Differential Gene Expression in CD4+ T Cells
Investigating the tumor immune cell infiltration microenvironment in hypopharyngeal cancer, we utilized the CIBERSORT algorithm to estimate the relative proportions of 22 immune cell types. The dominant frequencies of memory B cells, macrophages M0 and M1 types, activated mast cells, monocytes, resting natural killer cells, plasma cells, T cells CD4 memory resting cells, memory-activated T cells, CD4+ T cells, follicular helper T cells, and regulatory T cells (Tregs) are shown in Figure 1A. These proportions of immune cell subtypes served as feature data for Weighted Gene Co-expression Network Analysis (Figure 1D). Simultaneously, WGCNA was performed on the top 50% of genes based on standard deviation (n=9116). Setting the soft threshold power to 5 through the pickSoftThreshold function ensured that the gene network adhered to a scale-free distribution, with a scale-free topology model fit index of 0.9 achieved((Figure 1C). A hierarchical clustering tree was then generated using dynamic hybrid clustering. The resulting tree diagram depicted genes as individual leaves, with genes sharing similar expression data grouped together into branches to form gene modules. Modules with high similarity were merged at a cutoff of 0.25, resulting in the creation of 26 modules (Figure 1E,WGCNA Workflow Diagram depicted in Figure S2). The blue module comprised 724 genes, showing a strong correlation with CD4+ T cells (R2 = 0.75, P = 3e-04), while the blue-green module consisted of 278 genes, significantly linked to regulatory T cells (Tregs) (R2 = -0.81, P = 5e-05). To identify genes specifically associated with CD4+ T cells, sequencing count data from 12 hypopharyngeal cancer patients and 6 healthy controls (totaling 29,313 genes) were analyzed using the DESeq2 package for differential expression analysis. A total of 2,090 differentially expressed genes (DEGs) were identified, comprising 940 upregulated and 1,150 downregulated genes. The volcano plot (Figure 1B) illustrates the top 20 DEGs. After intersecting with the genes in the blue module, 195 differentially expressed genes associated with CD4+ T cells were identified. KEGG enrichment analysis of the differentially expressed genes mainly enriched in extracellular matrix structural constituents, integrin binding, protein polysaccharide binding, and Wnt receptor activity. Detailed results are provided in Figure S3 and Table S5.
Single-cell overview of different cell types in hypopharyngeal carcinoma
We performed single-cell gene expression analysis on hypopharyngeal cancer cells from five individuals in the GSE227156 dataset. After filtering out genes expressing red blood cells (<3%) and granulocytes (<10%), the datasets were merged and normalized. PCA dimensionality reduction was applied to 3000 highly variable genes, followed by batch correction using harmony to mitigate batch effects. During the data processing, no significant batch effects were observed(Figure S 4).
The resulting cell-gene matrix revealed an average of 1692 genes detected per cell. Utilizing umap/tsne clustering, we identified seven cell types: macrophages (Cluster 0), squamous epithelial carcinoma cells (Cluster 1), lymphocytes (Clusters 2 and 4), fibroblasts (Cluster 3), endothelial cells (Cluster 5), dendritic cells (Cluster 6), and epithelial cells (Cluster 7). Clusters were annotated manually using specific markers from the Cell Marker database and Durante et al.'s studies (Figure S 5). Further analysis uncovered 75 differentially expressed genes associated with tumor-related CD4+ T cells within Cluster 2.
PPI networks and Enrichment analysis of hub genes
To explore the protein-protein interactions of differentially expressed genes associated with CD4+ T cells, we inputted 75 genes into the STRING database to analyze their interactions. Disconnected nodes were removed during network construction, using a default interaction score of 0.4. Subsequently, the interaction data was imported into Cytoscape, resulting in the generation of Figure 3A. Utilizing the CytoHubba plugin, we identified central hub genes within the PPI network, including EGFR, Dlx5, DSG2, TP63, DLX2, and TSLP. Further analysis via ssGSEA revealed that high EGFR expression levels were significantly associated with tumor protein polysaccharides (Figure 3B). Conversely, high DLX5 expression levels were predominantly linked to signaling pathways regulating stem cell pluripotency (Figure 3C).
Identification of prognostic markers for hypopharyngeal cancer
Based on clinical data collected from Chaoyang Central Hospital, Kaplan-Meier analysis was conducted on EGFR and Dlx5 genes (Figures 4A-B), revealing an association between elevated Dlx5 gene expression and poorer prognosis. To validate these findings, hypopharyngeal cancer microarray datasets (GSE2379) from the Gene Expression Omnibus (GEO) database were obtained, consisting of the GPL-91 and GPL-8300 platforms. Survival analysis results are detailed in Figures 4C-D-E-F. To address potential biases due to sample size limitations, the investigation was expanded to include a larger set of head and neck tumor samples. RNA sequencing data comprising 546 cases of head and neck squamous cell carcinoma from the UCSC database were accessed for validation, yielding a hazard ratio of 1.42 (95% confidence interval: 0.82-0.904) for EGFR and 0.73 (95% confidence interval: 0.6872-0.769) for Dlx5, as depicted in Figures 4G-H. Additionally, immunohistochemical (IHC) staining results from the Human Protein Atlas database were retrieved to further confirm the expression levels of EGFR and Dlx5 genes (Figures 4I-J). This combined version effectively integrates the key findings and methodological steps in a coherent manner.
Correlation Analysis of Immune Checkpoint
Immune checkpoints[22] are a class of immune-inhibitory molecules expressed on the surface of immune cells. They regulate the activation level of the immune system to prevent excessive activation, which could lead to autoimmune reactions. However, cancer cells exploit these immune checkpoints, particularly T cell negative regulatory mechanisms, to dampen the immune system's attack, enabling immune evasion. To counter this, inhibitors such as PD-1, PD-L1, and CTLA-4 have been developed to alleviate immune response constraints, thereby reactivating T cells to target tumor cells and enhance cancer treatment efficacy. Despite these advancements, the majority of patients do not experience significant benefits, with response rates typically ranging from 10% to 25% [23], even in approved therapeutic indications. Therefore, the future of immunotherapy for head and neck squamous cell carcinoma may lean towards combination therapies or strategies that enhance immune response rates by combining with other targeted drugs to overcome resistance to immune checkpoint blockade. In light of this, we assessed the correlation between the DLX5 gene and immune checkpoints (Figure 5, GSE227156 platform GPL-91), revealing significant associations between Dlx5 and the immune checkpoint BTN3A1 (R=0.71), CCD28 with BTN2A1 (R=0.72), and HLA-F with HLA-G (R=0.72). Thus, interventions targeting DLX5 offer a promising avenue to augment the effectiveness of immune-based therapies.
Tumor Mutational Burden
Tumor mutational burden (TMB) [24] indicates the quantity of mutations in a tumor. Mutated proteins form neoantigens, which are presented to T cells by antigen-presenting cells through major histocompatibility complex (MHC) proteins. This process allows T cells to identify and release perforins and granzymes to attack and eliminate mutated tumor cells. Increased mutations, leading to a higher TMB, enhance the chances of immune recognition and targeting of tumor cells, improving the effectiveness of immunotherapy. Studies by Chabanon et al. (2016) [24] and Rooney et al. (2015) [25]have demonstrated a positive link between high tumor mutational burden (TMB-H) and positive outcomes post-treatment with immune checkpoint inhibitors (ICI). Clinical data also reveals a significant association between TMB levels and responses to PD-1/PD-L1 inhibitors[26]. The GSCA website (GSCA, http://bioinfo.life.hust.edu.cn/GSCA) [27] integrates data on gene expression, mutations, drug sensitivity, and clinical information from four public sources across 33 cancer types. By using the genomic alteration module to visualize the mutation burden of core genes, Figure 6 illustrates mutation percentages for EGFR, FAM83B, Dlx5, DSG2, IL1RAP, and CXADR, which are 41%, 21%, 18%, 12%, 12%, and 6%, respectively. This highlights the relatively high immunogenicity of Dlx5.
Correlation analysis of immune chemokines and receptors.
The tumor microenvironment plays a pivotal role in mediating interactions between tumor cells and the immune system. Various immune cell populations are attracted to this environment by specific chemokine factors, influencing tumor progression and treatment responses significantly[28]. Thus, therapeutic strategies targeting both pro-tumor and anti-tumor chemokine-receptor signaling pathways, in conjunction with immunotherapy, offer promising clinical benefits for cancer patients. To bolster this hypothesis, we conducted a comprehensive analysis correlating key genes with chemokines and their receptors.
We compared samples across three datasets: (i) 46 immune chemokines from different chemokine subfamilies, (ii) 26 corresponding immune chemokine receptors, and (iii) gene expression profiles of key genes. After logarithmically transforming gene expression data, we calculated distances between immune chemokines, receptors, and key genes using the Euclidean distance method. Subsequently, we utilized the linkET package in the R software to perform bias-corrected Mantel[29]correlation analysis.
Our analysis revealed significant correlations between EGFR and immune chemokines and receptors, as well as a robust correlation between DSG2 and these immune factors. Notably, while DLX5 exhibited a significant correlation with chemokines, its relationship with receptors did not reach statistical significance. This suggests that DLX5 may regulate chemokine activity, while its interactions with receptors could be influenced by unexplored factors.
Evaluating the Therapeutic Response
To predict the response of hypopharyngeal cancer to chemotherapy, we utilized the oncoPredict R package to estimate chemotherapy response based on half-maximal inhibitory concentration (IC50) data from the Cancer Cell Line Encyclopedia (CCLE) database, available for hypopharyngeal cancer patients. In our study, patients were stratified into high and low expression groups based on DLX5 expression levels, leading to the identification of 25 small molecule compounds with significantly different responses (Table S5). Figure 8 illustrates the top three small molecule compounds with the most statistically significant differences: BI.2536_1086 (P = 0.00019, Figure 8A), MN.64_1854 (P = 0.00103, Figure 8C), and Ulixertinib_2047 (P = 0.0013, Figure 8E).
Overexpression Dlx5 promotes cell proliferation and invasion
We conducted a detailed investigation into the role of Dlx5 in hypopharyngeal squamous cell carcinoma by introducing pCDNA3.1-DLX5 and empty pCDNA3.1 vectors into the FaDu cell line. RT-qPCR confirmed the successful overexpression of DLX5, and we evaluated the impact of Dlx5 on hypopharyngeal cancer cell proliferation through CCK-8 assays. Our findings revealed that upon DLX5 overexpression, cell proliferation was boosted, with an increase of over 42% in proliferation activity noted after 48 hours of overexpression (Figure 9A). Additionally, the outcomes from transwell migration and scratch healing experiments indicated a notable rise in cell migration speed and improved cell healing capability in FaDu cells (Figure 9C-D). Moreover, in the transwell invasion assay, the overexpression of Dlx5 substantially heightened the invasive potential of the tumor cells (Figure 9E). To summarize, the upregulation of Dlx5 stimulates the proliferation, migration, and invasion of hypopharyngeal cancer cells.
Knockdown of Dlx5 inhibits cell viability and cell proliferation
In order to confirm the potential involvement of Dlx5 in hypopharyngeal cancer, we developed three siRNAs (siRNA-1, siRNA-2, siRNA-3) to silence Dlx5 expression in FaDu cells. PCR validation demonstrated the effective suppression of Dlx5 expression by siRNA-1, achieving an mRNA knockdown efficiency of 80% (Table S7). To assess the impact of Dlx5 on cell proliferation, invasion, and migration, we performed CCK-8, Transwell, and scratch healing assays on FaDu cells transfected with siRNA-1 and a negative control (NC). The CCK-8 results revealed that silencing DLX5 decreased cell proliferation capacity, showing a maximum reduction of around 30% (Figure 9A). Transwell experiments indicated that downregulating Dlx5 constrained the migration and invasion abilities of FaDu cells (Figure 9D-E). The scratch healing assay further corroborated the diminished migration capability of FaDu cells following reduced Dlx5 expression (Figure 8B). These results suggest that suppressing Dlx5 expression can hinder the proliferation, migration, and invasion of hypopharyngeal cancer cells.