3.1 Identification of apoptosis-related genes in glioma patients
We treated glioma cell line U251 with different concentrations of doxorubicin (0, 100, 200, 500 nM) for four days to induce apoptosis in Supplementary Figure S2. After sequencing, 1626 differentially expressed apoptosis-related genes were acquired. We obtained 667 glioma samples from the TCGA database. The expression data of these genes were obtained from TCGA database. Through univariate regression analysis, we obtained 218 apoptosis-related genes associated with prognosis. By LASSO regression analysis, there were 27 apoptosis-related genes (Fig. 1A and 1B). In order to study apoptosis and immunity, we finally screened out four genes related to T lymphocytes.
3.2 Accessing prognostic merit of the risk signature
After screening, we obtained the most suitable 4 apoptosis-related and T-cell-related genes with prognostic value, which are CREM, TNFSF12, PEA15 and PRKCD (Fig. 1C). The survival analysis was displayed in Supplementary Figure S3. The risk characteristics of 667 glioma patients were constructed, and then computed the risk scores of the patients. On the basis of median score, 667 patients were classified into the high-risk and low-risk groups (Supplementary Table S1 and S2). In the high-risk group, CREM, TNFSF12 and PRKCD were upregulated, while PEA15 was downregulated. Kaplan-Meier (KM) curves demonstrated that the OS rate of the high-risk group was dramatically lower than the overall survival rate of the low-risk group (p < 0.001) (Fig. 1D and 1G). The risk curve and scatterplot revealed that the death rate of the high-risk group was higher than the death rate of the low-risk group (Fig. 1E and 1H). Time-varying receiver operating characteristic (ROC) curves were used to verify the predictive significance of the risk signatures, and the AUC values of the 1-, 3- and 5-year survival rates in the TCGA database were 0.881, 0.929, and 0.905, respectively (Fig. 1F). Furthermore, we validated the efficacy of this risk signature in the CGGA database, and the same results were achieved, with AUC of 1-, 3- and 5-year survival rates of 0.733, 0.722, and 0.737, respectively (Fig. 1I).
3.3 Exploration of the relationship between risk signature and clinical feature
A heat map was drawn to study the relationship between the risk signature and glioma patient feature, and the results showed the relationship between risk groups and the clinical parameters of the patients such as gender, age, pathological grade, IDH mutation status and 1p/19q deletion (Fig. 2A). There seemed to be no prominent difference in gender between the high- and low-risk groups, but there were more elderly patients in the high-risk group. Patients ≤ 60 years old had lower risk scores than those > 60 years old (p < 0.001). When comparing the WHO classification in the high- and low-risk groups, the high WHO grade was more likely to move towards the direction of the high-risk group, while the low-risk group was more likely to have more low WHO grade patients (p < 0.001). As for IDH mutation status, the risk scores of IDH wild type patients were higher than the risk scores of IDH mutation patients; that is, the high-risk group was mostly IDH wild type (p < 0.001). Then, we analyzed the status of the 1p/19q deletion, and our results showed that those in the high-risk group were more possibly to be the 1p/19q non-codeletion type group(p < 0.001) (Fig. 2B-F). The clinical feature of glioma patients in CGGA database were displayed in Supplementary Figure S4.
3.4 Establishment of a nomogram for predicting the 1-, 3- and 5-year survival rates of patients with glioma
We conducted univariate and multivariate Cox analyses, and our results indicated that this risk signature can be recognized as an independent prognosis factor for glioma patients (Fig. 3A and 3B). For better glioma patient prediction depending on the analysis results above, we utilized the TCGA database to construct a nomogram of risk score, age, grade and IDH mutation and validated it with the CGGA database (Fig. 3E and Supplementary Figure S5). The calibration plots showed an ideal match between the actual and nomogram-predicted probability of 1-, 3- and 5-year OS in the TCGA and the CGGA cohort (Fig. 3C). Furthermore, a time-dependent ROC curve was performed to evaluate the predictive capability of the nomogram and predictors (risk score, age, grade and IDH status (Fig. 3D). The results showed that the nomogram could well predict 1-year, 3-year and 5-year OS with AUC values of 0.628, 0.580, and 0.565 in TCGA database and of 0.603, 0.698, and 0.738 in CGGA database. The nomogram is more accurate in predicting the 1, 3 and 5-year survival rates of patients with glioma. These results demonstrate that the nomogram is more credible and precise prognostic predictor.
3.5 Relevance between the risk signature and immune landscape of the glioma microenvironment
Through functional enrichment analyses, we obtained the potential biological processes and relevant signaling pathways in which the apoptosis related genes risk signature was involved in glioma. As shown in Fig. 4A-D, the biological processes neuroactive ligand-receptor interaction, cell adhesion molecules (CAMs), ECM-receptor interaction and many other processes were markedly relevant to the signature. Regarding the KEGG pathway, immunity-related pathways (human T- cell leukemia virus 1 infection, complement and coagulation cascades, peptide ligand-binding receptors, regulation of IGF transport and uptake by IGFBPS, neurotransmitter receptors and postsynaptic signal transmission, GPCR ligand binding) and apoptosis-related pathways (P53 signaling pathway, cell cycle, cellular senescence, degradation of the extracellular matix) were significantly associated with the signature. The immune infiltrating cells of the high- and low-risk groups was indicated in Fig. 4E. We observed that Tregs, activated NK cells, monocytes, M0 macrophages, M1 macrophages, activated CD4 memory T cells, mast cells, follicular helper T cells and neutrophils were all markedly different between the two risk groups (p < 0.05). However, CD8 + T cells were not significantly different in the two groups. Due to the dual importance of CD8 + T cell number and function, we further calculated the relevance between risk scores and immune checkpoint expression. In Fig. 5E, we found that the number of immune checkpoints such as CD274, CD276, CTLA4, CD70, CD86, LILRB2 and TIMGD2, was positively relevant to the risk score, which fully demonstrated an immunosuppressive microenvironment in the high-risk group of patients with glioma.
3.6 Risk signature with tumor mutation burden and anti-PD-1/L1 immunotherapy
We analyzed the distribution of tumor mutation burden between low- and high-risk groups in TCGA cohort using “maftools” package. As indicated in Fig. 5A and 5B, low-risk group displayed more extensive tumor mutation burden than the high-risk group, with the rate of the 20th most noticeable mutated gene 99.37% versus 89.71%. More precisely, IDH1 and ATRX mutation happen more frequently in low-risk group than high one (92% vs 50%, 37% vs 31%), which predicts a better prognosis and is consistent with the results above. While several oncogenic mutations in high-risk group such as EGFR and PTEN happen a few times more than in low-risk group (14% vs 2%, 10% vs 2%). These all proved that high-risk group are more aggressive. Further, immunotherapies of PD-1 and PD-L1 blockade has indubitably arose a momentous breakthrough in tumor therapy. We inquired whether the apoptosis-related gene signature could predict patients’ response to immune checkpoint blockade therapy. The result from TIDE demonstrated that 58% patients have a response for anti-PD-1/L1 immunotherapy in low-risk group, while 69% patients have a response for anti-PD-1/L1 immunotherapy in high-risk group (Fig. 5D). The result from ImmuCellAI demonstrated that 39% patients have a response for anti-PD-1/L1 immunotherapy in low-risk group, while 54% patients have a response for anti-PD-1/L1 immunotherapy in high-risk group (Fig. 5C).
3.7 Expression level of the four apoptosis-related genes
We validated the level of the 4 apoptosis-related genes at mRNA and protein levels in the tissue sections of 28 gliomas and 15 paracancer patients. Quantitative PCR and immunohistochemistry showed that CREM, TNFSF12 and PRKCD were highly expressed in gliomas, while PEA15 was low expressed in gliomas (Fig. 6A-E).
3.8 Verified the relationship between these four genes and apoptosis by flow cytometry analysis
We constructed CREM, TNFSF12, PRKCD and PEA15 knockdown U251 glioma cell lines. As indicated in Fig. 6F, compared to the control group, the cell vitality of CREM, TNFSF12, PRKCD knockdown group decreased significantly, while that of PEA15 knockdown group increased significantly. Flow cytometry analysis demonstrated an increase in apoptotic cells in CREM, TNFSF12, PRKCD knockdown cells, and a decrease in apoptotic cells in PEA15 knockdown cells compared with the control group. We can conclude that CREM, TNFSF12 and PRKCD knockdown can promote glioma cells apoptosis, and PEA15 knockdown can restrain apoptosis of glioma cells (Fig. 6G-J).