Identify the pericytes concerning BTBB functions in GBM.
Nine GBM fresh tumor samples were selected for scRNA-seq (Table 1), and 9,3343 cells passed quality control and were included in this study. Graph-based clustering analysis, using known lineage markers, identified seven cell populations, with their characteristics shown on the heatmap (Fig. 1b-c). Highly heterogeneous population of cancer cells were observed, due to the GBM heterogeneity among samples. Pericytes are part of the stromal cells by definition. Stromal cells make comprise 2.38% of the total cell count in the 9 specimens, including 2217 cells, and further clustering divided the stromal cells into 12 sub clusters with their hallmarks shown in the bubble diagram (Fig. 1d-e).
Table 1
Clinical information of specimens selected to perform scRNA-seq.
Sample | Age | Gender | GFAP | Ki67 | P53 | IDH1 | Olig2 | EGFR | MGMT |
G3 | 66 | M | + | 15%+ | wt | - | + | + | |
G4 | 64 | M | + | 30%+ | wt | - | + | - | + |
G12 | 58 | M | + | 10%+ | + | - | + | + | - |
G14 | 65 | F | + | 30%+ | wt | - | + | - | |
G15 | 69 | F | + | 25%+ | wt | - | + | 2+ | |
G17 | 73 | M | + | 10%+ | + | - | + | 1+ | |
G18 | 72 | M | + | 70%+ | + | - | + | 1+ | |
G19 | 56 | F | + | 40%+ | wt | - | + | - | + |
G21 | 46 | M | + | 30%+ | wt | - | - | + | + |
G22 | 67 | M | + | 10%+ | + | - | + | + | - |
The results indicate that pericyte markers, from previous studies, display discrete distributions within sub clusters (Fig. 2a), suggesting significant heterogeneity among various pericyte research. BMX, which has been identified as a therapeutic target in GBM pericytes[44], exhibits low expression in all clusters. Due to the crossed and discrete marker expression, pericyte characteristics are not distinct. Garcia FJ et al[41] investigated human brain vasculature using single-nucleus RNA-sequencing (snRNA-seq) data, and we collected gene expression signatures from five clusters annotated as pericytes (Pericyte 1, Pericyte 2, Post Mortem Pericyte 1, Post Mortem Pericyte 2, Post Mortem Pericyte 3). Pericyte 1 and Pericyte 2 were annotated from ex vivo freshly resected surgical human brain tissue, while Post Mortem Pericyte 1, Post Mortem Pericyte 2, and Post Mortem Pericyte 3 were annotated from post mortem frozen human brain tissue. We used the top 50 DEGs of five pericytes clusters for the GSVA in our study. Cluster 1, 6, and 11 exhibit distinct pericyte characteristics, with cluster 6 demonstrating significant enrichment in all five pericyte expression signatures (Fig. 2b). Thus, cluster 1, 6, and 11 are potential pericyte clusters. To further confirm our findings, we conducted GSEA in cluster 6, resulting in significant enrichment outcomes in five different pericyte expression signatures (Fig. 2c).
To further investigate the function of different pericytes in GBM, we conducted Gene Ontology (GO) functional enrichment analysis on the top genes of each stromal cell cluster. In GO term analysis, we identified different biological functions of pericytes such as contraction, cell adhesion, immune functions and vascular processes that were enriched in distinct clusters (Fig. s1). Cluster 6 is characterised by BP terms that relate to vascular function and permeability, such as "vascular process in circulatory system", "transport across the blood-brain barrier", and "vascular transport". The CC terms in this cluster pertain to basal plasma membrane and cell adhesion, for example, "basal plasma membrane", "basolateral plasma membrane", "collagen-containing extracellular matrix", "cell-substrate junction", and "focal adhesion". The MF terms in cluster 6 are associated with transmembrane transporter activities (Fig. 3a). The GO functional enrichment analysis demonstrated significantly enriched functions relating to the structure and permeability of the BBB in cluster 6. This indicates a close correlation between cluster 6 and BTBB functions in GBM.
Additionally, cluster 1 is enriched in pathways relating to cellular adhesion, biosynthesis, and regulation of cellular morphology. It is worth noting that cluster 8 exhibits pathway enrichment with strong characteristics of smooth muscle cells (SMCs) (Fig s1), and cluster 8 highly expresses most pericyte markers (DES, αSMA, CALD1, Ang1), and GSEA findings for pericyte expression signatures are insignificant (Fig s2). The conflicting findings from GSVA, GO functional enrichment analysis, and biomarker expression emphasize the lack of specificity in pericyte markers within literary reviews.
Pertinent hallmark for pericyte of BTBB functions.
The results above demonstrate stromal cell sub cluster 6 to be pericyte of BTBB functions, to determine potential hallmarks for them, the top 50 up-regulated DEGs of cluster 6 and normal brain pericytes and GO terms genes related to capillary permeability were extracted. After intersection, only three genes remained: ATP1A2, SLC6A1, and PTH1R (Fig. 3b). These three genes were found to be expressed specifically in cluster 6 of stromal cells (Fig. 3c).
To verify those potential hallmarks, IF co-staining was conducted using histological sections obtained from nine scRNA-seq GBM samples. CD31 was selected as a hallmark for identifying endothelial cells in co-staining with potential hallmarks. The results indicate that cells marked by PTH1R, ATP1A2, and SLC6A1 exhibit morphological cohesion with the surrounding blood vessels of GBM tissue (Fig. 3d). Specifically, cells marked by PTH1R displayed significant specificity in the IF co-staining among the three markers and conform to the classic morphological definition of pericytes, which are cells located anatomically surrounding the walls of blood vessels. Therefore, the findings indicate that PTH1R can be regarded as a novel and highly specific hallmark of pericytes with respect to BTBB functions in GBM.
Multiple IF of PTH1R, CD31, and recognized hallmark of pericytes (ANPEP and Desmin) were conducted in GBM tissue to verify the specificity of PTH1R as a hallmark of pericytes related to BTBB functions in the brain. The results demonstrate that PTH1R has analogous specificity to ANPEP when labeling pericytes. PTH1R and ANPEP both have significantly higher specificity than Desmin (Fig. 3e). There is a significant negative correlation between higher expression of PTH1R and prognosis in the CGGA GBM cohort (Fig. 3f), and the expression of PTH1R is significantly lower in high grade glioma than in low grade glioma, as depicted in Fig. 3g.
PTH1R + Pericytes communicate with other cells by ECM-integrin signaling pathways and secreting heparin-binding growth factor.
To investigate the cell–cell interaction network among the cell types identified in our present work, we used the CellPhoneDB analysis and pericytes showed active interactions with other cell types, and they showed especially strong interactions with other stromal cells (Fig. 4a). The results identified crucial pathways within ECM-integrin signaling, including FN1, COL1A2, COL4A1 and other collagen-related genes enriched in PTH1R+ pericytes interacting with multiple integrin complexes in other cell populations, such as aVb1 complex, a4b1 complex, aVb5 complex, a5b1 complex, a1b1 complex, a10b1 complex, and others. Of significant interest to us are the pathways enriched between PTH1R+ pericytes and tumor cells/ECs. The FN1-aVb1 and FN1-a5b1 pair exhibit a high level of enrichment in the interaction between PTH1R+ pericytes and ECs, with FN1-aVb1 also showing significant enrichment between PTH1R+ pericytes and tumor cells. Collagen-integrin signalling pathways are primarily activated between PTH1R+ pericytes and other stromal cells. In addition, CD74 enriched in ECs interacts with APP in PTH1R+ pericyte as indicated (Fig. 4b).
Additionally, PTN was enriched in PTH1R + Pericytes via paracrine interactions with PTPRZ1 in various cell types, including tumor cells, oligodendrocytes, other stromal cells and multiple immune cells in GBM (Fig. s3).
To confirm the findings of the CellPhoneDB analysis, we examined the relationships of the interaction pairs above in the CGGA GBM cohort. We observed that all pairs exhibited significant positive correlations, particularly the ECM-integrins interaction pairs (Fig. 4c).
Difference between PTH1R + Pericytes in GBM and normal brain tissue.
Clustering was conducted and visualised on scRNA-seq data from 4 GBM samples containing the tumor core and paired surrounding peripheral tissues. Identification of cell types in different populations was conducted using marker genes provided by the original research (Fig. 5a). Stromal cells were subdivided into sub clusters (Fig. 5b). According to GSVA and GSEA, cluster 8 of the stromal cells exhibited expression signatures similar to pericytes in both previous research and our study (Fig. 5c-d). The pericyte hallmarks identified above - ATP1A2, SLC6A1, and PTH1R - exhibit a distinct expression in this cluster (Fig. 5e). Conversely, previously acknowledged pericyte hallmarks - RGS5, PDGFRβ, and NOTCH3 - demonstrate low specificity in the findings (Fig. 5f). Differential gene expression analyses were performed between pericytes in GBM and non-malignant tissue, followed by functional enrichment analysis. The findings indicate significant enrichment in pathways such as the complex of collagen trimers, granulocyte chemotaxis, basement membrane, endodermal cell differentiation, and collagen-containing extracellular matrix (Fig. 5g), with the proportion of pathways exhibited in a pie chart (Fig. 5h). The top 10 DEGs, including THY1, COL3A1, COL4A1, TIMP1, COL4A2, COL1A1, MGP, MIR4435-2HG, and FN1, are mostly ECM-related genes. These results demonstrate substantial up-regulation and interactions among ECM components in pericytes of GBM. COL4A1 and COL4A2 encode alpha proteins for collagen IV and are hub genes that combine pathways for collagen trimers and the basement membrane pathway. This indicates that additional collagen IV generated by pericytes is a structural characteristic of BTBB in GBM. The CellPhoneDB analysis has revealed interactions between ECM-related genes and the integrin family as a major pattern of PTH1R+ pericytes interacting with other cells in GBM tissue. The analysis of DEGs highlights active ECM-integrin family interactions led by PTH1R+ pericytes in GBM.
We then correlated the high and low expression groups of DEGs with the prognosis in the CGGA GBM cohort. All Top 10 DEGs, except THY1 (Fig. 6a), exhibited significant negative correlations. Collagen IV is an essential component of the blood-brain barrier. There is a substantial up-regulation of genes encoding collagen IV in GBM, which corresponds with the prognosis (HR = 0.67 Wilcoxon p value = 0.0151 in the COL4A2 group; HR = 0.61 Wilcoxon p value = 0.0021 in the COL4A1 group). These findings suggest that Collagen IV has potential value for prognosis prediction. FN1 is recognized as the hub gene in the ECM-integrin interaction network. The significant up-regulation of FN1 in GBM correlates with the prognosis (HR = 0.68 Wilcoxon p value = 0.0447). As such, Collagen IV and FN1 are selected as genes of interest for further validation.
30 samples of GBM and 20 LGG samples, all with complete follow-up data, underwent IHC staining to demonstrate collagen IV and FN1 expression levels. The expression levels of collagen IV and FN1 in GBM and LGG samples were presented using a violin plot, calculated with the median value of "IOD/Area" (collagen IV GBM: 0.0274 ± 0.01; LGG: 0.0202 ± 0.012 P = 0.026), (Fibronectin GBM: 0.018 ± 0.013; LGG: 0.0153 ± 0.008 P = 0.411) (Fig. 6b). The IHC results are indicated in Fig. 6c. Subsequent Kaplan-Meier survival analysis revealed a negative correlation between up-regulation of collagen IV and fibronectin and both progression-free survival (PFS) and overall survival (OS) in relation to GBM diagnosis. This correlation is illustrated in Fig. 6d, highlighting the potential of these markers as indicators of poor prognosis.
Correlation of TPRS and Immune characteristics in CGGA cohort
Pericytes and other stromal cells, in addition to diverse immune cells, have a crucial function in tumor immunity and potentially impact the choice of varied immunotherapies, such as mRNA vaccines[58]. To investigate the link between TPRS and immune characteristics in the cohorts, we analysed the correlation of TPRS with immune score, stromal score, and tumor purity in the CGGA cohort. As illustrated in Fig. 8a, we identified significant positive correlations between immune score and stromal score, whereas tumor purity was negatively correlated. Furthermore, Fig. 8b demonstrated a positive correlation with T cell infiltrated score. Enrichment of 28 immune cells was also assessed in the CGGA cohort using the ssGSEA method (Fig. 8c)[55]. Analysis revealed significant differences in immune cell components between TPRS high/low groups, with the TPRS high group scoring higher for activated CD4 T cells, central memory CD4 T cells, gamma delta T cells, memory B cells, and type 2 T helper cells, while the TPRS low group scored higher for activated B cells, eosinophils, immature B cells, monocytes, neutrophils, and type 17 T helper cells (Fig. 8d).
The cancer immunity cycle demonstrates various functions of immunomodulators objectively. In the TPRS high group, the release of cancer cell antigens (Step 1) and the infiltration of immune cells into the tumor (Step 5) were more activated. In contrast, dendritic cell recruiting (Step 4), NK cell recruiting (Step 4), and Th2 cell recruiting (Step 4) were more activated in the TPRS low group (Fig. 8e). Additional findings indicate that TPRS had negative correlations with most immune checkpoints, excluding CD276, HAVCR2, LAIR1, and LGALS3. However, TPRS was positively correlated with most pathways predicted for immunotherapy (Fig. 8f-g).