generalization across six cohorts, with an average C-index of 0.67 (Figure S1, Fig. [89]2B). Meta-analysis confirmed the model’s significance as a prognostic factor (all p < 0.05, Fig. [90]2 C). Patients were categorized into low- and high-risk groups based on median risk scores. Low-risk patients exhibited significantly better survival outcomes compared to high-risk patients (all p < 0.05, Fig. [91]2D). ROC analysis demonstrated the model’s predictive performance, achieving AUC values of 0.77 at one year, 0.74 at three years, and 0.72 at five years (Fig. [92]2E). Fig. 2. [93]Fig. 2 [94]Open in a new tab Prognostic Gene Selection and Model Construction.(A) Venn diagram demonstrating the intersection of astrocyte subtype-specific METCG-related differentially expressed genes, the METCG set, and common genes identified across six LGG cohorts. (B) C-index comparison of different machine learning models across cohorts, with the StepCox[forward] + plsRcox model achieving the highest mean C-index. (C) Meta-analysis illustrating the hazard ratio distribution across six LGG cohorts. (D) Kaplan-Meier survival analysis comparing high- and low-risk groups, based on the StepCox[forward]+ plsRcox model, with median survival times shown. P-values were calculated using the Log-rank test. (E) ROC curve analysis demonstrating the predictive performance of the StepCox[forward] + plsRcox model at 1, 3, and 5 years Evaluation of model generalization ability To assess robustness, the StepCox[forward] + plsRcox model was compared to existing prognostic models. Univariate Cox regression analysis confirmed its reliability (Fig. [95]3 A), while consistent C-index values across six cohorts (Fig. [96]3B) indicated stable performance. The model outperformed existing clinical and molecular markers in predictive accuracy (Fig. [97]3 C), validating its generalizability and prognostic value. Fig. 3. [98]Fig. 3 [99]Open in a new tab Evaluation of Model Generalization.(A) Univariate Cox regression analysis comparing the prognostic power of the proposed model to previously established LGG models. (B) Generalized C-index assessment across six cohorts, highlighting the stable performance of the proposed model in predicting overall survival compared to most other prognostic models. (C) Bar plot comparing the C-index values of the proposed model with various clinical and molecular markers. PQ = 1p19q co-deletion; IDH = IDH mutation Exploration of biological pathways based on risk model Pathway enrichment analyses revealed significant biological alterations associated with risk stratification. KEGG analysis indicated that high-risk patients were enriched in pathways related to oxidative phosphorylation, neurodegenerative diseases, and pyruvate metabolism. Hallmark analysis identified associations with oxidative phosphorylation, adipogenesis, and fatty acid metabolism. GO pathway analysis highlighted processes including ATP metabolism, the electron transport chain, and mitochondrial respiratory chain complex assembly (Fig. [100]4 A, p < 0.01). Subpopulation analyses corroborated these findings (Fig. [101]4B, adjusted p < 0.01). Additionally, high-risk patients exhibited significant mutations in CTNNB1 and APOB7, as well as chromosomal aberrations such as 5q35.3 gain and 8p23.2 loss (Fig. [102]4 C, p < 0.05). Fig. 4. [103]Fig. 4 [104]Open in a new tab Biological Function Analysis of the Model.(A) GSEA pathway enrichment analysis comparing high- and low-risk groups using KEGG, Hallmark, and GO gene sets. (B) GO term enrichment analysis showing biological processes, cellular components, and molecular functions enriched in the high-risk group (red) and low-risk group (blue). (C) Mutation analysis identifying key somatic mutations and chromosomal regions associated with high-risk patients. (D) Box plots illustrating the relative distribution of immune cells across six LGG cohorts, analyzed using the CIBERSORT algorithm.(E) Correlation analysis between immune modulators and the model-derived risk scores across six LGG cohorts Immune infiltration analysis revealed distinct microenvironmental characteristics between risk groups. The low-risk group demonstrated higher infiltration of follicular helper T cells and monocytes, while the high-risk group was associated with M2-type macrophages (Fig. [105]4D). Furthermore, the low-risk group exhibited a “hot” immune microenvironment, characterized by increased expression of immune stimulators, MHC pathways, and immune inhibitors (Fig. [106]4E). Identification of potential targeted drugs based on the model Drug sensitivity analysis using IC50 values from the GDSC2 database identified AZD1208_1449 as a potential targeted therapy, consistently validated across five LGG cohorts with an AUC > 0.65 (Fig. [107]5A-B). ROC curve analysis demonstrated the predictive efficacy of the model for drug sensitivity (AUC range: 0.68–0.83, Fig. [108]5 C). Low-risk patients were more sensitive to AZD1208_1449, as indicated by lower IC50 values compared to high-risk patients. Molecular docking predicts that AZD1208 targets SDHB by binding its catalytic pocket with high affinity (Vina score: −8.9 kcal/mol), forming interactions with key electron transfer residues (HIS270/HIS363) and substrate-binding residue ASP74 (Table 2), potentially inhibiting succinate binding or disrupting [2Fe-2 S] cluster function to block SDHB activity (Figure S2). Fig. 5. [109]Fig. 5 [110]Open in a new tab Drug Sensitivity Analysis Based on METCGs. (A) Flowchart of the drug selection process. GDSC2 = Genomic Data Sharing for Cancer Research, version 2; AUC = area under the curve; IC50 = half-maximal inhibitory concentration. (B) ROC curves evaluating the predictive performance of the model in identifying AZD1208_1449 sensitivity across six cohorts. (C) Bar plot comparing the calculated IC50 values between high- and low-risk groups Construction of the prognostic nomogram To enhance clinical utility, we performed univariate and multivariate Cox regression analyses to identify significant predictors of overall survival (OS) (Fig. [111]6 A, all p < 0.001). A nomogram incorporating model risk group, age, IDH mutation status, 1p19q co-deletion status, and WHO grade was developed to predict OS (Fig. [112]6 C). The nomogram outperformed other clinical predictors for 1-, 3-, and 5-year survival rates (Fig. [113]6D, DeLong test p < 0.05). Time-dependent C-index curves (Fig. [114]6E) and calibration plots (Fig. [115]6 F) further validated the nomogram’s accuracy and predictive performance. Fig. 6. [116]Fig. 6 [117]Open in a new tab METCG-Based Nomogram for Prognostic Evaluation in LGG Patients.(A-B) Univariate and multivariate Cox regression analyses comparing the model-derived risk groups with other clinical variables across LGG cohorts. (C) Nomogram plot integrating the model-derived risk groups with other clinical factors to generate predictive scores. (D) ROC curve analysis comparing the predictive performance of the nomogram and other variables for overall survival at 1, 3, and 5 years. (E) Time-dependent concordance index demonstrating the predictive performance of the nomogram compared to other clinical variables. (F) Calibration curves evaluating the accuracy of the nomogram in predicting 1-, 3-, and 5-year overall survival SDHB expression and its functional role in LGG progression and invasion To robustly assess the reliability of this prognostic model, we selected SDHB, one of the five candidate genes, for further single-cell analysis and in vitro experimental validation. The UMAP plot (Fig. [118]7 A) revealed the heterogeneous distribution of SDHB expression across cell types of LGG, with astrocytes showing the highest expression levels and proportions, as indicated by the accompanying dot plot (Fig. [119]7B). Kaplan-Meier survival analyses using CGGA_693 and TCGA_LGG datasets demonstrated that higher SDHB expression correlates with poorer overall survival in glioma patients (Figs. [120]7 C, D; log-rank p < 0.0001). Immunohistochemistry (IHC) of LGG tissue and adjacent peritumor tissue confirmed a significant elevated expression of SDHB in tumor regions (Fig. [121]7E). Immunofluorescence staining further demonstrated SDHB localized mitochondria of LGG cells, with its expression being significantly higher than normal astrocytes (HEB) cells (HS683, SHG-44; Figs. [122]7 F, G). BN-PAGE also confirmed that SDHB forms protein complexes in LGG cells, with the expression levels of these protein complexes significantly higher than those observed in HEB cells (Fig. [123]7H). Western blot analysis validated this differential expression at the protein level across HEB and LGG cells (Fig. [124]7I). The above results suggest that SDHB and Complex II may play a crucial role in the growth and invasion of LGG. Fig. 7. [125]Fig. 7 [126]Open in a new tab SDHB was upregulated in LGG and correlated with invasion and proliferation. (A) UMAP visualization of SDHB expression across different clusters. Each dot represents an individual sample, with the color intensity corresponding to SDHB expression levels (scale on the left). (B) Dot plot depicting the average expression of SDHB across various cell types, including astrocytes, microglia, oligodendrocytes, and T cells. The size of each dot reflects the percentage of cells expressing SDHB, while the color indicates the average expression level (scale at the bottom right). (C-D) Kaplan-Meier survival curves for patients with high (blue) and low (yellow) SDHB expression in the CGGA 693 and TCGA LGG cohorts. The log-rank test p-value is indicated, showing a significant difference in overall survival between the two groups. (E) Immunohistochemical staining of tumor and peritumoral tissues from three LGG patients revealed a marked upregulation of SDHB in the tumor tissues. Scale Bar=50μm. (F) Immunofluorescence analysis showed that the fluorescence intensity of SDHB in LGG cells was significantly higher than that in HEB cells.(G) Quantitative analysis of the green fluorescence intensity of SDH was performed on 30 cells in Figure 7F. ****P < 0.0001, n=30. (H-I) The results of BN-PAGE and SDS-PAGE indicated elevated expression levels of Complex II(H) and SDHB(I) in LGG and GBM cells. (J-K) Three siRNAs were used to knock down SDHB in HS683 and SHG44 cells, and SDS-PAGE confirmed that siRNA#2 and siRNA#3 achieved higher knockdown efficiency. (L-M) After knocking down SDHB in SHG-44 cells with siRNA#2 and siRNA#3 for 48 hours, Transwell (L-N) and CCK-8(O-P) assays confirmed that the knockdown of SDHB significantly inhibited the invasion and growth of LGG cells. ****P < 0.0001 Next, we used three different siRNAs to knock down SDHB in LGG cells and verified the knockdown efficiency through SDS-PAGE. siRNA#2 and siRNA#3 demonstrated higher knockdown efficiency. Therefore, we proceeded with further knockdown of SDHB in LGG cells using siRNA#2 and siRNA#3, and assessed the impact of SDHB knockdown on LGG cell growth and invasion through CCK-8 and Transwell assays. These results indicate that knockdown of SDHB in LGG cells (HS683 and SHG-44) leading to decreased cell invasion as shown by transwell assays (Fig. [127]7L, M, N) and significantly decreased proliferation rates (Figs. [128]7O, P). These findings suggest that SDHB can acts as a critical biomarker of glioma progression and invasion, potentially through its role in mitochondrial function. Knockdown of SDHB impairs mitochondrial function and alters metabolic activity in LGG cells To investigate the role of SDHB in mitochondrial function, we knocked down SDHB in HS-683 and SHG-44 cells using two siRNAs. Immunofluorescence analysis revealed a marked reduction in SDHB protein levels upon siRNA transfection, as evidenced by diminished SDHB staining (green) and disrupted mitochondrial structure, as visualized by Mitotracker staining (red) (Fig. [129]8 A and B). Quantification of Mitotracker fluorescence intensity demonstrated a significant increase in mitochondrial membrane potential (MMP) in siRNA-transfected cells compared to control siRNA (siCtrl)-treated cells (Fig. [130]8 C and D). Flow cytometry analysis of mitochondrial membrane potential, using Mitotracker fluorescence, further confirmed these findings, showing Increased MMP in SDHB-knockdown cells (Fig. [131]8E and F). To assess mitochondrial reactive oxygen species (ROS) levels, we also utilized MitoSOX staining. SDHB-knockdown cells exhibited significantly elevated mitochondrial ROS levels compared to siCtrl-treated cells (Fig. [132]8G and H), highlighting increased oxidative stress upon SDHB depletion. Fig. 8. [133]Fig. 8 [134]Open in a new tab Knockdown of SDHB impairs mitochondrial ETC complexes and alters metabolic activity in LGG cells. (A-D) siRNA#2 and siRNA#3 were utilized to knock down SDHB in HS683 and SHG-44 cells. Immunofluorescence staining was conducted to assess Mitotracker (red fluorescence), SDHB (green fluorescence), and mitochondrial morphology (A-B). Quantitative analysis of fluorescence intensity indicated that knockdown of SDHB led to a significant increase in fluorescence intensity in both HS683 (C) and SHG-44(D) cells. ****P < 0.0001, n=30. (E-H) Flow cytometric analysis was performed to evaluate the changes in fluorescence intensity of Mitotracker and MitoSOX in HS683 and SHG-44 cells following SDHB knockdown. (I-K) The Luciferase ATP Assay Kit was used to measure changes in ATP levels in HEB cells, LGG cells, and SDHB knockdown LGG cells. ** P <0.01, **** P < 0.0001, n=8. (L-N) The Succinate Dehydrogenase (SDH) Activity Assay Kit was used to measure changes in SDH activity in HEB cells, LGG cells, and SDHB knockdown LGG cells. ** P <0.01, **** P < 0.0001, n=8. (O-Q) The Mitochondrial Respiratory Complex II Activity Assay Kit was used to measure changes in Complex II activity in HEB cells, LGG cells, and SDHB knockdown LGG cells. **** P < 0.0001, n=8. (R-S) SDS-PAGE(R) and BN-PAGE(S) were performed to detect the expression of the five mitochondrial respiratory chain complexes in HS683 and SHG-44 cells following SDHB knockdown Next, we aimed to investigate whether the expression level of SDHB in LGG affects ATP production, SDH activity, and Complex II (succinate dehydrogenase, SDH) activity. We found that ATP levels were significantly higher in LGG cells (Fig. [135]6I). Additionally, SDHB knockdown inhibited ATP production in LGG cells (Figs. [136]6 J, K). We also assessed the enzymatic activities of SDH, which were markedly reduced in SDHB-low expressed cells, as demonstrated in both the HS-683 and SHG-44 cell models (Figs. [137]8L-N). In contrast, SDHA activity remained unaffected (Figure S3). As one of the most important subunits of Complex II, we hypothesized that SDHB knockdown would also inhibit Complex II (SDH) activity. Indeed, Complex II activity was significantly increased in SDHB-high expressed LGG cells (Fig. [138]8O), while SDHB knockdown reduced Complex II activity in both HS-683 and SHG-44 cell models (Figs. [139]8P-Q). Thus, we can conclude that high expression of SDHB promotes ATP production, SDH and Complex II activity in LGG cells. In summary, we conclude that SDHB knockdown increases oxidative stress and significantly impairs Complex II in LGG cells (Figs. [140]8A-Q), leading to widespread disruption of the electron transport chain (ETC). To further investigate the underlying mechanisms, we used SDS-PAGE and BN-PAGE to examine the expression and assembly of the five ETC complexes. Western blot analysis confirmed the efficient knockdown of SDHB and revealed concomitant reductions in the protein levels of several ETC components, including NDUFS1 (Complex I), SDHA (Complex II), UQCRC1 (Complex III), MTCO2 (Complex IV), and ATP5A1 (Complex V) in siRNA-transfected cells (Fig. [141]8R). Additionally, BN-PAGE further substantiated that SDHB knockdown led to a decrease in all five complexes, indicating compromised assembly and activity of the ETC complexes in SDHB-deficient cells (Fig. [142]8S). These results demonstrate that SDHB knockdown leads to mitochondrial dysfunction, reduced ATP production, elevated ROS levels, and impaired activities of respiratory complexes in glioma cells. These findings highlight the critical role of SDHB in maintaining mitochondrial integrity of ETC and metabolic homeostasis. Discussion This study represents a pioneering integration of single-cell sequencing and advanced bioinformatics to elucidate the distribution, functionality, and prognostic significance of METCGs in LGG. Leveraging single-cell RNA sequencing from the [143]GSE117891 dataset, we accurately annotated four distinct cell types in brain tissue, including astrocytes, oligodendrocytes, T cells, and microglial cells, using a combination of CellMarker 2.0 and classical markers. The application of UMAP and DotPlot visualizations further enabled a comprehensive understanding of cell-specific characteristics, particularly highlighting astrocytes as the dominant cell type with unique metabolic profiles. Innovatively, we employed multiple scoring algorithms, including AUCell, UCell, and ssGSEA, to calculate METCGs scores, revealing astrocytes’ metabolic dominance in oxidative phosphorylation pathways. To validate whether this dominance was statistically significant, we conducted comprehensive inter-group comparisons across all scoring algorithms. As shown in Table 1, astrocytes exhibited significantly higher METCG scores compared to oligodendrocytes, T cells, and microglia (all adjusted p-values < 0.001). These results remained consistent across AUCell, UCell, ssGSEA, Singscore, JASMINE, and viper, confirming the robustness of our scoring framework. This significant enrichment suggests that astrocytes may serve as a key metabolic hub within the LGG microenvironment, with elevated oxidative phosphorylation activity potentially contributing to tumor-supportive functions. These findings underscore the power of single-cell analysis in revealing intratumoral heterogeneity. In parallel, we applied advanced machine learning (StepCox[forward] + plsRcox) to build a robust prognostic model, which demonstrated superior accuracy and generalizability across six LGG cohorts. Pathway and immune profiling further revealed distinct metabolic and immunological features between risk groups. Together, this work establishes a novel framework for LGG stratification and highlights the clinical potential of integrating single-cell and bioinformatics approaches. To further validate the reliability of the METCGs prognostic model in predicting potential diagnostic and therapeutic molecular targets in LGG, we conducted additional confirmation through cellular experiments. The model is composed of five genes, including SDHB, SDHC, SLC25A27, UQCRB, and NDUFA13, each contributing to various mitochondrial functions. SDHB and SDHC are subunits of Complex II, while the others belong to Complexes I and III or are involved in mitochondrial energy regulation [[144]17, [145]35, [146]36]. The role of Complex II in tumors is complex and context-dependent: it can promote tumor progression through succinate accumulation and activation of HIF-1α [[147]37], while also inhibiting tumor growth by maintaining mitochondrial function and inducing apoptosis [[148]17]. Therefore, a deeper understanding of the specific mechanisms through which Complex II regulates tumorigenesis in LGG is crucial for the development of novel metabolism-targeted therapeutic strategies. Thus, we selected SDHB for further study. SDHB is a key subunit of Complex II in the electron transport chain (ETC). It contributes to the proper functioning of mitochondrial oxidative phosphorylation, which is essential for ATP production and maintaining cellular energy homeostasis [[149]38]. In this study, we first observed that SDHB and Complex II were upregulated in LGG. Additionally, knockdown of SDHB inhibited the proliferation and invasion of LGG cells. Notably, SDHB exhibited the highest expression levels and proportions in astrocytes. In conclusion, our findings suggest that the elevated expression of SDHB and Complex II may be correlated with tumorigenesis and could serve as potential therapeutic targets for LGG. However, the underlying mechanisms by which SDHB modulates Complex II assembly, thereby promoting LGG proliferation and invasion, remain to be elucidated. To further investigate the mechanism by which SDHB affects mitochondrial function and inhibits LGG growth and invasion through disrupt the structure of the mitochondrial respiratory chain, we knocked down SDHB in two LGG cell lines and explored the effects using immunofluorescence, flow cytometry and BN-PAGE techniques. Our findings reveal that knockdown of SDHB impairs mitochondrial integrity, leading to a significant increase in oxidative stress, disrupted MMP, and elevated ROS levels. Notably, we also demonstrate that SDHB depletion reduces the activity of SDH and Complex II, indicating a global impairment of mitochondrial respiration chain. To further validate this hypothesis, we utilized BN-PAGE and SDS-PAGE to examine the assembly of the five mitochondrial ETC complexes and the expression levels of their key subunits. The results revealed widespread dysfunction across the ETC complexes, with a pronounced impact on Complex II. In summary, we propose the scientific hypothesis that the high expression of SDHB in LGG contributes to maintaining elevated SDH and Complex II activity, ensuring the structural and functional integrity of mitochondrial ETC complexes and ATP production. This supports the high ROS production and MMP required for the rapid growth of LGG, thereby promoting its proliferation and invasion. The novelty of this study lies in the comprehensive assessment of SDHB’s impact on both mitochondrial structure and function in LGG cells. While previous studies have linked SDHB to tumorigenesis and metabolic reprogramming [[150]38–[151]40], our work specifically addresses how SDHB knockdown affects the assembly and activity of all five mitochondrial ETC complexes, a mechanism not yet fully elucidated in LGG research. Additionally, our findings that SDHB depletion compromises the assembly of the ETC and impairs mitochondrial energy production provide new insights into the metabolic vulnerabilities of glioma cells. This underscores the potential of targeting SDHB or its associated pathways as a therapeutic strategy to disrupt mitochondrial function and limit tumor progression in LGG. These results contribute to the growing body of evidence linking mitochondrial ETC structure and functions to cancer biology, highlighting the potential for targeting METCGs in LGG therapy. From a clinical perspective, our findings hold significant translational implications. The METCG-based risk model demonstrates reliable prognostic value and may serve as a powerful tool for stratifying LGG patients and guiding individualized treatment planning. Moreover, SDHB emerges not only as a biomarker of poor prognosis but also as a promising therapeutic target. Our data suggest that pharmacological inhibition of SDHB, or disruption of Complex II function, may impair the metabolic fitness of LGG cells and suppress tumor growth. The identification of AZD1208_1449 as a candidate small molecule with selective efficacy in low-risk groups further supports the feasibility of developing mitochondria-targeted therapies based on the metabolic status of individual tumors. Further investigation into the downstream signaling pathways affected by SDHB depletion and their role in glioma progression could pave the way for novel therapeutic interventions targeting mitochondrial metabolism. Acknowledgements