Abstract Background Renal Cell Carcinoma (RCC) is a leading cause of cancer-related mortality worldwide, with Clear Cell Renal Cell Carcinoma (ccRCC) comprising 75% of cases. Surgical resection remains the cornerstone of treatment for localized RCC, but its asymptomatic progression and lack of reliable early biomarkers often result in advanced disease at diagnosis. Collagen VI alpha-2 chain (COL6A2), an extracellular matrix protein, has been implicated in tumor progression and metastasis. Despite its established roles in other malignancies, the specific contribution of COL6A2 to ccRCC pathogenesis is poorly understood. Objective This study aims to systematically investigate COL6A2 expression in ccRCC, its prognostic value, and its potential impact on the tumor immune microenvironment, cancer stem cell characteristics, and drug response. Methods The mRNA and protein expression data for ccRCC were sourced from TCGA, GEO, CPTAC, and ICPC. Single-cell and spatial transcriptomic data were processed using Seurat with quality control measures. Clinical correlations and survival analyses, including immune infiltration and COL6A2 expression, were assessed using Cox regression and Kaplan-Meier curves. Cancer stemness was evaluated using six stemness indices. Differential expression and pathway analyses (GO, KEGG, GSEA) were performed with DESeq2 and clusterProfiler. Drug sensitivity and immunotherapy response were predicted using GDSC, CTRP, and TIDE databases. Functional studies, including colony formation and invasion assays, as well as in vivo xenograft models, assessed the impact of COL6A2 on tumor progression and therapy response. Results COL6A2 expression was significantly upregulated in ccRCC compared to normal tissues. High COL6A2 expression correlated with poorer overall survival (OS), progression-free interval (PFI), and progression-free survival (PFS), establishing it as an independent prognostic factor for ccRCC. Additionally, COL6A2 expression was positively associated with immune-suppressive cell infiltration, suggesting an immunosuppressive tumor microenvironment. COL6A2 was also linked to enhanced stem cell-like properties, invasiveness, and metastatic potential. Pathway enrichment analyses revealed that COL6A2 may influence tumor progression by regulating the epithelial-mesenchymal transition (EMT) process and activating the PI3K-Akt signaling pathway. Notably, high COL6A2 expression correlated with enhanced responsiveness to sunitinib but resistance to immunotherapy, highlighting its dual role in therapy selection. Conclusion This study identifies COL6A2 as a powerful prognostic biomarker and a driver of ccRCC progression through EMT and immune suppression. Targeting COL6A2 holds promise for improving immunotherapy efficacy and personalizing treatment strategies, offering new hope for ccRCC patients facing limited options. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-025-06793-9. Keywords: Renal cancer, COL6A2, Tumor microenvironment, Pharmacological prediction, Prognostic significance Introduction Renal Cell Carcinoma (RCC), is the predominant form of malignant kidney tumors, with Clear Cell Renal Cell Carcinoma (ccRCC) accounting for 75% of cases [[30]1]. Globally, approximately 315,000 new cases of RCC are diagnosed annually, ranking it among the top ten most common cancers worldwide [[31]2]. Due to RCC’s low responsiveness to traditional radiotherapy and chemotherapy, surgical resection remains the preferred treatment for localized RCC. However, because of RCC’s asymptomatic growth and the lack of effective early diagnostic markers, approximately 30% of patients present with distant metastasis at diagnosis [[32]3]. Metastatic RCC treatment primarily relies on targeted therapies, such as Sorafenib and Sunitinib, but their high cost, resistance, toxicity, and side effects limit their widespread clinical application [[33]4]. Therefore, it is imperative to delve into the molecular mechanisms of RCC and identify new biomarkers and molecular targets. Collagen VI (COL6), a member of the collagen family, is ubiquitously expressed in various tissues [[34]5]. COL6-dependent assembly of vascular basement membranes is crucial for vascular function and tumor progression [[35]6]. The COL6A2 gene encodes the α2 chain of Collagen VI, which, along with the α1 (VI) and α3 (VI) chains encoded by COL6A1 and COL6A3, forms the functional collagen VI structure [[36]7]. Studies have shown that COL6A2 is strongly associated with tumor proliferation, migration, and invasion. For instance, COL6A2 significantly inhibits the proliferation of human bladder cancer cells, induces G1-phase cell cycle arrest, and suppresses wound healing and invasion by inhibiting matrix metalloproteinases (MMP-2 and MMP-9) activity [[37]8]. Additionally, COL6A2 is implicated as a key gene in the recurrence of Low-Grade Glioma (LGG), with the LncRNA HOXA-AS2/miR-184/COL6A2 axis serving as a potential regulatory mechanism for LGG recurrence [[38]9]. Although previous studies have shown that COL6A2 expression is significantly elevated in ccRCC [[39]10], its functional role and clinical significance in ccRCC remain unclear. This study aims to systematically explore the expression pattern of COL6A2 in ccRCC, its prognostic value, and its potential impact on the tumor immune microenvironment, cancer stem cell characteristics, and drug responsiveness. Materials and methods Data sources and gene expression analysis Bulk RNA-seq databases mRNA expression profiles and clinical data for various tumor types including bladder urothelial carcinoma (BLCA), breast invasive carcinoma (BRCA), cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), cholangiocarcinoma (CHOL), colon adenocarcinoma (COAD), esophageal carcinoma (ESCA), glioblastoma multiforme (GBM), head and neck cancer (HNSC), kidney chromophobe (KICH), kidney renal clear cell carcinoma (KIRC), kidney renal papillary cell carcinoma (KIRP), liver hepatocellular carcinoma (LIHC), lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), pancreatic adenocarcinoma (PAAD), pheochromocytoma and paraganglioma (PCPG), prostate adenocarcinoma (PRAD), rectum adenocarcinoma (READ), sarcoma (SARC), skin cutaneous melanoma (SKCM), stomach adenocarcinoma (STAD), thyroid carcinoma (THCA), thymoma (THYM), uterine corpus endometrial carcinoma (UCEC) were obtained from The Cancer Genome Atlas (TCGA) database. Public mRNA expression datasets for ccRCC were obtained from the Gene Expression Omnibus (GEO) database, including [40]GSE66272 (27 tumor and 27 adjacent normal samples), [41]GSE53757 (76 normal samples, 68 tumor samples), [42]GSE36895 (23 normal samples, 29 tumor samples), [43]GSE46699 (63 normal samples, 69 tumor samples), [44]GSE167093 (155 normal samples, 348 tumor samples), and [45]GSE126964 (11 normal samples, 55 tumor samples).Protein expression profiles for ccRCC and normal tissues were obtained from the Clinical Proteomic Tumor Analysis Consortium (CPTAC) and the International Cancer Proteogenome Consortium(ICPC) databases. Immunohistochemistry (IHC) staining images for COL6A2 were retrieved from the Human Protein Atlas (HPA) database. Single Cell and Spatial Transcriptomics RNA-seq Databases: Single cell and spatial transcriptomics RNA-seq datasets were obtained from the GEO database ([46]GSE210038, [47]GSE210041), including 7 ccRCC samples and 2 adjacent normal samples. Quality control and downstream analysis were performed using the standard pipeline of the R package Seurat (version 5.0.1) [[48]11], filtering out low-quality cells with fewer than 200 expressed genes, more than 30% mitochondrial genes, and genes expressed in fewer than three cells. After excluding housekeeping and mitochondrial genes, a total of 22,935 genes and 61,343 cells were retained. Cluster annotation was conducted following standard data normalization, dimensionality reduction, and clustering, utilizing previously reported cellular markers. Pseudotime analysis was conducted using the R package Monocle [[49]12]. Human samples In accordance with ethical guidelines, informed consent was obtained from patients, and approval was granted by the institutional research ethics committee of Zhongnan Hospital, Wuhan University. Fresh renal cancer tissues and corresponding adjacent normal tissues were obtained from Zhongnan Hospital of Wuhan University. These tissues were stored at -80 °C. Clinical correlation analysis and prognostic analysis The most recent survival data for the all cohorts of TCGA were retrieved from the study [[50]13].The relationship between COL6A2 expression levels and clinical parameters such as TNM, Stage, and age in TCGA-KIRC cohort was assessed. Tumor samples from the TCGA-KIRC cohort were stratified into high-expression and low-expression groups using the median COL6A2 expression level as the cutoff. The association between COL6A2 expression and overall survival (OS), progression-free interval (PFI), progression-free survival (PFS) in ccRCC patients was analyzed using univariate Cox regression. The association between COL6A2 expression groups and OS, PFI, PFS was evaluated with Kaplan-Meier (KM) survival analysis. Multivariate Cox regression was employed to determine the independent impact of COL6A2 expression and clinical variables on OS, PFI, and PFS. To enhance prediction reliability of ccRCC patient outcomes, a prognostic nomogram integrating COL6A2 expression and clinical features was constructed based on the multivariate Cox regression results. Calibration curves were plotted to compare nomogram-predicted survival probabilities against observed outcomes. Predictive accuracy was assessed using receiver operating characteristic (ROC) curves. Statistical analyses were conducted with the R package “survival” and visualized using the “survminer” package. Nomogram construction was performed using the “rms” package. Analysis of the correlation between COL6A2 expression and immune cell infiltration We used the ESTIMATE algorithm to calculate the correlation between immune scores and COL6A2 expression levels, elucidating the relationship between COL6A2 and the immune contexture of ccRCC. We utilized several algorithms, including TIMER, Quantiseq, CIBERSORT, EPIC, and MCPcounter, to investigate the correlation between immune subset proportions and COL6A2 expression levels, assessing the impact of COL6A2 on immune cell infiltration in the ccRCC immune microenvironment. Analysis of the correlation between COL6A2 and cancer stemness index In this study, six cancer stemness indices, derived from previous research, were calculated based on mRNA expression and DNA methylation signatures [[51]14]. These include the RNA Stemness Score (RNAss), based on RNA expression, the Epiregulin Expression Stemness Score (Epiregulin Expression Stemness Score), based on epigenetically regulated RNA expression involving 103 genes, the DNA Stemness Score (DNAss), based on DNA methylation, combining 219 stem cell signature probes, the Epigenetically Regulated DNA Methylation Stemness Score (EREG-METHss), based on 87 probes, the Differentially Methylated Positions Stemness Score (Differentially Methylated Positions Stemness Score), based on 62 differentially methylated probes, and the Enhancer Elements/DNA Methylation-based Stemness Score (ENHss), based on 82 enhancer elements/DNA methylation probes. Additionally, this study integrates the stemness indices with COL6A2 gene expression data to explore the correlation between COL6A2 expression and tumor stemness. Differential gene and pathway enrichment analysis To investigate the functions and pathways associated with COL6A2, we used the R package DESeq2 to identify differentially expressed genes (DEGs) between high and low COL6A2 expression groups. The selection criteria were based on an adjusted P-value < 0.05 and an absolute log fold change (|logFC|) > 1. Next, we used the R package ‘clusterProfiler’ to perform Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and Gene Set Enrichment Analysis (GSEA) on the identified DEGs [[52]15]. GSEA was performed using gene sets from the MSigDB database, including “c5.go.v2023.2”, “c2.cp.kegg_legacy.v2023.2”, and HALLMARK gene sets, to evaluate the enrichment of distinct pathways and molecular mechanisms between high and low COL6A2 expression groups. For GSEA, we set parameters with a minimum gene set size of 5 and a maximum of 5000, performing 1000 resampling iterations. Additionally, gene sets related to the Epithelial-Mesenchymal Transition (EMT) pathway were compiled from CancerSEA, and GSVA enrichment scoring was performed using the R package ‘GSVA’. Drug sensitivity prediction Data from the GDSC (Genomics of Drug Sensitivity in Cancer) and CTRP (Cancer Therapeutics Response Portal) databases were used to construct our training set. We then employed the R package ‘oncoPredict’ to predict drug sensitivity scores for each patient with ccRCC, assessing their response to a variety of pharmaceuticals [[53]16]. Briefly, ridge regression models were trained using large-scale gene expression and drug sensitivity data (training dataset). These models were subsequently applied to independent gene expression profiles (testing dataset) to generate drug sensitivity predictions. Predicted drug sensitivity scores demonstrated close alignment with experimental IC50 values, where lower scores corresponded to lower IC50 levels, indicating potentially enhanced patient therapeutic response. Additionally, the TIDE (Tumor-Immune Dysfunction and Exclusion) online tool was used to predict the response of patients with ccRCC to immunotherapy, based on TCGA-KIRC transcriptome data. The TIDE tool provides an in-depth analysis of immune cell infiltration and functional status within the tumor microenvironment, aiding in the evaluation of immune cell composition and their potential impact on tumor growth and therapeutic response [[54]17]. Cell culture and transfection Human RCC cell lines and RENCA cell were obtained from Land Biotechnology (Guangzhou, China). Cells were all cultured in RPMI 1640 medium (Gibco, China), supplemented with 10% fetal bovine serum (FBS, Gibco, USA), and maintained in a humidified incubator at 37 °C with 5% CO^2. ShRNA plasmids were synthesized by Beijing Tsingke Biotech Co.,Ltd. Transfection of ACHN and OS-RC-2 cells was conducted using Lipofectamine 3000 (Invitrogen, USA). 48 h post-transfection, cells were collected for further experimental analyses. RT-qPCR & Western blot RT-qPCR and Western blot were performed as previously described [[55]18]. Primer sequences for amplification were as follows: COL6A2 forward primer, 5’-TCAAGGAGGCTGTCAAGA-3’ and reverse primer, 5’-GGCGATGGAGTAGAGGTT-3’; GAPDH forward primer, 5’-GAAGGTGAAGGTCGGAGTC-3’ and reverse primer, 5’-GAAGATGGTGATGGGATTTC-3’. Primary antibodies, anti-COL6A2 (1:1000), anti-E-cadherin (1:1000), anti-N-cadherin (1:1000), anti-β-catenin (1:1000), anti-Vimentin (1:1000), anti-Snai1 (1:1000), anti-GAPDH(1: 10000, proteintech), were used for probing, with GAPDH serving as a loading control. Colony formation assay, transwell migration assay, scratch assay, cell counting kit-8 and IC50 assay Colony formation, tumor invasion, and scratch assays were performed as previously described [[56]18]. Cell proliferation of ccRCC cell lines was assessed using the CCK-8 assay (MedChem Express). 2000 cells per well were seeded in 96-well plates with 100 µL of RPMI 1640 medium supplemented with 10% FBS for 96 h. After adding 10 µL of CCK-8 solution, the cells were incubated for 2 h. Absorbance at 450 nm was measured daily using a DNM-9606 plate reader (Perlong, Beijing). These cells were exposed to varying concentrations of sunitinib (0, 0.01, 0.05, 0.10, 0.50, 1.00, 2.50, 5.00, 7.50, 10.00 µmmol/L) for 24 h, followed by the addition of 10 µL of CCK-8 reagent and incubation at 37 °C for 1 h. Each experiment was performed in triplicate. The IC[50] (half maximal inhibitory concentration) value for sunitinib was determined using best-fit dose-response inhibition curves in GraphPad Prism 10.0 software. In vivo tumor xenograft model In accordance with standard statistical methods, we randomly selected 6 mice from each group, following the approach commonly used in most studies. BALB/c-nu and BALB/c mice (4–5 weeks old, female) were purchased from Sibeifu Company (Beijing, China). The mice were randomly assigned to different experimental groups. The cells were mixed with 50 µL of Matrigel (Corning) and 50 µL of PBS, then injected subcutaneously into the backs of the mice. A total of 6 × 10^6 OS-RC-2 cells were transfected with shControl and sh-COL6A2. Starting on the 5th day after subcutaneous tumor implantation, the mice were treated with or without sunitinib (25 mg/kg, administered orally once daily for 14 days). Tumor size was monitored and assessed every 2 days. Anti-mouse PD-1 monoclonal antibody (clone RMP1-14), purchased from Bio X Cell, was used for anti-PD-1 treatment, while saline was used as the isotype control. Additionally, 1 × 10^7 Renca cells were separately infected with shControl and sh-COL6A2. From the 5th day after subcutaneous tumor implantation, the mice were treated with or without anti-mouse PD-1 antibody (10 mg/kg, administered via intraperitoneal injection twice weekly). Tumor size was monitored and assessed every 3 days. Tumor volume was calculated using the formula: (L × W²)/2. Mice were euthanized, and tumors were excised and weighed. The experiment was conducted in accordance with the guidelines established by the Medical Ethics Committee of Zhongnan Hospital, Wuhan University (approval MRI2024-LAC209). Statistical analysis All statistical analyses in this study were performed using R software (version 4.1.3) and GraphPad Prism 10 software (La Jolla, CA, USA). Intergroup differences were assessed for significance using Student’s t-test and the Wilcoxon rank-sum test, while correlation analyses were conducted using the Spearman correlation coefficient. Statistical significance was defined as * P < 0.05, ** P < 0.01, *** P < 0.001, and **** P < 0.0001. Results Pan-cancer landscape and prognostic significance of COL6A2 expression We first utilized TCGA database to analyze the expression patterns of the COL6A2 gene across various cancer types. The results showed that COL6A2 expression was significantly elevated in BRCA, CHOL, GBM, HNSC, KIRC, and significantly decreased in BLCA, CESC, COAD, KICH, PRAD, UCEC compared to normal tissues (P < 0.05) (Fig. [57]1A). For cancer types with significant expression differences, survival data, including OS, PFI, and PFS, were analyzed using univariate Cox regression models to assess the relationship between COL6A2 expression and prognosis. The analysis showed that COL6A2 acted as an independent risk factor for poor prognosis in most cancers with significant expression differences (Fig. [58]1B). Further analysis revealed that COL6A2 had statistical significance in BLCA, KICH, GBM, and KIRC (Fig. [59]1C). However, in BLCA and KICH, COL6A2 expression was inversely correlated with survival, requiring further investigation into the underlying mechanisms. Given the established role of COL6A2 in GBM, this study focused on its role in KIRC. COL6A2 expression in ccRCC was analyzed using TCGA, CPTAC, and ICPC databases, revealing significantly higher expression at both the transcriptional and protein levels compared to normal tissues (P < 0.05) (Fig. [60]1D, E). To validate this, COL6A2 expression was analyzed using the [61]GSE66272, [62]GSE53757, [63]GSE36895, [64]GSE46699, [65]GSE167093, and [66]GSE126964 datasets from the GEO database, with results confirming higher expression in ccRCC compared to normal tissues (P < 0.05) (Fig. [67]1H). Additionally, IHC staining images from HPA confirmed the high expression of COL6A2 in renal cancer tissues (Fig. [68]1F). Finally, COL6A2 expression in clinical samples was validated at both the RNA and protein levels, confirming higher expression in cancer tissues compared to adjacent normal tissues (P < 0.05) (Fig. [69]1G, I). Fig. 1. [70]Fig. 1 [71]Open in a new tab COL6A2 Expression and the Correlation with Prognostic Indicators in Pan-Cancer, Particularly ccRCC. A. RNA expression of COL6A2 across various cancers and normal tissues from TCGA. B. Univariate COX regression forest plot for OS, PFI, and PFS of COL6A2. The horizontal line segments represent the 95% confidence intervals, with the two ends representing the lower limit (lower 95% HR) and upper limit (higher 95% HR) of the 95% confidence interval; the dots represent the HR values for each variable, where blue dots indicate HR < 1 and red dots indicate HR > 1; there is a vertical dashed line at HR = 1. If the line segment of a variable does not intersect with the dashed line, it means that the 95% confidence interval of the variable’s HR does not include 1, the p-value is less than 0.05, and the variable has an impact on patient survival with statistical significance. C. Venn diagram displaying tumors with differential gene expression and significant univariate COX regression for OS, PFI, and PFS. D. Differential RNA expression of COL6A2 in paired tumor and normal samples from TCGA-KIRC. E. Protein expression of COL6A2 in ccRCC tumors and normal samples from the CPTAC. F. Immunohistochemical staining of COL6A2 in renal cancer and normal samples from HPA. G. RNA expression of COL6A2 in tumor and adjacent clinical samples. H. RNA expression of COL6A2 in cancer and adjacent samples from GEO datasets. I. Protein expression of COL6A2 in collected tumor and adjacent clinical samples COL6A2 as independent prognostic factor in ccRCC Further analysis of the TCGA-KIRC dataset revealed correlations between COL6A2 expression levels and clinical characteristics of ccRCC patients. The analysis showed that patients over 65 had significantly lower COL6A2 expression than younger patients (P < 0.05) (Fig. [72]2A). Gender had no significant effect on COL6A2 expression (P > 0.05) (Fig. [73]2A). In TNM, patients in T3-4 had significantly higher COL6A2 expression than those in T1-2 (P < 0.05). Similarly, patients in stage N1 had higher expression than those in stage N0 (P < 0.05), with no significant difference observed between M stages (P > 0.05) (Fig. [74]2A). In stage, patients in Stage3-4 had significantly higher COL6A2 expression than those in Stage1-2 (P < 0.05) (Fig. [75]2A). Kaplan-Meier survival curve analysis showed that patients with high COL6A2 expression had significantly shorter OS, PFI, and PFS (Fig. [76]2B). Similar trends were observed across subgroups defined by gender, age, TNM, and Stage classifications (P < 0.05) (Fig. [77]2C, SupFigure [78]1 A, F). Multivariate Cox regression analysis confirmed COL6A2 as an independent prognostic factor for OS in ccRCC patients (P < 0.05) (Fig. [79]2D). A nomogram clinical prediction model, incorporating age, N stage, and M stage, showed high predictive accuracy for 1-year, 3-year, and 5-year survival, as demonstrated by ROC curves and calibration plots (Fig. [80]2E-G). Analysis of PFI and PFS confirmed the trends observed for OS, with high COL6A2 expression correlating with poorer prognoses. Multivariate Cox regression analysis also confirmed COL6A2 as an independent prognostic factor for these survival indicators (P < 0.05) (SupFigure [81]1B, G). Similarly, a nomogram clinical prediction model, based on PFI and PFS multivariate Cox regression analysis results, demonstrated high predictive accuracy for 1-year, 3-year, and 5-year survival, as shown by ROC curves and calibration plots (SupFigure [82]1 C-E, H-J). Fig. 2. [83]Fig. 2 [84]Open in a new tab Association of COL6A2 with clinical characteristics and prognostic indicators in KIRC. A. Expression of COL6A2 across subgroups defined by age, gender, TNM, and stage in KIRC. Clinical characteristics (left to right): TNM stage, Clinical stage, Age, Gender. B. Kaplan-Meier survival curves for OS, PFI, and PFS in high vs. low COL6A2 expression groups. C. Kaplan-Meier survival curves for OS in high vs. low COL6A2 expression groups across clinical subgroups. D. Multivariate COX regression forest plot for OS including COL6A2 and clinical indicators. E. Nomogram based on significant indicators from multivariate COX regression for predicting OS. F. Calibration curves at 1, 3, and 5 years for the OS nomogram. G. Receiver operating characteristic (ROC) curves at 1, 3, and 5 years for the OS nomogram Immune microenvironment remodeling by COL6A2 We used the ESTIMATE algorithm to calculate the correlation between immune scores and COL6A2 expression, elucidating the relationship between COL6A2 and the immune microenvironment in ccRCC. The results showed a negative correlation between COL6A2 expression and tumor purity (P < 0.05), and a positive correlation with StromalScore, ImmuneScore, and ESTIMATEScore (P < 0.05) (Fig. [85]3A). Furthermore, we used various bioinformatics tools, including TIMER, Quantiseq, CIBERSORT, EPIC, and MCPcounter, to analyze immune cell infiltration within the tumor microenvironment. TIMER analysis showed no significant correlation between COL6A2 expression and the proportion of B cells (P > 0.05), while a positive correlation was observed with CD8 + T cells, CD4 + T cells, neutrophils, macrophages, and dendritic cells, which were more abundant in samples with high COL6A2 expression (P < 0.05) (P < 0.05) (Fig. [86]3B). EPIC analysis showed that samples with high COL6A2 expression had significantly higher proportions of B cells, cancer-associated fibroblasts, CD4 + T cells, and endothelial cells compared to low expression samples (P < 0.05), while CD8 + T cells, macrophages, and NK cells showed no significant differences (P > 0.05) (Fig. [87]3C). Quantiseq analysis showed that samples with high COL6A2 expression had higher proportions of both M1 and M2 macrophages, with M2 macrophages being more abundant than M1, as well as higher proportions of NK cells, Tregs, and dendritic cells (P < 0.05), while proportions of B cells, monocytes, neutrophils, CD4 + T cells, and CD8 + T cells showed no significant differences (P > 0.05) (Fig. [88]3D). MCPcounter analysis showed that samples with high COL6A2 expression had significantly higher proportions of B cells, NK cells, monocytes, myeloid dendritic cells, neutrophils, endothelial cells, and fibroblasts (P < 0.05)(Fig. [89]3E). Although T cells and cytotoxic lymphocytes were more abundant in high expression samples (P < 0.05), the proportion of CD8 + T cells showed no significant difference (P > 0.05) (Fig. [90]3E). CIBERSORT analysis showed that Tfh cells, activated NK cells, and monocytes were more abundant in samples with low COL6A2 expression (P < 0.05), while Tregs were more abundant in samples with high COL6A2 expression (P < 0.05). No significant differences were observed in other cell types between high and low COL6A2 expression groups, including immature B cells, memory B cells, plasma cells, and CD8 + T cells (Fig. [91]3F). The comprehensive analysis suggests that samples with high COL6A2 expression have higher proportions of immune-suppressive cells, such as macrophages, Tregs, and dendritic cells, while proportions of CD8 + T cells and B cells show no significant differences. Additionally, the proportions of stromal cells, such as cancer-associated fibroblasts and endothelial cells, are significantly higher in samples with high COL6A2 expression compared to those with low expression, consistent with ESTIMATE analysis results, indicating a strong positive correlation between StromalScore and COL6A2 expression. These results suggest that, although samples with high COL6A2 expression have a higher immune score, the enrichment of tumor-associated fibroblasts and immune-suppressive cells may create an immunosuppressive microenvironment, thereby affecting tumor progression. Fig. 3. [92]Fig. 3 [93]Open in a new tab Exploration of the Relationship Between COL6A2 and Immune Infiltration in ccRCC Using Various Immune Infiltration Analyses. A. ESTIMATE. B. TIMER. C. EPIC. D. Quantiseq. E. MCPcounter. F. CIBERSORT. TCGA-KIRC tumor samples were dichotomized into high-expression (red) and low-expression (blue) groups based on the median COL6A2 expression level, as stratified in the box plot COL6A2 promotes tumor stemness and EMT progression This study further analyzed DMPss, DNAss, ENHss, EREG − METHss, EREG.EXPss and RNAss to explore the association between COL6A2 expression levels and tumor stemness. The findings revealed that in tumor samples with high COL6A2 expression, DMPss, DNAss, ENHss, EREG − METHss and EREG.EXPss scores were significantly higher than in samples with low expression (P < 0.05), while RNAss scores were relatively lower (P < 0.05) (Fig. [94]4A). These results suggest that tumor cells with high COL6A2 expression may possess stronger stem cell characteristics, invasiveness, and metastatic potential. Incorporating these stemness scores, we hypothesize that the expression level of COL6A2 may influence the sensitivity of tumor cells to treatment, thereby providing potential biomarkers for the development of personalized therapeutic strategies. Fig. 4. [95]Fig. 4 [96]Open in a new tab Relationship between COL6A2 and tumorigenesis in ccRCC: exploration of its function and molecular pathways. A. Distribution of tumorigenic scores in high vs. low COL6A2 expression groups. B. Volcano plot of differentially expressed genes between high and low COL6A2 expression groups. C. GSEA-KEGG enrichment bar plot for differentially expressed genes. D. GO Biological Process (BP) and KEGG enrichment bar plots for genes with high expression. E. GO-BP and KEGG enrichment bar plots for genes with low expression. F. Top 5 GSEA-GOBP enrichment plots for highly expressed genes. G. Uniform Manifold Approximation and Projection (UMAP) plot showing single-cell subtypes. H. UMAP plot showing COL6A2 expression across cell types. I. Volcano plot of differential analysis between tumor epithelial cells and normal renal tubular epithelial cells at the single-cell level. J. UMAP plot showing tumor cell subtypes. K. UMAP plot showing COL6A2 expression across tumor cell subtypes. L. UMAP plot showing EMT scores across tumor cell subtypes. M-N. Pseudotime trajectory plot for tumor cell subtypes. O. COL6A2 expression in the pseudotime trajectory plot Using the TCGA-KIRC dataset, we stratified tumor samples into high and low COL6A2 expression groups and performed differential gene expression analysis, identifying 996 differentially expressed genes. Specifically, compared to the low expression group, 585 genes were upregulated and 411 downregulated in the high expression group (Fig. [97]4B). GSEA was performed on these differentially expressed genes, sorted by P-value from smallest to largest. The top 10 GSEA-KEGG enriched pathways revealed that Protein digestion and absorption, ECM − receptor interaction, PI3K − Akt signaling pathway, Proteoglycans in cancer, Focal adhesion, Relaxin signaling pathway, and other pathways were significantly enriched in the high COL6A2 expression group, while Metabolic pathways were enriched in the low expression group (Fig. [98]4C). The top 5 GSEA-GOBP (biological process) enriched pathways showed that biological processes significantly associated with the high COL6A2 expression group included extracellular matrix organization, anatomical structure morphogenesis, external encapsulating structure organization, extracellular structure organization, and system development (Fig. [99]4F). Further GO and KEGG enrichment analysis of the upregulated and downregulated genes indicated that the upregulated gene set was closely related to pathways such as PI3K − Akt signaling pathway, Focal adhesion, ECM − receptor interaction, TGF − beta signaling pathway, and primarily enriched in biological processes like extracellular matrix organization, external encapsulating structure organization, collagen fibril organization, and collagen metabolic process (Fig. [100]4D, SupFigure [101]2B). The downregulated gene set was related to metabolic pathways such as steroid hormone biosynthesis, retinol metabolism, pyruvate metabolism, and involved in ion transport processes including organic anion transport, carboxylic acid transport, organic acid transport, monocarboxylic acid transport, inorganic anion transport, inorganic anion transmembrane transport, nucleosome assembly, and sodium ion transport (Fig. [102]4E, SupFigure [103]2 C). The comprehensive analysis suggests that COL6A2 influences tumor invasiveness and metastatic capacity by regulating the EMT process and is associated with the activation of the PI3K-Akt signaling pathway. Additionally, the EMT-related gene set from the CancerSEA website confirmed the role of COL6A2 in the EMT process. Correlation analysis showed that high COL6A2 expression was significantly positively correlated with the expression of EMT-related genes (SupFigure [104]2 A), and EMT-related genes were significantly upregulated in the high COL6A2 expression group (P < 0.05) (SupFigure [105]2D), confirming that COL6A2 plays an important role in the progression of ccRCC by promoting the EMT process. Single-cell and spatial profiling reveals mesenchymal-specific COL6A2 expression At the single-cell level, we used the Seurat software package to cluster cells, identifying major cell populations, including mesenchymal cells, B cells, MAST cells, immune cells, malignant cells, myeloid cells, NK cells, endothelial cells, mesangial cells, NKT cells, T cells, and normal tubule cells (Fig. [106]4G, SupFigure [107]3 A-C). By analyzing COL6A2 expression across different cell populations, we found that COL6A2 expression was significantly higher in tumor epithelial cells than in normal renal tubular cells (P < 0.05) (Fig. [108]4I). Additionally, COL6A2 expression was significantly elevated in tumor-associated fibroblasts and endothelial cells (Fig. [109]4H). Additionally, we performed a secondary clustering analysis on tumor epithelial cells, dividing them into four subtypes based on cellular markers reported in the literature: epithelial, inflammatory, intermediate, and mesenchymal (Fig. [110]4J). The analysis showed that COL6A2 was predominantly expressed in the mesenchymal subtype of tumor cells (Fig. [111]4K). Using the R package GSVA, EMT enrichment scores were analyzed for tumor epithelial cells and were found to be predominantly concentrated in the mesenchymal subtype (Fig. [112]4L). Pseudotime analysis showed epithelial tumor cells transitioning to mesenchymal tumor cells, marked by a gradual increase in COL6A2 expression, consistent with the trends of EMT-related genes (Fig. [113]4M-O, SupFigure [114]3D). Spatial transcriptomic analysis also showed a strong spatial correlation between COL6A2 expression and EMT scores (SupFigure [115]3E). These results further confirm the potential key role of COL6A2 in the EMT process of ccRCC. COL6A2 drives tumor progression via PI3K-AKT/TGF-β and EMT We began by examining the RNA expression levels of COL6A2 at the cellular level. COL6A2 expression was significantly higher in the 786-O, ACHN, and OS-RC-2 cell lines compared to normal HK2 cells (Fig. [116]5A). Among these, ACHN and OS-RC-2 cell lines, which exhibited the highest COL6A2 expression, were selected to create stable knockdown models for further functional analysis. Fig. 5. [117]Fig. 5 [118]Open in a new tab Functional exploration of COL6A2 in ccRCC at the cellular level. A. RNA expression of COL6A2 in HK2 and ccRCC cell lines. B. RNA expression of COL6A2 in OS-RC-2 and ACHN cell lines after knockdown. C. Cell proliferation curves of OS-RC-2 and ACHN cell lines following COL6A2 knockdown, measured by CCK-8 assay. D-F. Transwell Migration Assays, Colony formation assays, Scratch assays in OS-RC-2 and ACHN cell lines following COL6A2 knockdown. G. Bar graphs of Transwell Migration Assay, Colony formation assay, Scratch Assay results. H. Protein expression of COL6A2 and EMT-related markers in OS-RC-2 and ACHN cell lines following knockdown. I. Expression changes of PI3K-AKT and TGF-β signaling pathway-related markers in OS-RC-2 and ACHN cell lines following COL6A2 knockdown After successfully silencing COL6A2 (Fig. [119]5B), we used CCK-8 assays and colony formation tests to assess its impact on ccRCC cell proliferation. Inhibition of COL6A2 significantly reduced the proliferation of both ACHN and OS-RC-2 cells (Fig. [120]5C, D, G). Transwell Migration and Scratch assays showed that silencing COL6A2 significantly impaired the migratory potential of these cell lines (Fig. [121]5E, F, G). These findings underscore the critical role of COL6A2 in promoting ccRCC cell proliferation and migration. Bioinformatics analysis revealed a strong correlation between COL6A2 and the EMT process. Based on this, we investigated changes in EMT-associated markers following COL6A2 knockdown. The results showed an increase in E-cadherin, β-catenin expression and a decrease in N-cadherin, Vimentin, and Snail1 levels (Fig. [122]5H). Additionally, the study identified a strong relationship between COL6A2 and the PI3K-AKT and TGF-β signaling pathways, suggesting that COL6A2 regulates EMT, as well as tumor cell growth and invasion, through these pathways. To further validate this hypothesis, we analyzed the expression of key markers in the PI3K-AKT and TGF-β pathways after COL6A2 knockdown. Although no significant changes were observed in the protein levels of PI3K and AKT, the phosphorylation levels of these proteins were notably reduced. Furthermore, the expression of TGF was markedly decreased following COL6A2 knockdown. These results provide support for the hypothesis that COL6A2 regulates EMT, tumor growth, and invasion through the PI3K-AKT and TGF-β signaling pathways (Fig. [123]5I). COL6A2 modulates therapeutic response to targeted agents and immunotherapy Drug sensitivity analysis revealed that patients with high COL6A2 expression exhibited an increased response to targeted therapies, including sunitinib, pazopanib, axitinib, cabozantinib, and sorafenib (Fig. [124]6A). To investigate the relationship between COL6A2 and immunotherapy, we used the TIDE tool to evaluate the response of tumor samples from the TCGA-KIRC dataset to immunotherapy. Tumor samples with high COL6A2 expression had significantly higher TIDE scores and lower immunotherapy response rates compared to the low-expression group (Fig. [125]6B). Further TIDE analysis revealed elevated scores for Dysfunction, Exclusion, Myeloid-derived suppressor cell (MDSC), and cancer-associated fibroblasts (CAF) in the high-expression group, while Microsatellite Instability (MSI) scores and CD274 levels were lower, despite higher Cytotoxic T lymphocyte (CTL) proportion (Fig. [126]6B). Collectively, these findings suggest that patients with high COL6A2 expression exhibit an overall immunosuppressive profile and a poorer response to immunotherapy. Fig. 6. [127]Fig. 6 [128]Open in a new tab Key Role of COL6A2 in Regulating Sensitivity to Sunitinib and Immunotherapy in ccRCC. A. Distribution of drug sensitivity scores for Sunitinib, Pazopanib, Axitinib, Cabozantinib, and Sorafenib in high vs. low COL6A2 expression groups. B. Distribution of TIDE prediction results, TIDE scores, and other immune infiltration scores for immune therapy response in high vs. low COL6A2 expression groups. C. IC50 curves of OS-RC-2 and ACHN cell lines in response to Sunitinib following COL6A2 knockdown. D-F. Tumor growth rate and body weight changes in BALB/c-nu mice following COL6A2 knockdown with/without Sunitinib. G-I. Tumor growth rate and body weight changes in BALB/c mice following COL6A2 knockdown with/without RMP1-14 To validate the functional role and clinical relevance of COL6A2, we examined changes in tumorigenicity and sunitinib sensitivity following COL6A2 knockdown in both cellular and animal models. At the cellular level, IC50 experiments demonstrated that tumor cells with COL6A2 knockdown exhibited significantly increased IC50 values compared to controls (Fig. [129]6C). In the animal model, a subcutaneous tumor implantation experiment was conducted in nude mice. Tumor growth was significantly suppressed in the COL6A2 knockdown group compared to controls (Fig. [130]6D-F). However, after sunitinib treatment, no significant difference in tumor size was observed between the knockdown and control groups (Fig. [131]6D-F). Additionally, to explore the relationship between COL6A2 and immunotherapy, we performed subcutaneous tumor implantation experiments on immunocompetent mice using normal and COL6A2-knockdown RENCA cells. These mice were also treated with anti-PD-1 antibodies. The results showed that the knockdown group exhibited significantly reduced tumor growth rates and body weight, and displayed higher sensitivity to anti-PD-1 treatment (Fig. [132]6G-I). Discussion Renal cell carcinoma, particularly ccRCC, exhibits high incidence and mortality rates worldwide [[133]4]. COL6A2, a component of collagen VI, has demonstrated significant clinical relevance in various tumors [[134]8, [135]19, [136]20]. Research on the mechanisms of action and clinical value of the COL6A2 gene in ccRCC is still in its early stages. This study focuses on COL6A2 to investigate its potential clinical significance and biological functions in ccRCC. Through a comprehensive analysis of public databases and clinical samples, we found that COL6A2 expression in ccRCC is significantly elevated and closely associated with clinical features such as TNM staging and stage grouping. We confirmed using various statistical methods (including multivariate Cox regression, Kaplan-Meier survival curves, nomograms, and ROC curves) that COL6A2 is an independent prognostic factor for ccRCC, with high expression significantly associated with poor survival outcomes (OS, PFI, PFS). These findings suggest that COL6A2 plays a key role in prognosis assessment for ccRCC. This study explores the specific mechanisms of COL6A2 in ccRCC, focusing on differential gene analysis and pathway enrichment. The analysis reveals that ccRCC with varying COL6A2 expression levels exhibit distinct biological and functional characteristics. In ccRCC with high COL6A2 expression, pathway enrichment analysis shows that tumor biological processes are primarily related to extracellular structures, matrices, and collagen fibers, consistent with previous reports on COL6A2 function. Additionally, the high-expression group is associated with several key signaling pathways, including extracellular matrix-receptor interaction, focal adhesion, PI3K-Akt, and TGF-β pathways, which are known to play critical roles in tumor proliferation, invasion, EMT, and immune escape [[137]21–[138]25]. In contrast, ccRCC with low COL6A2 expression is primarily associated with metabolic processes, including organic/inorganic anion transport, retinol metabolism, pyruvate metabolism, and cytochrome P450-mediated drug metabolism. These findings suggest that varying COL6A2 expression levels in ccRCC may reflect tumor heterogeneity, with high expression promoting tumor development, while low expression impacts development through changes in metabolic pathways. EMT is a key process in tumor progression, influencing tumor invasiveness, metastasis, immune escape, and drug resistance, thus holding critical significance in cancer therapy [[139]26–[140]28]. Previous studies have shown that COL6A2 promotes EMT in gliomas and colorectal tumors [[141]29, [142]30]. To explore the relationship between COL6A2 and EMT in ccRCC, we compiled an EMT-related gene set and calculated an EMT score using the GSVA algorithm. In the TCGA-KIRC database, COL6A2 showed a strong positive correlation with EMT-related genes, which were significantly upregulated in the high-expression group compared to the low-expression group. At the single-cell level, tumor cells were classified into four subtypes: epithelial, inflammatory, intermediate, and mesenchymal, with COL6A2 predominantly expressed in mesenchymal tumor cells, consistent with the distribution of EMT scores. Pseudotime analysis simulating the EMT transformation from epithelial to mesenchymal tumor cells showed that CDH1 and KRT18 expression gradually decreased, while CDH2, VIM, SNAI2, TIMP1, and TGFBI were upregulated, confirming the reliability of the simulation. Meanwhile, COL6A2 expression gradually increased during this process. Spatial transcriptomic analysis showed that COL6A2 expression closely correlates with the spatial distribution of EMT scores. In summary, this study confirms that COL6A2 is closely associated with the EMT process in ccRCC. Through functional exploration and pathway enrichment analysis, this study identified three potential roles of COL6A2 in ccRCC. First, COL6A2 may promote proliferation, invasion, and metastasis in ccRCC by modulating the PI3K-AKT signaling pathway. Second, COL6A2 may promote EMT via the TGF-β signaling pathway, thereby influencing tumor development. Lastly, as a key extracellular matrix component, COL6A2 promotes tumor development by regulating the tumor microenvironment. At the cellular level, COL6A2 knockdown reduced the expression of key molecules in the PI3K-Akt and TGF-β pathways, as well as EMT-related markers, with increased E-cadherin, β-catenin expression and decreased levels of N-cadherin, vimentin, Snail1. Furthermore, COL6A2 knockdown significantly reduced the proliferation, migration, and invasion of renal cancer cells, providing preliminary evidence of COL6A2’s role in ccRCC development. Regarding the tumor microenvironment, we conducted immune infiltration analysis in this study. ESTIMATE analysis showed that high COL6A2 expression correlates with low tumor purity and high immune and stromal scores, reflecting an increased content of immune and stromal cells. Several immune infiltration methods revealed that COL6A2 is strongly correlated with immune-suppressive cells, such as M2 macrophages, myeloid-derived suppressor cells, Tregs, and dendritic cells, but not with anti-tumor immune cells like CD4 + T cells, CD8 + T cells, or NK cells. COL6A2 is strongly associated with CAFs and endothelial cells, with high expression in CAFs. CAFs promote tumor growth and invasion by producing extracellular matrix (ECM), cytokines, and growth factors, influencing the immune microenvironment [[143]31]. The ECM provides structural support for tumor cells and plays a critical role in tumor development by modulating cell signaling via interactions with surface receptors [[144]32]. ECM alterations regulate immune cell behavior, creating an immunosuppressive microenvironment and impairing immunotherapy efficacy [[145]33]. The comprehensive results suggest that COL6A2 promotes tumor immune escape by regulating the infiltration of immunosuppressive cells, thereby creating an immunosuppressive tumor microenvironment. Additionally, the interaction between COL6A2 and stromal cells enhances the tumor’s immunosuppressive environment, particularly in immune escape and EMT, creating a microenvironment that helps tumor cells evade immune surveillance and resist anti-tumor treatments. Therefore, COL6A2 plays a key role in renal cancer invasion and metastasis, and it also regulates immune escape by modulating the immune microenvironment. The Cancer Stemness Index (CSI) is a measure of the gene expression characteristics associated with cancer stem cells (CSCs) in tumor cells [[146]14]. CSCs are tumor cells with self-renewal and pluripotent differentiation capabilities, and they contribute to tumor initiation, progression, metastasis, and resistance to therapy [[147]34]. Cancer stemness analysis showed that the high COL6A2 expression group had higher tumor stemness scores, suggesting that COL6A2 is associated with the stemness characteristics of ccRCC. Tumor stem cells possess self-renewal and multidirectional differentiation potential, playing a critical role in tumor initiation, development, invasion, and metastasis. Tumor stem cells acquire invasiveness through the EMT process, which involves the loss of adhesion and enhanced migration, contributing to metastasis and drug resistance. This study also included pharmacological predictive analysis to assess the correlation between COL6A2 expression and renal cancer patients’ sensitivity to various treatments. Higher COL6A2 expression was associated with increased sensitivity to targeted drugs like sunitinib, pazopanib, and cabozantinib. TIDE analysis revealed that patients with high COL6A2 expression had higher TIDE scores and a significantly lower proportion of responders to immunotherapy compared to the low-expression group, suggesting poorer responses to immune checkpoint inhibitors (e.g., PD-1/PD-L1 inhibitors). TIDE analysis further showed that the high COL6A2 expression group had lower MSI scores and CD274 levels at immune checkpoints, while Dysfunction and Exclusion scores, as well as MDSC and CAF proportions, were significantly higher compared to the low-expression group. This suggests that COL6A2 plays a significant role in immune evasion in renal cancer, consistent with preliminary analysis results. Experimental validation demonstrated that COL6A2 knockdown significantly increased the IC50 values of tumor cells, while no significant change in tumor size was observed following intratumoral sunitinib injection. However, in the subcutaneous tumor-bearing model, tumor size was significantly reduced in the COL6A2 knockdown group compared to the non-knockdown group. In the anti-PD-1 treatment model, mice with COL6A2 knockdown exhibited significantly enhanced sensitivity to anti-PD-1 treatment compared to the normal group. These findings suggest that COL6A2 not only serves as a prognostic marker but may also be a potential therapeutic target for guiding clinical treatment in ccRCC patients. Although this study outlines the potential role of COL6A2 in ccRCC, further experimental validation is required. Limitations of this study include the unclear molecular mechanisms underlying COL6A2’s interaction with the PI3K-AKT and TGF-β pathways, as well as its potential synergy with other ECM proteins. Future research should focus on COL6A2’s role in the tumor immune microenvironment and its influence on immune evasion and EMT. The small sample sizes in existing studies limit generalizability, highlighting the need for large-scale, multicenter studies. While the association between COL6A2 and tumor prognosis is preliminarily established, its clinical application as a biomarker and its potential in targeted therapies for renal cancer require further validation through extensive clinical trials. Conclusion Overall, this study demonstrates a strong association between high COL6A2 expression in ccRCC and tumor progression, the tumor immune microenvironment, and clinical prognosis. COL6A2 serves as a potential prognostic marker and offers new insights into the selection of targeted and immunotherapies for renal cancer. Future research should further investigate the biological role of COL6A2 in renal cancer and its clinical application, aiming to identify new targets for early diagnosis and personalized treatment. Electronic supplementary material Below is the link to the electronic supplementary material. [148]Supplementary Material 1^ (2.9MB, docx) Acknowledgements