Abstract Background Evidence has been presented that the tumor protein D52 (TPD52) family plays a critical role in tumor development and progression. As a member of the TPD52 family, the changes in TPD52L2 gene status are instrumental in kinds of cancer development. However, its effects on patient prognosis and immune infiltration in Head and Neck Squamous Carcinoma (HNSCC) are still poorly understood. Methods The Tumor Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and c-BioPortal database was used to explore the expression pattern, prognostic value, and variation of gene status in HNSCC. The LinkedOmics database was used to obtain the co-expression genes of TPD52L2 and identify the diagnostic value of TPD52L2 in HNSCC. The correlations between TPD52L2 expression and six main types of immune cell infiltrations and immune signatures were explored using Tumor Immune Estimation Resource (TIMER). The correlation between TPD52L2 expression and immune checkpoint genes (ICGs) was analyzed by TCGA database. Immunohistochemistry (IHC) was performed to validate the expression of three ICGs (PDL1, PDL2, EGFR) and TPD52L2 using 5 paired HNSCC and normal head and neck tissues. Polymerase Chain Reaction (PCR) and Western Blot (WB) of HNSCC and normal head and neck cell lines were performed to verify the high level of TPD52L2 mRNA and protein expression. protein expression of TPD52L2 in pan-cancer was also validated using UALCAN. Results TPD52L2 was overexpressed in tumor tissues, and it predicted worse survival status in HNSCC. ROC analysis suggested that TPD52L2 had a diagnostic value. Multivariate Cox analysis identified TPD52L2 as an independent negative prognostic marker of overall survival. Functional network analysis suggested that TPD52L2 was associated with immune-related signaling pathways, cell migration pathways, and cancer-related pathways. High expression of TPD52L2 was associated with a more mutant frequency of TP53. Notably, we found that the expression of TPD52L2 was closely negatively correlated with the infiltration levels of 15 types of immune cells and positively correlated with several immune markers. PCR, WB experiments, and UALCAN database verified the high level of TPD52L2 mRNA and protein expression. Conclusion TPD52L2 is upregulated in HNSCC, which is an independent factor for adverse prognosis prediction. It probably plays a role in the negative regulation of immune cell infiltration. TPD52L2 might be a promising prognostic biomarker and therapeutic target in HNSCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12903-024-04977-1. Keywords: Head and neck squamous carcinoma, TPD52L2, Diagnosis, Prognosis, Immune cells, Bioinformatics Introduction As the sixth most common cancer worldwide, HNSCC affects approximately 900,000 patients annually. The incidence of this disease is rising, while therapeutic options have only marginally improved [[34]1, [35]2]. Approximately 95% of head and neck cancers are HNSCC, which develops in the mouth, hypopharynx, throat, or oropharynx [[36]3]. Current therapies of HNSCC mainly include surgery, radiotherapy, chemotherapy, and multidisciplinary comprehensive sequence therapy. As reported, sixty percent of patients are already in advanced stages (stage III or IV) at the time of diagnosis [[37]4, [38]5]. The survival rate among advanced HNSCC patients is only 34.9% and the effectively multidisciplinary treatment for HNSCC is still limited [[39]6, [40]7]. Thus, there is an urgent need for novel biomarkers and new therapeutic options for HNSCC. The tumor protein D52-like family is a group of coiled proteins. This family consists of TPD52, TPD52L1 (TPD53), TPD52L2 (TPD54), and TPD52L3 (TPD55). The TPD52 family has been reported to play an important role in the proliferation and metastasis of various cancer cells [[41]8, [42]9], TPD52 was shown to be overexpressed in breast cancer and prostate cancer and identified as a candidate oncogene [[43]10, [44]11]. In oral squamous cell carcinoma (OSCC), TPD52 was elevated under hypoxic conditions in a HIF-independent manner and promoted cell proliferation and survival [[45]12]. These data suggested that inhibition of TPD52 could contribute to cancer treatment. TPD52L2, a member of the TPD52-like protein family, is reported to be involved in a new class of intracellular transport vesicles: intracellular nanovesicles [[46]13]. And proteomic analysis revealed that TPD52L2 was one of the most abundant proteins in HeLa cells, ranking 180th out of 8,804 proteins, with an estimated 3:3 × 106 copies per cell [[47]12]. TPD52L2 has been proved to be highly expressed and linked to poor prognosis in several types of cancers, including lung adenocarcinoma [[48]14], pancreatic adenocarcinoma [[49]15, [50]16], and liver cancer [[51]17]. Recent studies indicate TPD52L2 to be a potential prognostic biomarker of survival for many cancers, but to date, no systematic reviews have been conducted on the role of TPD52L2 in HNSCC. In the current study, we performed a bioinformatic analysis by extracting and analysing TCGA, GEO, TIMER, LinkedOmics, c-BioPortal and UALCAN datasets to explore the potential oncogenic mechanism of TPD52L2 in HNSCC, as well as by using Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), pathway enrichment to investigate the potential functions of TPD52L2. Furthermore, we performed an IHC experiment to verify the correlation between TPD52L2 and tumor immune cell infiltration/immune checkpoint, whereas expression of TPD52L2 in HNSCC were validated by PCR and WB experiments. These findings offer fresh perspectives on TPD52L2’s role as well as brand-new targets for HNSCC diagnosis and prognosis. Materials and methods Data source and processing Pan-cancer TCGA dataset (PANCAN, N = 10,535, G = 60,499, N stands for the number of cases, G stands for genes) was downloaded from UCSC cancer genome browser ([52]https://xenabrowser.net/) The expression of TPD52L2 values were log[2](1 + x) transformed for downstream analysis. After excluding cancer species less than 3 samples, the final cancer species for the analysis was 26. The data of the TPD52L2 expression and the associated clinical outcome data in HNSCC patients were downloaded from TCGA ([53]https://portal.gdc.cancer.gov/), including 502 tumor samples and 44 normal samples, and then the data was converted to log[2] Transcripts Per Million (TPM). [54]GSE39400 [[55]18] dataset downloaded from GEO database ([56]https://www.ncbi.nlm.nih.gov/) was chosen as a validation set. T-test was used to compare the distribution in the expression levels of TPD52L2 between tumor and normal samples. The Copy Number Variation (CNV) datasets of HNSCC were obtained from TCGA and analyzed using the R package “maftools” [[57]19]. In addition, unpaired Wilcoxon’s ranksum and signed rank tests in R were performed to determine the significance of the difference between sample groups. Analysis of the clinicopathological features and diagnostic value Genomic data and clinicopathological features from a total of 502 tumor samples of HNSCC published were downloaded from TCGA for inclusion in this study. The patients were divided into two groups according to high and low TPD52L2 expression level, separated by the median value. To evaluate the relationship between the expression of TPD52L2 and clinicopathological features in HNSCC samples, Pearson’s chi-square test and Fisher’s Exact Test were performed. The diagnostic performance of TPD52L2 to differentiate between normal tissue and tumor was assessed by receiver operating characteristics (ROC) curve analysis. Prognostic analysis The dataset ([58]GSE65858 [[59]20], which was derived from the GEO database including 270 HNSCC tumor samples and corresponding gene expression and survival data) was used to validate the OS curves and Cox regression analysis in HNSCC. The samples in GEO database were stratified into two groups of high- and low-expression as described above. Then, the Kaplan–Meier analyses with the log-rank test was used for survival analysis in pan-cancer. Univariate and multivariate Cox regression analyses were conducted to explore the influence of clinical variables and TPD52L2 on the survival of patients with HNSCC using R packages “survminer” and “survival”. The prognostic variables p < 0.1 in the univariate Cox regression analysis entered the multivariate Cox regression model. Identification and enrichment analysis of co‑expression genes Co-expression analysis with Pearson correlation test was performed to identify co-expressed genes of TPD52L2 in the data of HNSCC from TCGA via the database LinkedOmics ([60]http://www.linkedomics.org/login.php). The top 120 co-expression genes were selected to perform GO and KEGG pathway enrichment analysis using the R package “clusterProfiler” (version 3.14.3) to explore the TPD52L2-related molecular mechanisms. Gene mutation and CNV status analysis TCGA-pan-cancer gene mutation and CNV data of TPD52L2 were retrieved from cBioPortal database ([61]https://www.cbioportal.org/). Gene expression data of TPD52L2 were extracted from the Pan-cancer TCGA dataset (PANCAN, N = 10,535, G = 60,499). GISTIC copy number data was downloaded from TCGA dataset. We calculated gene expression of TPD52L2 in tumor samples with different CNV status with the R statistical software (version 3.6.4), then compared differences between groups by unpaired Wilcoxon’s signed-rank test. The Kruskal–Wallis nonparametric statistical tests were performed using “kruskal.test” function from the stats package in R. Gene mutation frequency analysis We further graphed mutational landscape of TPD52L2 using ComplexHeatmap [[62]21] in R from TCGA database. The chi-square test was used to assess the difference in the frequency of mutations in each group of samples. Tumor immunity estimation resource database analysis TIMER ([63]https://cistrome.shinyapps.io/timer/) was utilized to analyze the comprehensive correlation between TPD52L2 and tumor-infiltrating immune cells signatures. The correlation between the CNV of TPD52L2 and the abundance of six types of tumors infiltrating immune cells (TIICs) including B cells, CD4 + T cells, CD8 + T cells, neutrophils, macrophages, and dendritic cells were explored. Gene expression and immune cell infiltration analysis Calculation of immune infiltration scores were based on the gene expression data previously identified. The level of infiltration of 24 immune cells in the tumors was evaluated based on gene expression using the R packages IOBR (version 0.99.9, [64]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8283787/). The relative enrichment score of these immune cells in HNSCC was assessed by single-sample GSEA, which was accomplished using the R package GSVA. The correlation between the expression of TPD52L2 and these immune cells was investigated using the Spearman’s correlation analysis, and the differences in the level of immune infiltration between the high and low TPD52L2 expression groups were evaluated using the Wilcoxon ranksum test. The Estimation of Stromal and Immune cells in Malignant Tumor tissues using Expression (ESTIMATE) algorithm was used to calculate and compare stromal scores, immune scores, and ESTIMATE scores between low and high TPD52L2 expression groups of HNSCC by Wilcoxon rank-sum test. Immune checkpoint gene analysis Patients with high expression of immune checkpoint inhibitors (ICIs) will benefit more from ICI treatment. We analyzed the correlation between the expression of TPD52L2 and more than 50 ICGs using the TCGA database. Five paired HNSCC and normal tissue specimens from Cancer Hospital of Shantou University Medical College were used (The immunohistochemistry involved patients who had undergone surgery and had not received chemoradiotherapy). The Ethical Committee of the Hospital approved the study protocol. IHC analysis used UltraSensitive SP IHC Kit-9710 (MXB, China), according to the manual instructions. The primary antibody used in IHC included: CD274 (PDL1) Monoclonal Antibody (14–5983-82, eBioscience invitrogen), Recombinant Anti-PDL2 antibody (ab205921, abcam), Recombinant Anti-EGFR antibody (ab52894, abcam) and Anti-TPD52L2/D54 Antibody (A07219, bosterbio). The staining intensity was scored as (0 = negative, 1 = weak, 2 = moderate, 3 = strong) according to the expression level and the staining area was scored as (0 = 0%, 1 = 1–25%, 2 = 26–50%, 3 = 51–100%) based on the percentage of positive cells. The immunostaining score (IS) was calculated by adding the score of staining intensity and area [[65]22]. RT-PCR and WB experiments The mRNA expression was analyzed by reverse transcription-polymerase chain reaction (RT-PCR). Primers for TPD52L2 transcripts amplification were forward 5’-CTCCATCCGCCACTCAAT-3’, reverse5’- CACCCACAACCTTAGACTTTAT-3’. Total RNA was extracted from five HNSCC different cell lines (cells (FD-LSC-1, Fadu, cal27, HN-6, SCC-25) vs. normal head and neck cells (NOK, FM)) using TRIzol® Reagent, and the above cells were all purchased from Guangzhou Yeshan Biotechnology Co., Ltd. After identification purity and concentration, RNA was reverse transcribed to cDNA using M-MLV Reverse Transcriptase (Promega, U.S.A.). Real-time PCR amplification was performed using an SYBR Green Realtime PCR Master Mix (Toyobo, Japan). The mRNA expression level was normalized to the expression level of the housekeeping gene GAPDH. Relative gene expression levels were calculated using 2^−△△ Ct. Each experiment was performed in triplicate. Proteins were also extracted from the above cell lines. From sample preparation to detection, the reagents for WB were in Western Blotting Application Solutions Kit. After sample preparation, equal amounts of sample protein and internal control protein were loaded onto two SDS-PAGE gels (12% and 10% concentrations, respectively). Under the premise of maintaining the same conditions as much as possible, electrophoresis and transfer of the samples to a nitrocellulose membrane were performed. The membrane was blocked for 2 h at room temperature and incubated overnight at 4℃. Primary antibodies used are as followed: anti-TPD52L2 (1:1000, A07219, Bosterbio), HRP labeled GAPDH antibody (1:10,000, KC-5G5, Aksomics), Rabbit Anti-Mouse IgG (H + L) (1:4000, 6170–05, Southern Biotech), Goat Anti-Rabbit IgG (H + L chain specific) (1:5000, 4050–05, Southern Biotech), peroxidase- Rabbit Anti- Goat IgG (1:3000, BA1060, Bosterbio), peroxidase- Rabbit Anti- Rat IgG(1:2000, BA1058, Bosterbio).Proteins were detected using Medical X-ray film (Kodak). Each experiment was performed in triplicate. Protein expression analysis in UALCAN The protein of TPD52L2 expression between normal and tumor tissues in pan-caner was analyzed using CPTAC data from the UALCAN website ([66]http://ualcan.path.uab.edu). Differences in protein level were compared using the Student’s t-test, p < 0.05 was considered statistically significant. Results Elevated expression of TPD52L2 in HNSCC Analysis of the expression of TPD52L2 in pan-cancer showed higher expression in 18 cancer subtypes compared to their normal counterpart tissue (Fig. [67]1A). Especially, TPD52L2 was over-expressed in HNSCC, which was concluded from paired and unpaired analyses in TCGA database (p < 0.001) (Fig. [68]1B, C). In [69]GSE39400 cohort, higher expression of TPD52L2 in HNSCC patients in peripheral blood cells (PBC) was validated (p < 0.001) (Fig. [70]1D). As shown in Table [71]1, TPD52L2 expression was observed to have a strong association with T stage (p = 0.019). However, TPD52L2 expression level showed no significant correlation with other clinicopathologic features. ROC analysis showed a good diagnostic accuracy in discriminating normal from cancer tissues based on TPD52L2 mRNA expression level, with an area under the curve (AUC) of 0.891 [95% confidence interval (CI): 0.856–0.926], a sensitivity of 95.5%, and a specificity of 71.7% (Fig. [72]1E). Fig. 1. [73]Fig. 1 [74]Open in a new tab Expression and diagonostic value of TPD52L2. A Pan-cancer expression of TPD52L2. Analyses of TPD52L2 expression in (B) unpaired and (C) paired samples in TCGA cohort. D TPD52L2 expression was significantly increased in HNSCC tissues compared to normal tissues from the GEO datasets([75]GSE39400). E ROC curve analysis of TPD52L2 expression in HNSCC Table 1. Clinicopathological features of HNSCC patients from TCGA dataset Clinicopatogical features Low expression of TPD52L2 High expression of TPD52L2 P value n 251 251 Gender, n (%) 0.545 Female 70 (13.9%) 64 (12.7%) Male 181 (36.1%) 187 (37.3%) Age, n (%) 0.624  <  = 60 125 (25%) 120 (24%)  > 60 125 (25%) 131 (26.1%) T stage, n (%) 0.019 T1 21 (4.3%) 12 (2.5%) T2 80 (16.4%) 64 (13.1%) T3 69 (14.2%) 62 (12.7%) T4 74 (15.2%) 105 (21.6%) N stage, n (%) 0.195 N0 124 (25.8%) 115 (24%) N1 32 (6.7%) 48 (10%) N2 83 (17.3%) 71 (14.8%) N3 3 (0.6%) 4 (0.8%) M stage, n (%) 1.000 M0 236 (49.5%) 236 (49.5%) M1 3 (0.6%) 2 (0.4%) Smoker, n (%) 0.422 No 52 (10.6%) 59 (12%) Yes 195 (39.6%) 186 (37.8%) Alcohol history, n (%) 0.866 No 81 (16.5%) 77 (15.7%) Yes 168 (34.2%) 165 (33.6%) [76]Open in a new tab Prognostic significance of TPD52L2 Survival analysis demonstrated that TPD52L2 over-expression predicted worse overall survival (OS) of patients with BLCA, MESO, LGG, LUADLUSC, LIHC, BRCA, HNSCC (Fig. [77]2A, C-H). A similar result in HNSCC was confirmed in the GEO validation dataset ([78]GSE65858) (Fig. [79]2B). Fig. 2. [80]Fig. 2 [81]Open in a new tab TPD52L2 is associated with survival outcome. A Overall survival (OS) of TPD52L2 in TCGA HNSCC cohort. OS of TPD52L2 analysis from Kaplan–Meier survival curves on [82]GSE65858 (B), in TCGA BLCA (C), MESO (D), LGG (E), LUADLUSC (F), LIHC (G), BRCA (H) cohort As shown in the forest plot (Fig. [83]3A, B), univariate analysis of TCGA showed age, gender, N stage, M stage and TPD52L2 were significant prognostic factors for OS in HNSCC patients, and multivariate analysis showed that TPD52L2 (HR = 1.439, p = 0.01) had an independent prognostic value. these results were consistent with that of the [84]GSE65858 dataset (Fig. [85]3C, D). Fig. 3. [86]Fig. 3 [87]Open in a new tab Univariate (A) and multivariate (B) cox regression analyses of TPD52L2 associated with overall survival in HNSCC patients from TCGA. Univariate (C) and multivariate (D) regression analysis of TPD52L2 from GEO database Tpd52L2 co-expression gene analysis As shown in Fig. [88]4A, 4653 genes (dark red dots) showed significant positive correlation with TPD52L2, while 9518 genes (dark green dots) had significant negative correlation. The top fifty significant genes positively or negatively associated with TPD52L2 are shown by heat map in Fig. [89]4B, C. TPD52L2 expression had a strong positive relationship with the expression levels of UBE2V1 (r = 0.749), PSMA7 (r = 0.689), and ARFGAP1 (r = 0.655). It also had a strong negative association with MYLIP (r = -0.621), BCL2 (r = -0.621), and REPIN1 (r = -0.617). Fig. 4. [90]Fig. 4 [91]Open in a new tab Co-expression genes of TPD52L2 in HNSCC analyzed by LinkedOmics database. A All genes highly correlated with TPD52L2 as identified by Pearson correlation test in the HNSCC cohort. B, C Heat maps showing the top 50 genes positively and negatively correlated with TPD52L2 in HNSCC. Red indicates positively correlated genes, and blue indicates negatively correlated genes. D – G Significantly enriched GO annotations and KEGG pathways of TPD52L2 in HNSCC cohort The outcomes of the GO analysis showed that TPD52L2 co-expressed genes were involved mainly in cell junction organization, negative regulation of calcineurin-NFAT signaling cascade, focal adhesion, cell-substrate adherent junction, structural constituent of cytoskeleton (Fig. [92]4D-F). The KEGG pathway analysis showed enrichment in Phagosome, Gap junction (Fig. [93]4G). Gene alteration of TPD52L2 The genomic alteration frequency of TPD52L2 was higher than 1% in HNSCC patients with “amplification” as the primary alterations (Fig. [94]5A). In evaluating the CNV status of TPD52L2 in pan-cancer, we found that TPD52L2 expression was positively correlated with CNA in HNSCC (p < 0.001) (Fig. [95]5B). Fig. 5. [96]Fig. 5 [97]Open in a new tab Gene alteration of TPD52L2. A Mutation and CNA status of TPD52L2 in TCGA-pan-cancer using the cBioPortal database. B Correlation between TPD52L2 mRNA and CNA in TCGA-pan-cancer Tpd52L2 genetic alteration status analysis As the prognosis of patients with high expression of TPD52L2 is worse than that of the patients with low expression, we speculated whether the expression of TPD52L2 is related to the mutations of some common cancer-promoting genes. A total of 518 samples were tested for mutations, of which 422(81.5%) were plotted. We further analyzed the gene mutations (such as TP53, PIK3CA, and FAM135B) in high and low TPD52L2 expression groups (Fig. [98]6A). These results demonstrated that the expression of TPD52L2 was potentially related to the mutation of TP53, PIK3CA, and FAM135B genes. Fig. 6. [99]Fig. 6 [100]Open in a new tab Gene mutant frequency (top 20) in high- and low-TPD52L2 expression groups. A total of 518 samples were tested for mutations, of which 422(81.5%) were plotted. Gene (such as TP53, PIK3CA, and FAM135B) mutations in high and low TPD52L2 expression groups were analyzed using ComplexHeatmap in R from TCGA database Correlation between TPD52L2 and immune infiltration An immune infiltration score for 22 immune cells was assigned for 39 cancers based on their gene expression signatures which were downloaded from TCGA pan-cancer dataset. Data analysis using the “psych” package showed that TPD52L2 expression was significantly correlated with immune cell infiltrate in 38 cancers, including HNSCC (Fig. [101]7A). Fig. 7. [102]Fig. 7 [103]Open in a new tab Correlation Between TPD52L2 and Immune Infiltration. A Based on TCGA pan-cancer dataset, an immune infiltration score of 39 cancers for 22 immune cells was analyzed. B TPD52L2 CNV status affected the infiltrating levels of B cells, CD8 + T cells, CD4 + T cells, Macrophages, neutrophils and dendritic cells. C The distribution of 24 subtypes of immune cells in low- and high- TPD52L2 expression groups. Box diagram showing the stromal scores (D), immune scores (E), and ESTIMATE scores (F) in the low- and high- TPD52L2 groups analyzed by the ESTIMATE algorithm The TIMER analysis suggested that the CNV of TPD52L2 was significantly related to the infiltration levels of B cells, CD8 + cells, CD4 + cells, macrophages, neutrophils and dendritic cells (Fig. [104]7B). Additionally, the differences of 24 types of TIICs between the high-TPD52L2 expression and low- groups were compared. The results indicated that the high-expression group had fewer activated DCs (aDC) (p < 0.001), B cells (p < 0.001), CD8 T cells (p < 0.001). Other TIICs showed no statistically significant intergroup differences (Fig. [105]7C). The stromal scores (Fig. [106]7D), immune scores (Fig. [107]7E) and ESTIMATE scores (Fig. [108]7F) of high-TPD52L2 and low- group were performed by R package “ESTIMATE”. The ESTIMATE algorithm suggested that the high-TPD52L2 group had lower immune scores (p < 0.001) and ESTIMATE scores than the low- group. Correspondingly, we evaluated the correlations between immune responses and the expression of TPD52L2 by Spearman's correlation coefficient. There were 14 types of immune cells negatively correlated with TPD52L2 expression, including aDCs (r =  − 0.177, p < 0.001), B cells (r =  − 0.361, p < 0.001), CD8^+ T cells (r =  − 0.270, p < 0.001), cytotoxic cells (r =  − 0.322, p < 0.001), DCs (r =  − 0.141, p = 0.002), Mast cells (r =  − 0.177, p < 0.001), NK CD56 bright cells (r =  − 0.188, p < 0.001), NK CD56 dim cells (r =  − 0.163, p < 0.001), pDC (r =  − 0.241, p < 0.001), T cells (r =  − 0.332, p < 0.001), T helper cells (r =  − 0.119, p = 0.008), TFH (r =  − 0.274, p < 0.001), Th17 cells (r =  − 0.194, p < 0.001), and Treg (r =  − 0.223, p < 0.001) (Fig. [109]7E). Correlation between the expression of TPD52L2 and icgs in HNSCC In this study, the correlation between the expression of TPD52L2 and that of the over 40 common immune checkpoint genes was analyzed using spearman correlation coefficient. In HNSCC, TPD52L2 expression was positively correlated with 4 immune checkpoint markers, including PDCD1LG2, EGFR, CD44, CD274 (Fig. [110]8A). Fig. 8. [111]Fig. 8 [112]Open in a new tab Correlation between TPD52L2 expression and ICGs in HNSCC. A Results based on TCGA database. B Immunohistochemistry on the tissue microarrays displayed CD274, EGFR, PD-L2, TPD52L2 protein levels in normal tissues (n = 5) and HNSCC tissues (n = 5) (40*10 times), and representative images scored by Image-Pro Plus 6.0 software In addition, protein levels of TPD52L2 and three ICGs (PDL1, PDL2, EGFR) were further explored in our own clinical samples (n = 5 paired) based on IHC assay. In our samples, the protein level of TPD52L2 and three ICGs PDL1, PDL2, EGFR) was also significantly up-regulated in HNSCC samples compared with the paired normal tissues (p < 0.05) (Fig. [113]8B). These results demonstrated that TPD52L2 gene may play a crucial role in tumor immunity and may have impactful implications for the use of ICIs against HNSCC. Cell line experiment verifies the overexpression of TPD52L2 in HNSCC Seven cell lines (five HNSCC cell lines (FD-LSC-1, Fadu, cal27, HN-6, SCC-25)) vs. two normal control cell lines (NOK, FM) were used to explore the mRNA and protein expression by qRT-PCR and WB. As shown in Fig. [114]9, the expression level of TPD52L2 in the specimens of HNSCC cell lines was significantly higher than that in normal cell lines. The WB assay result showed that the protein levels of these HNSCC cells were also significantly higher than in normal control cells (p < 0.001). The original blots are presented in Supplementary Fig. 1A, B. Fig. 9. [115]Fig. 9 [116]Open in a new tab The expression of TPD52L2 in vitro. A PCR analysis of the TPD52L2 mRNA levels in HNSCC cells and normal head and neck cells. B Western blotting analysis of the expression of TPD52L2 in HNSCC cells and normal head and neck cells. GAPDH was used as a loading control. The quantitative analysis result is shown in the lower panel. C TPD52L2 protein expression in pan-cancer cohorts in TCGA by UALCAN To validate the TPD52L2 protein levels of normal and tumor tissues, the UALCAN website (http:// ualcan.path.uab.edu) was utilized. Based on CPTAC data, the protein expression of TPD52L2 was upregulated in HNSCC compared with normal controls (p < 0.001). Discussion The tumor protein D52 (TPD52) family includes TPD52, -53, -54 (also known as TPD52L2) and -55 [[117]23], and they have been demonstrated to associated with the tumorigenesis and progression in several cancers. To exemplify, the overexpression of TPD52 in nonmalignant 3T3 fibroblasts induced an increase in the growth speed and enabled the cells to exist in an anchorage-independent manner in vitro as well as to undergo metastatic growth in vivo [[118]24], and overexpression of TPD52 led to increased tumor growth in cancer cells [[119]25]. TPD53 regulates the cell cycle and is highly upregulated at the G2-M phase transition [[120]26]. In this study, we used data from TCGA database and UALCAN website to explore the expression of TPD52L2 and its clinical significance in HNSCC. We found that mRNA level and protein level of TPD52L2 (TPD54) in HNSCC tissues was higher than that in normal tissues. Next, qRT-PCR and WB verified the previous bioinformatics analysis results. ROC analysis also showed the overexpression of TPD52L2 can be used to distinguish between HNSCC and normal tissues. In addition, we found that the patients with high expression of TPD52L2 showed a much worse prognosis. Our cox regression analysis demonstrated that the expression level of TPD52L2 and age, N stage and M stage were independent factors for the prognosis of HNSCC patients. In pan-cancer analysis, we determined the high expression of TPD52L2 in pan-cancer and the KM analysis showed that TPD52L2 was significantly correlated with OS prognostic indicators of BLCA, MESO, LGG, LUADLUSC, LIHC and BRCA. Recent research has also shown that TPD52L2(TPD54) was upregulated in a variety of cancer tissues, and its expression level was associated with tumor progression and poor prognosis in prostate cancer [[121]16], basal breast cancer [[122]27] and lung adenocarcinoma [[123]14]. Pan et al. [[124]17] found that depletion of TPD52L2 could remarkably inhibit proliferative and colony-forming ability of liver cancer cells (SMMC-7721). To better understand the potential mechanism and function of TPD52L2 in the development of HNSCC, we conducted GO and KEGG enrichment analysis of TPD52L2 co-expression genes. Interestingly, the results showed that cell senescence and immune-related pathways were significantly enriched, e.g., calcineurin-NFAT (nuclear factor of activated T cells) signal cascade negative regulation pathway and calcineurin-mediated signal transduction negative regulation pathway. Previous studies have shown that inhibition of calcineurin-NFAT can inhibit cancer cell senescence, downregulate p53 expression, and promote the occurrence of skin squamous cell carcinoma [[125]28]. Cell aging is of great significance to the life activities of human body, including the regulation of embryonic development, tissue repair and tumor inhibition. And the loss of ‘Cell Aging’ characteristics of cells is related to the occurrence and progression of tumors. Our results suggest that TPD52L2 may affect the aging process of cells by regulating the negative cascade of calcineurin-NFAT signal cascade and play an important role in the occurrence of cancer. At the same time, our gene enrichment results also found that cell migration-related pathways such as focal adhesion and ER to Golgi vesicle-mediated transport were significantly enriched. Larocque [[126]13] first found that TPD52L2 is involved in a variety of membrane transport pathways: anterograde transport, recycling and Golgi integrity. Another study [[127]29] found that the only known marker of intracellular nano-vesicles (INVs, diameter 30 nm) is TPD52L2, and TPD52L2 binds directly to the cell membrane and binds to INVs through its C-terminal conserved positively charged motif, which researchers point out may be related to cancer cell migration and invasion. Therefore, TPD52L2 is likely to use its own INVs to transport goods to help achieve tumor cell migration. It is well known that tumor cell migration plays an important role in the development of cancer, and focal adhesion kinase (FAK) [[128]30] has been proved to be involved not only in tumor proliferation, but also in many processes such as tumor cell migration and angiogenesis, which eventually leads to further metastasis and deterioration of tumor. In general, our enrichment analysis results suggest that TPD52L2 may affect the progression of HNSCC and the prognosis of patients by inhibiting immunity and promoting tumor cell migration. Apart from that, our study revealed that the mutation frequency of TP53 in 442 HNSCC samples was 77.4% and it was significantly higher in high-TPD52L2 group than the low- TPD52L2 group. Mutations in the TP53 gene can lead to loss of tumor suppressor activity of its encoded protein p53, which is seen as a driver of a variety of tumorigenesis. TP53 mutations often occur in cancers and are associated with poor prognosis in many cancers [[129]31]. Growing evidence showed that tumor microenvironment (TME) is important for tumor pathogenesis, where tumor cells usually establish tumor microenvironment to enrich kinds of properties [[130]32, [131]33]. Immune-related cells and stromal cells are the main components of TME. We found that overexpression of TPD52L2 and activation of dendritic cells (p < 0.001), B cells (p < 0.001), CD8^+ T cells (p < 0.001) were negatively correlated. There were also differences in immune score and ESTIMATE score between TPD52L2 high expression group and low expression group, and the two scores in high expression group were significantly lower than those in TPD52L2 low expression group (p < 0.001). These results suggest that the overexpression of TPD52L2 may affect the progression and prognosis of HNSCC by regulating the level of immune cell infiltration and inhibiting immune anti-tumor function. As a new treatment method, immunotherapy is based on a variety of ways to activate the immune system in order to achieve the purpose of cancer treatment, which has dramatically improved the outlook of cancer treatment [[132]34]. Now many literatures have reported the clinical application with the immune checkpoint inhibitor in patients with locally advanced, HPV-unrelated head and neck cancer [[133]35]. Therefore, we explored the correlation between TPD52L2 expression and immune checkpoint molecules by TCGA database and IHC. We observed that the expression of TPD52L2 was positively correlated with four immune checkpoint molecules, including PDCD1LG2, EGFR, CD44 and CD274. The results show that TPD52L2 may play a key role in tumor immunity and may play an important role in the treatment of HNSCC with ICIs. However, it is important to acknowledge the limitations of our study. Firstly, our research primarily relied on bioinformatic analyses and experiments conducted on cell lines, lacking in vivo and in vitro functional experiment verification. A previous study reported that their in vitro experiments suggested that TPD52L2 has negative effects on cell proliferation and migration in OSCC cells, while this result was not confirmed by in vivo experiments [[134]36]. Although our study confirmed the elevated expression of TPD52L2 mRNA and protein in a variety of head and neck squamous cell carcinomas (including OSCC), no further in vitro and in vivo experiments were conducted. It is worth noting that in vivo experiments can be influenced by specific cytokines that interact with TPD52L2, thereby potentially contributing to the variability in cellular backgrounds. Subsequently, we intend to conduct in vitro functional experiments and develop a TPD52L2-deletion mouse model to substantiate the role of TPD52L2 in regulating tumor proliferation and metastasis. This verification is crucial for advancing our results towards potential clinical applications. Conclusion Together, our study confirmed that TPD52L2 is elevated in HNSCC and it could act as a reliable independent prognostic factor. In addition, TPD52L2 was associated with TIICs and immune checkpoints. Our results suggest that TPD52L2 may serve as a prognostic biomarker and it may provide a new therapeutic option for patients with HNSCC. Supplementary Information [135]Supplementary Material 1.^ (159.9KB, pdf) Acknowledgements