The expression of HDAC1 is significantly elevated in the MES subtype GBM and shows a positive correlation with the expression of MES-specific genes.
To assess the variation in HDAC1 expression across different GBM subtypes, TCGA GBM data were analyzed, revealing significantly elevated mRNA expression in the MES subtype and reduced expression in the PN subtype (Fig. 1A). Previously validated MES subtype GBM cell line TBD0220L/N9 and PN subtype GBM cell line TBD0207B/U251 [13] were used in this study. HDAC1 mRNA expression was notably higher in TBD0220L than in TBD0207B (Fig. 1B) and in N9 compared to U251 (Fig. 1C), with similar results for HDAC1 protein expression (Fig. 1D). Characteristic genes for each subtype were evaluated using Verhaak RG classification data (Table S5). Cerebrospinal fluid specimens from four patients with GBM (Table S1) were analyzed to evaluate the correlation between HDAC1 mRNA and subtype marker gene. HDAC1 expression positively correlated with the MES subtype marker gene MMP7 and negatively with the PN subtype marker gene HOXD3 (Fig. 1E). Immunofluorescence detection confirmed a positive correlation between HDAC1 and MMP7 expression (Fig. 1F). To inhibit HDAC1, siHDAC1 was designed to prevent protein synthesis, and an HDAC1 inhibitor (RG2833) was used to block its biological function (Supplementary Fig. 1). Both methods decreased MES subtype marker gene expression and increased PN subtype marker gene expression (Fig. 1G-H). TCGA data analysis further revealed a positive correlation between HDAC1 and MES subtype marker genes and a negative correlation with PN subtype marker genes (Fig. S2).
The in vivo experiments conducted on PDX models demonstrated that RG2833 is effective in treating MES subtype GBM, inducing the transformation of the MES subtype to the PN subtype.
First, after intracranial transplantation of heterologous MES GBM tissue in nude mice, RG2833 was administered by gavage, demonstrating its inhibitory effect on MES GBM proliferation and its ability to extend the survival time of nude mice (Fig. 2A-C). Immunohistochemical experiments were conducted using paraffin sections of intracranial tumor tissue, and mRNA from the tumor tissue was extracted for q-PCR analysis. The HDAC1 inhibitor reduced the expression of MES subtype marker genes and increased the expression of PN subtype marker genes in vivo (Fig. 2D-E).
The combination of RG2833 and bevacizumab inhibited the proliferation and invasion of MES GBM cells.
Analysis of survival data from 52 patients with GBM using the TCGA database revealed that MES subtype patients had a poorer prognosis and shorter median survival compared to those with PN and CL subtypes (Fig. S3A-B). This finding prompted an exploration of therapeutic strategies aimed at transforming the poorly prognostic MES subtype into other subtypes. The PN subtype, for example, shows an effective therapeutic response to bevacizumab [15]. Our experiments demonstrated that bevacizumab inhibited the proliferation and invasion of the PN subtype cell line U251 in vitro (Fig. 3A-C). Although bevacizumab had no significant effect on MES cell lines, RG2833 significantly inhibited their proliferation and invasion. Additionally, combining RG2833 with bevacizumab had a more pronounced inhibitory effect on the proliferation and invasion of the N9 cell line (Fig. 3D-F). Western blotting analysis was conducted to investigate the protein expression of MES marker genes following bevacizumab treatment in N9 cells, revealing no significant changes. However, the combination of bevacizumab and RG2833 significantly reduced the protein expression of MES marker genes. These changes were mirrored at the mRNA level, showing a consistent trend with the protein expression (Fig. 3G-H).
Perform differential analysis of RNA-seq data for MES cells before and after treatment with RG2833.
To investigate the molecular mechanism by which RG2833 induces subtype conversion, the N9 cell line was divided into a control group and an RG2833 treatment group, each with two replicates. Differential gene analysis via clustering identified the 40 genes with the most significant differences, all with p-values less than 0.0001 (Fig. 4A). The intersection of identified genes (p < 0.05) between replicates revealed gene overlap rates of 83% and 87% in the control and RG2833 treatment groups, respectively (Fig. 4B). Volcano plot analysis of overlap genes indicated that RG2833 treatment significantly upregulated 833 genes, downregulated 575 genes, and left 15,340 genes unchanged (p-adjust value of 0.05 and log fold change (log FC) of 1) (Fig. 4C). GO analysis linked the differentially expressed genes to cell growth, death, and cancer-related pathways (Fig. 4D).
HDAC1 interacts with p-SMAD3, and RG2833 induces alterations in the subcellular localization of HDAC1.
Immunofluorescence experiments using specific antibodies revealed significant co-localization of HDAC1 and p-SMAD3 in TBD0220L cells, indicating a strong correlation between these proteins (Fig. 5A). In the N9 cell line, Co-IP assays demonstrated a reciprocal recruitment interaction between HDAC1 and p-SMAD3 (Fig. 5B). Additionally, we used antibodies against H2AK5ac, H2BK5ac, H3K9ac, and H3K27ac to assess changes in histone acetylation following RG2833 treatment. The results showed increased acetylation levels within the nucleus (Fig. 5C). Furthermore, RG2833 treatment altered HDAC1 protein distribution, decreasing its presence in the nucleus while increasing its presence in the cytoplasm (Fig. 5D). Intracellular immunofluorescence staining of TBD0220L cells showed consistent alterations, confirming the observed trends (Fig. 5E).
By inhibiting HDAC1 to block histone acetylation leads to the recruitment of p-SMAD3 to the genome, thereby influencing transcription in MES GBM cells.
The N9 cell line was used for ChIP-seq experiments, enriching proteins with the p-SMAD3 ChIP-grade antibody, precipitating genomic DNA, and conducting high-throughput sequencing analysis [16]. The p-SMAD3 binding sequence was identified (https://jaspar.elixir.no/) (Fig. S4). Cell samples were divided into a DMSO control group and an RG2833 treatment group, each with two replicates. Post-treatment comparisons revealed a significant increase in p-SMAD3 enrichment around the transcription start site (TSS) following RG2833 treatment (Fig. 6A). Analysis of p-SMAD3 distribution in different coding DNA regions showed an increased distribution ratio of p-SMAD3 in the promoter region, particularly the regions within 1kb base pairs of the actual TSS after RG2833 treatment (Fig. 6B). Comparing sequencing coding gene data from the control and treatment groups, we found 89% gene overlap in the control repeated group and 93% in the treatment repeated group (Fig. 6C). Combining differential genes between the control and treatment groups with genes showing significant binding changes, we performed clustering analysis to identify genes with p-adjust values < 0.0001. The results indicated that 18 genes showed a significant decrease in binding, while 31 genes showed a significant increase in binding (Fig. 6D). In summarizing the RNA-seq results before and after RG2833 treatment (Fig. 4C), we took the intersection to identify genes with increased mRNA expression (p-adjust value < 0.05) and increased p-SMAD3 binding with ChIP-seq analysis (p-adjust value < 0.05), and obtained 107 potential downstream target genes (Fig. 6E). Targeted gene GO analysis revealed associations with axon guidance, ECM-receptor interaction, and cell adhesion molecules (Fig. S5).
TP53I11 maintains the characteristics of PN subtype GBM and inhibits the proliferation and invasion of MES GBM both in vivo and in vitro.
TP53I11 emerged as a significant downstream regulated gene cluster from our previous analyses (Fig. 4A and 6D). ChIP-qPCR experiments revealed that the p-SMAD3 antibody effectively enriched TP53I11, with enrichment levels increasing following RG2833 treatment (Fig. 7A, Table S6). Analysis of TCGA GBM patient data indicated that patients with higher TP53I11 mRNA expression in tumor tissue had significantly longer survival times compared to those with lower TP53I11 mRNA expression (Fig. 7B).
By utilizing the subtype-specific TCGA GBM data, we observed a significant elevation of TP53I11 expression in the PN subtype compared to the MES subtype at the mRNA level (Fig. 7C). Gene set enrichment analysis (GSEA) based on the Verhaak GBM PN gene set showed that patients with GBM exhibiting high TP53I11 expression had higher enrichment scores, while those with low TP53I11 expression had lower enrichment scores (Fig. 7D). Receiver operating characteristic (ROC) curve analysis of TCGA GBM data indicated that TP53I11 has high sensitivity in distinguishing PN patients from other subtypes, with an area under the curve (AUC) of 84% (Fig. 7E). TP53I11-specific interfering RNA (siRNA) was constructed and applied to the PN subtype GBM cell line U251, resulting in increased cell proliferation and invasion as shown by CCK-8 and Transwell assays (Fig. 7F-H). Additionally, a shTP53I11 lentiviral vector was developed, transfected into U251 cells, and then injected into the brains of mice to form tumors. Compared to the control group, the shTP53I11-treated group displayed larger tumor volumes and weights (Fig. 7I-L). Enhanced Ki-67 positivity, detected by immunohistochemistry, indicated increased proliferative capacity in shTP53I11-treated tumors (Fig. 7M and N).