Abstract Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by limited treatment options and poor prognosis. Recent evidence highlights the crucial role of cancer-associated fibroblasts (CAFs) in TNBC progression, yet their molecular characteristics remain incompletely understood. In this study, we performed a comprehensive analysis combining bioinformatics approaches with experimental validation to investigate CAF-related genes in TNBC. Using weighted gene co-expression network analysis (WGCNA) of TNBC samples from TCGA and METABRIC datasets, we identified 185 CAF-related genes significantly associated with extracellular matrix organization and TGF-β signaling pathways. Through rigorous statistical modeling, we developed a 3-gene prognostic signature (CERCAM, JAM3, PLAU) that effectively stratified TNBC patients into high- and low-risk groups with distinct survival outcomes. Importantly, we validated the functional role of PLAU, one of the signature genes, through in vitro and in vivo experiments. Results showed that CAF-derived PLAU played key role in the malignant behaviors of TNBC. Our findings provide new insights into CAF-mediated TNBC progression and suggest potential stromal targets for therapeutic intervention. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-025-03867-y. Keywords: TNBC, Cancer-associated fibroblasts, Prognostic signature, PLAU, Tumor microenvironment Introduction Triple-negative breast cancer (TNBC) represents the most aggressive subtype of breast cancer, characterized by the absence of estrogen receptor, progesterone receptor, and HER2 expression [[42]1]. Despite accounting for only 15–20% of all breast cancer cases, TNBC contributes disproportionately to breast cancer mortality due to its intrinsic biological aggressiveness and limited treatment options [[43]2]. The lack of targeted therapies has made chemotherapy the mainstay of treatment, yet many patients develop resistance, highlighting the urgent need for novel therapeutic strategies [[44]3, [45]4]. Cancer-associated fibroblast (CAF) is a major component in the tumor microenvironment in established solid tumors [[46]5]. Recent advancements in molecular biology have underscored the pivotal role of cancer-associated fibroblasts (CAFs) within the tumor microenvironment [[47]6]. TNBC often exhibited massive fibrosis with abundant infiltration of CAFs, which protect tumor from antitumor lymphocyte infiltration and contribute to immunosuppression, progression and treatment resistance [[48]7]. In TNBC, CAF secreted factors were proved to promote the cancer cell progression through inhibiting the tumor suppressor NR4A1 [[49]8]. C. Gaggioli reported that CAFs were acting as the leading cell in the migration and invasion process of carcinoma cells [[50]9]. In addition, lung-resident fibroblasts modify cancer cells metabolism in the lung and promoting angiogenesis in TNBC [[51]10]. Due to the tumor-promoting properties, a number of therapeutic strategies are designed to target the TNBC CAFs [[52]11]. However, there is still a long way to go before it can be implemented in clinical practice. Understanding the intricate interplay between CAFs and tumor cells has thus emerged as a critical area of research, promising new insights into prognostic biomarkers and therapeutic strategies. PLAU, a serine protease, is known for its role in degrading the extracellular matrix (ECM), thereby facilitating tumor invasion and metastasis. The expression of PLAU in stromal cells, particularly CAFs, has been associated with aggressive tumor phenotypes and poor prognosis in various cancers. For instance, in esophageal squamous cell carcinoma (ESCC), PLAU secreted by tumor cells has been shown to promote the conversion of fibroblasts into inflammatory CAFs, which in turn enhance tumor progression through the uPAR/Akt/NF-κB/IL8 signaling pathway [[53]12]. In pancreatic ductal adenocarcinoma (PDAC), PLAU expression has been linked to the activation of oncogenic signaling pathways and the modulation of stromal-cancer cell interactions [[54]13]. Nevertheless, the specific contribution of CAF-derived PLAU to TNBC progression has not been systematically investigated. This study employs Weighted Gene Co-expression Network Analysis (WGCNA) to delineate a distinct CAF signature in TNBC, aiming to predict patient outcomes and therapeutic responses more accurately. Furthermore, the identification of PLAU as a novel prognostic biomarker highlights its potential therapeutic implications in breast cancer management. Materials and methods Data acquisition The transcriptome, mutation, and clinical data of breast cancer samples were downloaded from the TCGA database ([55]https://gdc-portal.nci.nih.gov/). A total of 116 samples with ER negative, PR negative and HER2 negative were identified as triple negative breast cancer patients (TCGA-TNBC) for our analysis. We also downloaded the transcriptome and clinical data of METABRIC dataset from cBioPortal ([56]https://www.cbioportal.org/) and enrolled 320 triple negative breast cancer patients (METABRIC-TNBC) as the validation cohort. The single-cell RNA-seq data of fresh TNBC samples ([57]GSE118389 and [58]GSE176078) were downloaded from GEO database ([59]https://www.ncbi.nlm.nih.gov/geo/). Calculating the abundance of CAFs from bulk tumor gene expression data After downloading the bulk RNA expression data from two different database, we calculate the CAFs abundance of each TNBC samples by R package “EPIC” and “MCP-counter”. The Estimate the Proportion of Immune and Cancer cells (EPIC) algorithm was employed to assess the proportion of immune cells, fibroblasts and cancer cells under different expression conditions of specific genes [[60]14]. The Microenvironment Cell Population counter (MCP-counter) algorithm was processed to calculate the absolute quantification of immune cells and fibroblasts in each sample [[61]15]. The R package “ESTIMATE” was used to estimate the stromal score, immune score and tumor purity in tumor microenvironments [[62]16]. Performing weighted gene Co-expression network analysis (WGCNA) and screening for hub genes WGCNA was performed on both the TCGA-TNBC and METABRIC-TNBC cohorts with the R package “WGCNA” [[63]17]. Genes with similar expression patterns were clustered together into one module and marked with same color. Then the Pearson’s correlations between module eigengenes and EPIC-quantified CAF infiltrations as well as the stromal score were evaluated. Genes in the most correlated module of two cohorts were collected, and the common genes were selected as CAF related genes. The pathway enrichment analysis was performed using R package “clusterProfiler”. Prognostic model construction and validation We first performed univariate Cox regression analysis to find genes significantly related to survival. Then we utilized least absolute shrinkage and selection operator (LASSO) regression analysis to further screen prognostic-related genes. Finally, we performed stepwise multivariate COX regression analysis to find optimal prognostic-related CAF genes to constructed CAF signature score. The R package “Survival” was used to plot the survival curves by Kaplan-Meier methods. Screening for CAF markers and conducting correlation analysis To validate the accuracy of CAF risk model, we compared the CAF abundances and stromal score between high- and low- CAF risk score groups. Through literature consulting, we collected 23 CAF specific marker genes. The correlations between the risk model and the expression of CAF specific marker genes were also analyzed. Immune infiltration analysis The relative proportions of immune cell subsets in the TCGA-TNBC cohort were quantified using CIBERSORT algorithm of R package [[64]18]. Differential immune infiltration in the tumor microenvironment between low- and high-risk groups was assessed using the limma R package. Spearman’s correlation analysis was performed to evaluate associations between immune cell infiltration levels and the expression of CAF signature genes. Prediction of immunotherapy and chemotherapy response Tumor Immune Dysfunction and Exclusion (TIDE) analysis ([65]https://tide.dfci.harvard.edu/) were utilized to evaluate the immunotherapy response. Drug sensitivity analysis was conducted by pRRophetic package. Data visualization was performed using the ggplot2 package for graphical representation. Validating the CAF signature genes in Cancer Cell Line Encyclopedia (CCLE) and Human Protein Atlas (HPA) Databases The mRNA expression of CAF signature genes in fibroblasts and breast cancer cell lines were downloaded from the CCLE database and compared by Wilcoxon tests [[66]19]. The protein expression patten of the CAF signature genes in breast cancer tissues were obtained from the HPA database ([67]https://www.proteinatlas.org/). Single-cell RNA-seq data analysis We processed the single-cell RNA-seq data by R package “Seurat”. The clusters were annotated by R package “singleR”. Conditioned medium preparation PLAU was selected for further investigation based on its high coefficient and weight in the multivariable prognostic model, which suggested its strong correlation with TNBC progression. A comprehensive review of the literature also revealed that PLAU is a well-established factor in tumor progression and metastasis. CAFs was generated by stimulating MRC5 cells with recombinant human transforming growth factor β1 for 24 h. Lentivirus knocking down PLAU was purchased from Genechem (Shanghai, China). CAFs, either with PLAU knockdown or as controls, were cultured in DMEM supplemented with 10% FBS and antibiotics until they reached approximately 80% confluence. The culture medium was then collected and centrifuged at 1200 g for 10 min. The freshly obtained conditioned medium (CM) was subsequently used to culture MDA-MB-231 cells. Western blot analysis Protein lysates were resolved via SDS-polyacrylamide gel electrophoresis (SDS-PAGE) and subsequently electrotransferred onto PVDF membranes. The membranes were then blocked with 5% skim milk and probed with the following primary antibodies: anti-GAPDH (1:1000, #2118, CST) and anti-PLAU (1:1000, #15800, CST). Following an overnight incubation at 4 °C, unbound antibodies were removed by washing, and the membranes were exposed to HRP-conjugated secondary antibodies (1:5000, Proteintech). Immunoreactive bands were visualized using an enhanced chemiluminescence (ECL) substrate and imaged with a G: BOX Chemi X system (Syngene, UK). CCK8 assay Cell proliferation was assessed using the CCK-8 assay. Briefly, cells were cultured in 96-well plates with CM from control or PLAU-knockdown CAFs. At the predetermined time, the CM was removed and 100 µL of medium containing 10% CCK-8 reagent (MedChem Express, USA) was added to each well, followed by incubation at 37 °C for 1–2 h. Optical density (OD) was measured at 450 nm using a microplate reader (BioTek, Winooski, VT, USA). EdU cell proliferation assay Cells were seeded in 12-well plates and allowed to adhere overnight. Then the culture median was replaced by CM from control or PLAU knockdown CAFs. After 48 h, cells were treated with the designated reagents following the manufacturer’s protocol (EdU kit, Meilunbio, China). Proliferating cells (EdU-positive) were visualized and quantified using a fluorescence microscope (Leica). Wound healing assay Cells were seeded in 12-well plates and adhere overnight. A linear scratch wound was created in the monolayer using a 200 µL pipette tip, followed by incubation in CM from control or PLAU knockdown CAFs. Wound closure was monitored at baseline (0 h) and 48 h post-scratching, with phase-contrast images captured using an optical microscope (Nikon) to quantify migration rates. Transwell migration and invasion assays Cell migration and invasion were assessed using 24-well Transwell chambers (8-µm pores, NEST Biotechnology). For the invasion assay, upper chambers were pre-coated with 50 µL diluted Matrigel (1:8, BD Biosciences) and incubated overnight at 37 °C. 50,000 cells suspended in 200 µL serum-free medium were seeded into the upper chamber, while the lower chamber contained CM from control or PLAU knockdown CAFs. After 24–48 h of incubation, non-migratory cells on the upper membrane surface were gently removed with cotton swabs. Migrated/invaded cells on the lower membrane were fixed in 4% paraformaldehyde, stained with 0.1% crystal violet, and quantified under a microscope. In vivo tumor xenograft study The female BALB/c nude mice (six weeks old) were used to construct xenograft model by subcutaneously injecting the mixture of NC or sh-PLAU CAFs cells with MDA-MB-231 into the right axilla of each mouse (n = 5 per group). The tumor volumes were measured every three days over a 22-day period with the formula: tumor volume (mm3) = 1/2 (length × width^2). Immunohistochemistry Tissue samples from experimental animals were first fixed in 10% neutral buffered formalin for 24 hours and then processed through a routine paraffin embedding procedure. Tissue sections, 4 µm in thickness, were cut using a microtome and placed on glass slides. Prior to antigen retrieval, all sections were washed with distilled water and dehydrated through a graded ethanol series. Antigen retrieval was performed by heating the sections in citrate buffer (pH 6.0) at 95–100°C for 30 minutes, followed by cooling to room temperature. Next, the sections were incubated with 5% hydrogen peroxide solution to block endogenous peroxidase activity for 10 minutes. The sections were then incubated with blocking solution containing 5% normal goat serum for 1 hour at room temperature to reduce non-specific binding. The primary antibody was applied to the sections and incubated overnight at 4°C. On the following day, a biotinylated secondary antibody was applied to the sections and incubated for 1 hour at room temperature. Following this, the ABC method was used to amplify the secondary antibody signal. Finally, the sections were stained with 3,3’-diaminobenzidine (DAB) and counterstained with hematoxylin. Afterward, the sections were dehydrated through a graded ethanol series, cleared in xylene, and mounted with a coverslip for examination. The primary antibodies were listed as follows: PLAU (Proteintech, 17968-1-AP), Ki-67 (Abcam, ab15580), α-SMA(Servicrbio, [68]GB111364), MMP2(Servicrbio, GB11130). Statistical analysis Statistical analyses were performed using R software (version 4.1.0) and GraphPad Prism 8.0. Continuous variables between two groups were compared using Student’s t-test for normally distributed data or Wilcoxon rank-sum test for non-parametric data. Survival analysis was conducted using Kaplan-Meier curves with log-rank tests for group comparisons. Spearman’s rank correlation analysis was employed to evaluate bivariate associations. A two-tailed p-value < 0.05 was considered statistically significant for all analyses. Results Higher CAF abundance and stromal scores indicate poorer prognosis in TNBC We used EPIC and MCP-counter to calculate CAF abundance in TNBC patients. The ESTIMATE algorithm was used to calculated the stromal score of TNBC samples. Kaplan-Meier revealed that higher CAF abundance and stromal scores were associated with worse overall survival of TNBC patients in both TCGA and METABRIC cohorts (Fig. [69]1A-F). These results demonstrated the role of CAF in the malignancy of TNBC and highlighted the importance of further exploration of CAF related genes for TNBC. Fig. 1. [70]Fig. 1 [71]Open in a new tab Higher CAF abundance and stromal scores indicate poorer prognosis in TNBC. (A) Kaplan-Meier curves of TCGA-TNBC patients stratified by CAF abundance estimated by EPIC algorithm. (B) Kaplan-Meier curves of TCGA-TNBC patients stratified by CAF proportion estimated by MCPcounter algorithm. (C) Kaplan-Meier curves of TCGA-TNBC patients stratified by StromalScore. (D) Kaplan-Meier curves of METABRIC-TNBC patients stratified by CAF abundance estimated by EPIC algorithm. (E) Kaplan-Meier curves of METABRIC-TNBC patients stratified by CAF proportion estimated by MCPcounter algorithm. (F) Kaplan-Meier curves of METABRIC-TNBC patients stratified by StromalScore WGCNA analysis of TCGA-TNBC and METABRIC-TNBC To obtain key genes related to CAF, we firstly performed WGCNA on both TCGA-TNBC and METABRIC-TNBC cohort. For TCGA-TNBC, the soft threshold power is 11 and scare-free R2 is 0.97 (Fig. [72]2A, B). A total of 8 co-expression modules were clustered, with the yellow module showing the strongest positive correlation with CAF abundance (Cor = 0.62, P = 2e-13) and stromal score (Cor = 0.74, P = 4e-21) (Fig. [73]2E). For METABRIC-TNBC, the soft threshold power is 5 and scare-free R2 is 0.97 (Fig. [74]2C, D). A total of 14 co-expression models were clustered, with the black module showing the strongest positive correlation with CAF abundance (Cor = 0.91, P = 2e-126) and stromal score (Cor = 0.82, P = 5e-79) (Fig. [75]2F). Finally, a total of 353 genes in the yellow module of TCGA-TNBC and 422 genes in the black module of METABRIC-TNBC were screened out as hub genes which were highly associated with CAF and stromal score. Fig. 2. [76]Fig. 2 [77]Open in a new tab WGCNA in the TCGA-TNBC cohort and METABRIC-TCGA cohort. (A) Scale independence and mean connectivity in the TCGA-TNBC cohort. (B) Gene dendrogram and modules before merging in the TCGA-TNBC cohort. (C) Scale independence and mean connectivity in the METABRIC-TNBC cohort. (D) Gene dendrogram and modules before merging in the METABRIC-TNBC cohort. (E) Pearson correlation analysis of merged modules and CAF abundance in the TCGA-TNBC cohort. (F) Pearson correlation analysis of merged modules and CAF abundance in the METABRIC-TNBC cohort Functional analyses of CAF-related genes A total of 422 and 353 genes were included in the TCGA-TNBC yellow and METABRIC-TNBC black modules, respectively. These genes were considered to be highly associated with CAF related phenotype. By intersecting two hub gene sets, 185 common genes were found out and visualized via Venn diagram (Fig. [78]3A, Supplementary Table [79]1). To explore the function of the common hub genes, GO and KEGG analyses were carried out. As showed in Fig. [80]3B, these genes were mainly associated with “extracellular matrix organization”, “connective tissue development”, “response to transforming growth factor beta”, “collagen fibril organization” of the biological process (BP) category. In the cellular component (CC) term, “collagen-containing extracellular matrix”, “collagen trimer” and “banded collagen fibril” was significantly enriched. The molecular function (MF) category mainly included “extracellular matrix structural constituent”, “growth factor binding” and “extracellular matrix binding”. KEGG pathway enrichment analysis showed that these genes were mainly enriched in “ECM-receptor interaction”, “TGF-beta signaling pathway” and “focal adhesion”(Fig. [81]3C). These results suggested that these genes are representative genes and play key roles in the CAF development, collagen synthesis and extracellular matrix formation in TNBC. We can conclude that the 185 genes identified by WGCNA were CAF-specific genes in TNBC. Fig. 3. [82]Fig. 3 [83]Open in a new tab Functional analysis of CAF related genes. (A) Venn diagram showing the overlapped CAF-related genes in the TCGA-TNBC and METABRIC-TNBC cohort. (B) GO enrichment analysis of the common CAF-related genes. (C) KEGG enrichment analysis of the common CAF-related genes Screening out prognosis-related CAF-specific genes and constructing prognostic gene signature Our next step is to identify more representative genes that are closely associated with the prognosis of TNBC. Firstly, univariate Cox regression analysis was conducted on the 185 CAF-specific genes, and 37 OS-related genes with p < 0.05 were selected (Fig. [84]4A). Among these prognosis-related genes, COL5A1 showed the highest mutation rate of 4% in TNBC patients. CDH11, SERPINE1 and COL12A1 had a mutation rate of 3%. POSTN, VCAN, COL3A1, COL5A2, COL1A2 and COL11A1 had a mutation rate of 2%. RAB31, DPYSL3, NUAK1, PRSS2, SRPX2, SPOCK1, COL1A1 and MFAP5 had a mutation rate of 1% (Fig. [85]4B). Fig. 4. [86]Fig. 4 [87]Open in a new tab Screening out prognosis-related CAF-specific genes and constructing prognostic gene signature. (A) Forest plot showing effect of 37 prognostic CAF-related genes on survival. (B) Somatic mutation landscape of 37 prognostic genes. (C and D) LASSO regression analysis. (E) Survival difference between the high- and low-CAF score groups in TCGA-TNBC cohort. (F) Survival difference between the high- and low-CAF score groups in METABRIC-TNBC cohort Secondly, LASSO regression analysis and stepwise multivariate regression analysis was conducted to further narrow the scope, and three crucial genes were finally found out to construct a CAF-related signature score (CAF score) (Fig. [88]4C and D). The CAF risk score = expression of CERCAM*0.011 + expression of JAM3*0.424 + expression of PLAU*0.452. Patients were divided into high-risk CAF score group and low risk CAF score group with the median score as the cutoff value. As displayed in Fig. [89]4E and F, TNBC patients in the high-risk CAF score group showed significantly worse overall survival than those in the low-risk CAF score group both in the TCGA cohort (HR = 23.067, 95%CI: 2.834 ~ 187.727, log-rank p < 0.001) and METABRIC cohort (HR = 2.2, 95%CI: 1.601 ~ 3.024, log-rank p < 0.001). We validated the CAF-score in independent cohorts from the GEO database ([90]GSE25066). The model retained predictive power (HR = 2.19, 95%CI:1.107 ~ 4.331, log-rank p = 0.021) (Supplementary Fig. [91]1). CAF score and signature genes were crucial indicators of CAF infiltrations In order to verify the accuracy of the CAF risk model, we compared between two groups the stromal score as well as CAF abundances predicted by ESTIMATE and other three methods: EPIC, MCP-counter and TIDE. As a result, patients in high- CAF score group showed significantly higher stromal score and CAF score than those in low- CAF score group (Fig. [92]5A, B, C and D). Patients with high- CAF scores showed obviously higher expression of documented CAF markers such as ACTA2, FN, COL1A1, COL1A2 and MMP2 (Fig. [93]5E). Additionally, the CAF score and expression of three signature genes exhibited positive correlation with the documented CAF markers (Fig. [94]5F). GSEA revealed that pathways closely related to extracellular formation were significantly enriched in high CAF score group, such as “ECM receptor interaction” and “focal adhesion and regulation” (Fig. [95]5G). While genes in low- CAF score group were significantly enriched in “oxidative phosphorylation”, “primary immunodeficiency” and “ribosome” (Fig. [96]5H). Furthermore, ssGSEA results also showed that the CAF score was positively correlated with pathways such as “ECM receptor interaction”, “TGFbeta signaling pathway” and “focal adhesion” (Fig. [97]5I, J and K). Fig. 5. [98]Fig. 5 [99]Open in a new tab The CAF signature could predict the CAF infiltration in TNBC. (A) Differences in stromal score evaluated by ESTIMATE algorithm between high- and low- CAF score groups. (B) Difference in CAF infiltration evaluated by EPIC algorithm between high- and low- CAF score groups. (C) Difference in CAF infiltration evaluated by MCPcounter algorithm between high- and low- CAF score groups. (D) Difference in CAF infiltration evaluated by TIDE algorithm between high- and low- CAF score groups. (E) Heatmaps of expression of CAF markers in high- and low- risk groups. (F) Correlation analysis between the signature genes and CAF markers. (G) GSEA plot of high-risk group. (H) GSEA plot of low-risk group. (I) Correlation analysis between risk score and ECM receptor interaction (left), TGF-beta signaling pathway (middle) and focal adhesion (right) CAF score and signature genes were potential predictive biomarker for immunotherapy and chemotherapy Since tumor-associated fibroblasts are associated with the efficacy of immunotherapy and chemotherapy, we aim to investigate whether the CAF score is correlated with immune infiltration and whether it can predict the efficacy of both immunotherapy and chemotherapy. As depicted in Fig. [100]6A, high-CAF score group exhibits a higher infiltration of immunosuppressive M2 macrophages and a lower infiltration of immune-promoting CD8 + T cells. Moreover, PLAU expression was positively correlated with M2 macrophages and negatively correlated with CD8 + T cells (Fig. [101]6B). The TIDE algorithm predicted that fewer patients in the high-CAF score group would benefit from immunotherapy compared to those in the low-CAF score group (Fig. [102]6C). Drug sensitivity analysis showed that patients in high-CAF score group were more sensitive to oxaliplatin, fludarabine, lapatinib and gefitinib (Fig. [103]6D-G). These findings suggest that PLAU, secreted by CAFs, may contribute to the immune-suppressive environment in TNBC, which could promote tumor aggressiveness and resistance to immune therapies. Fig. 6. [104]Fig. 6 [105]Open in a new tab CAF score and signature genes were potential predictive biomarker for immunotherapy and chemotherapy. (A) Differences in the immune cells infiltrated in the tumor microenvironment between the low- and high-CAF score groups. (B) Correlations between infiltrated immune cells and the expression of CAF signature genes. (C) The proportion of patients with varying immunotherapy responses in the two risk groups. (D) Drug sensitively to oxaliplatin between the high- and low-CAF score group. (E) Drug sensitively to fludarabine between the high- and low-CAF score group. (F) Drug sensitively to lapatinib between the high- and low-CAF score group. (G) Drug sensitively to gefitinib between the high- and low-CAF score group Two groups exhibited different mutation spectrum We wondered whether there were correlations between the CAF score and somatic mutation in TNBC. Firstly, the mutation profile of all breast cancer patients and TNBC patients were depicted separately. Among all breast cancer patients in TCGA database, PIK3CA showed the highest mutation rate of 34%, followed by TP53 (34%), TTN (17%) and CDH1 (13%) (Fig. [106]7A). While in TNBC, the most frequently mutated gene were TP53 (83%), followed by TTN (19%), MUC16 (12%) and SYNE1 (13%) (Fig. [107]7B). Of note, two CAF score groups exhibited different mutation spectrum. Mutations of PIK3CA, TP53, TTN, MAP3K1, HMCN1, FLG and SPTA1 were higher in high-CAF score group than in low-CAF score group, while the mutation frequency of CDH1, MUC16, KMT2C and PTEN was the same between two groups (Fig. [108]7C and D). Fig. 7. [109]Fig. 7 [110]Open in a new tab The mutation profile differed in two risk groups. (A) The top 10 mutated genes in all BRCA samples in TCGA. (B) The top 10 mutated genes in TNBC samples in TCGA. (C) The mutation profile of samples in low-risk group. (D) The mutation profile of samples in high-risk group Analyze CAF signature genes in single-cell RNA-sequencing data In order to investigate the expression differences of these three genes among different cell types in tumor tissues, we downloaded single-cell RNA-sequencing data of TNBC from GEO database. After quality control and normalization of the dataset [111]GSE176078, the RNA-seq data of 39,375 cells from ten fresh TNBC tumors were obtained (Fig. [112]8A). Using dimensionality reduction visualization analysis, 39,375 cells were divided into 22 clusters (Fig. [113]8B). The R package “SingleR” was used to annotate these 22 clusters (cluster 3, 7, 14, 15, 18, 19 and 22 were Epithelial cells; cluster 0, 2, 10 and 13 were T cells; cluster 9 were Endothelial cells; cluster 1 and 4 were Monocyte; cluster 11 were Fibroblasts; cluster 12 were Smooth muscle cells, Fig. [114]8C). Among the three signature genes, CERCAM was mainly expressed in fibroblast. JAM3 was detected in smooth muscle cells, fibroblast and endothelial cells. While PLAU was detected in fibroblasts, monocyte and endothelial cells (Fig. [115]8D). Fig. 8. [116]Fig. 8 [117]Open in a new tab Single-cell RNA sequence analysis reveals fibroblasts as the major source of CERCAM, JAM3, and PLAU. (A) The tsne map of 10 mixed samples from [118]GSE176978. (B) The findCluster function was used to obtain clusters. (C) 23 clusters annotated to nine-cell types by singlR. (D) The expression of three signature genes in different cell types. (E) The tsne map of 6 mixed samples from [119]GSE118389. (F) The findCluster function was used to obtain clusters. (G) 11 clusters annotated to six-cell types by singlR. (H) The expression of three signature genes in different cell types We performed the same analysis on the dataset [120]GSE118389 and obtained a similar result. The RNA-seq data of 1534 cells from six fresh TNBC tumors were obtained and divided into 11 clusters (Fig. [121]8E and F). The R package “SingleR” was used to annotate these 11 clusters (cluster 0, 1, 2,4,8,9 were Epithelial cells; cluster 3 were T cells; cluster 5 were Endothelial cells; cluster 6 were Monocyte; cluster 7 were Fibroblasts; cluster 10 were Smooth muscle cells, Fig. [122]8G). The three signature genes, i.e., CERCAM, JAM3, and PLAU, all exhibited high expression in fibroblasts. JAM3 was also expressed in endothelial cells, while PLAU was detected in monocyte and endothelial cells (Fig. [123]8H). PLAU plays key role in the malignant behaviors of TNBC To further elucidate the expression characteristics of the CAF signature genes, we download the gene expression data of fibroblast and breast cancer cell lines from Cancer Cell Line Encyclopedia database. Results showed that the mRNA expression of CERCAM, KAM3 and PLAU were all significantly higher in fibroblast cell lines than in breast cancer cell lines (Fig. [124]9A). The IHC images of the CAF signature genes were downloaded from the HPA database. As presented in Fig. [125]9B, CERCAM is hardly expressed in normal breast tissue but found positive in cytoplasm and membrane of breast tumor cells. JAM3 is lowly expressed in glandular cells of normal mammary gland and moderately expressed in breast cancer tissues. In normal mammary gland, PLAU was medium expressed in adipocytes and glandular cells, not detected in myoepithelial cells. In contrast, PLAU was highly expressed in entire breast cancer nest in breast cancer tissues. Fig. 9. [126]Fig. 9 [127]Open in a new tab PLAU knockdown in CAFs suppressed tumor cell proliferation, migration, and invasion in vitro. (A) The expression levels of CERCAM, JAM3, and PLAU in fibroblast and breast cancer cell lines in Cancer Cell Line Encyclopedia database. (B) The IHC images of CERCAM, JAM3, and PLAU in the HPA database. (C) Efficacy of shRNA in suppressing expression of PLAU in CAFs. (D) OD values of MDA-MB-231 cells cultured in CM from control or PLAU knockdown CAFs in CCK8 assay. (E) Proliferative activity of MDA-MB-231 cells cultured in CM from control or PLAU knockdown CAFs in Edu assay (40×). (F and H) Wound healing assay of MDA-MB-231 cells cultured in CM from control or PLAU knockdown CAFs (40×). (G and I) Migration and invasion abilities of MDA-MB-231 cells cultured in CM from control or PLAU knockdown CAFs in transwell assays (200×). (*, p<0.05; **, p<0.01; ***, p<0.001) In our multivariable prognostic model, PLAU was identified as a key prognostic factor, with the highest coefficient and weight, underscoring its critical role in TNBC progression. Furthermore, PLAU has been reported to be involved in the development, metastasis, and prognosis of various cancers. Thereby, we selected PLAU for in-depth study. We wonder whether regulating PLAU in fibroblasts affects the proliferation and invasion ability of triple-negative breast cancer cells. Firstly, we knockdown PLAU by lentivirus in the fibroblast (Fig. [128]9C) and turn the fibroblast cells into CAFs by stimulated it with rhTGFβ. Then We stimulated the MDA-MB-231 cells with conditioned medium (CM) from control and PLAU knockdown CAFs. Results showed that the proliferation rate of MDA-MB-231 cells decreased in CM from PLAU knockdown CAFs (Fig. [129]9D, E). In wound healing assays, MDA-MB-231 cells migrated more slowly in CM from PLAU knockdown CAFs than in CM from control CAFs (Fig. [130]9F, H). What’s more, MAD-MB-231 cells exhibited weaker migration and invasive abilities in CM from PLAU knockdown CAFs in transwell assays (Fig. [131]9G, I). PLAU knockdown in CAFs suppressed tumor growth of TNBC in vivo To further investigate the role of PLAU in vivo, we co-injected MDA-MB-231 cells and CAFs at a 1:1 ratio into the subcutaneous space of nude mice. Tumor growth was significantly slower in the PLAU knockdown group compared to the control group (Fig. [132]10A, B). Immunohistochemical (IHC) staining confirmed successful downregulation of PLAU expression in tumor tissues from the PLAU knockdown group (Fig. [133]10C). Additionally, the expression of Ki-67, a marker of cell proliferation, was markedly reduced in this group (Fig. [134]10C). Importantly, we also observed a significant decrease in the expression of key extracellular matrix remodeling markers, such as αSMA and MMP2, in the PLAU knockdown tumors (Fig. [135]10C). Spearman’s correlation analysis further revealed a significant positive correlation between PLAU expression and the levels of MMP2, ACTA2, ITGAV, and CTSB—genes well-established for their roles in ECM remodeling (Fig. [136]10D). Collectively, these findings suggest that PLAU contributes to the progression of TNBC, at least in part, by regulating the ECM remodeling pathway (Fig. [137]10E). Fig. 10. [138]Fig. 10 [139]Open in a new tab PLAU knockdown in CAFs suppressed tumor growth of TNBC in vivo. (A) Gross appearance of tumor xenografts in control and sh-PLAU groups. n = 5. (B) Tumor growth curve of two groups. (*, p<0.05). (C) IHC staining of PLAU, Ki-67,α-SMA and MMP2 in the tumor tissues. (D) Scatter plot showing correlation between PLAU with the expression of ACTA2, MMP2, ITGAV and CTSB in TCGA-TNBC dataset by Spearman’s correlation tests. (E) The mechanism diagram of the study. Discussion TNBC is an aggressive tumor and the prognosis is difficult to be improved by existing treatment modalities [[140]20]. The mechanism of TNBC progression is still unclear. Recent studies have identified various markers and molecular pathways associated with CAFs that influence the tumorigenic process. Recent advancements in CAF heterogeneity have highlighted the diverse subpopulations of CAFs, each contributing differently to tumor progression [[141]21]. Furthermore, recent studies have developed risk models incorporating CAF signatures to predict patient outcomes, with significant implications for personalized treatment in breast cancer [[142]22, [143]23]. In the current study, we found that higher CAF abundance in the tumor microenvironment was associated with worse prognosis in TNBC. We establish a robust CAF-derived prognostic signature in TNBC through integrative multi-omics analysis and functional validation, identifying PLAU as a pivotal mediator of CAF-driven tumor progression. Our findings not only resolve critical gaps in understanding stromal-tumor crosstalk but also provide actionable insights for precision oncology in this aggressive breast cancer subtype. The robust association between high CAF abundance and poor survival (HR = 23.07, TCGA cohort) aligns with emerging evidence that CAFs sculpt immunosuppressive niches via extracellular matrix (ECM) remodeling and cytokine secretion [[144]6]. The WGCNA-identified genes, particularly those enriched in ECM organization (e.g., COL5A1, COL12A1) and TGF-β signaling, corroborate recent work demonstrating that CAF-derived collagens activate integrin-β1/FAK pathways to drive TNBC metastasis [[145]24]. Notably, the inclusion of PLAU in our prognostic model resolves a long-standing controversy regarding the role of urokinase-type plasminogen activator (uPA) in breast cancer. While earlier studies attributed uPA’s pro-metastatic effects primarily to tumor cell autonomy [[146]25], our single-cell RNA-seq data and functional assays definitively implicate CAF-secreted PLAU as a key driver of TNBC progression [[147]26]. The prognostic power of our CAF signature surpasses previous stromal models [[148]16], likely due to its unique integration of junctional proteins (CERCAM, JAM3) that mediate direct CAF-tumor interactions—a dimension overlooked in prior transcriptomic analyses [[149]27]. This innovation addresses a critical limitation of bulk RNA-seq deconvolution methods, which often fail to capture spatial biology nuances [[150]28]. Furthermore, the divergent mutation landscapes between high- and low-CAF score groups suggest that CAF-induced stromal pressure may accelerate genomic instability, a hypothesis supported by recent reports linking CAF-secreted ROS to TP53 mutagenesis [[151]29]. Clinically, our model’s consistent performance across ethnically diverse cohorts (TCGA and METABRIC) provides a urgently needed tool for risk stratification in TNBC, a disease marked by heterogeneous treatment responses [[152]20]. The strong association between high-CAF scores and ECM receptor interaction pathways offers a mechanistic explanation for the failure of immune checkpoint inhibitors in fibrotic TNBC subtypes [[153]30], advocating for combined stromal-immune targeting strategies. For instance, PLAU inhibition could synergize with anti-PD1 therapies by normalizing the fibrotic microenvironment. Given the growing body of evidence linking CAFs to immune suppression in the tumor microenvironment, future research could investigate whether PLAU expression in CAFs contributes to the activation of inflammatory pathways or impedes immune cell infiltration, which would further strengthen our understanding of how PLAU promotes tumor progression in TNBC. Targeting PLAU or its downstream signaling pathways could thus offer a promising therapeutic strategy to disrupt the immunosuppressive microenvironment and enhance anti-tumor immunity. Despite these advances, limitations warrant consideration. First, our xenograft model, while informative, does not fully recapitulate human CAF heterogeneity. Recent advances in patient-derived CAF-organoid co-cultures could overcome this constraint [[154]31]. Second, spatial transcriptomics is needed to validate the PLAU expression gradient observed in single-cell data. Lastly, prospective validation in clinical trials is essential to assess the CAF signature’s utility in guiding neoadjuvant therapy selection [[155]32]. In conclusion, this study establishes PLAU as a linchpin of CAF-driven TNBC progression and provides a translatable prognostic framework. By contextualizing stromal biology within the genomic and clinical landscape of TNBC, our work paves the way for mechanism-informed therapeutic strategies targeting the tumor microenvironment. Electronic supplementary material Below is the link to the electronic supplementary material. [156]12935_2025_3867_MOESM1_ESM.jpeg^ (151.4KB, jpeg) Supplementary Material 1: Supplementary Figure 1. Survival difference between the high- and low- CAF score groups in GSE25066-TNBC dataset from NCBI GEO database. [157]12935_2025_3867_MOESM2_ESM.txt^ (1.4KB, txt) Supplementary Material 2: Supplementary table 1. The list of 185 CAF-related genes. Author contributions Xianglin Yuan and Binghe Xu performed study concept and design; Jun Zou performed the bioinformatics analysis and writing; Wan Qin conducted the experiments; Zhang and Yu Li performed review and revision of the paper; Baowen Yuan, Yuanyi Wang, Yalong Qi and Qian Wang provided technical and material support. All authors read and approved the final manuscript. Funding This work was supported by the National Natural Science Foundation of China for Young Scholars (Grant No.82203972). Data availability No datasets were generated or analysed during the current study. Declarations Competing interests The authors declare no competing interests. Footnotes Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Contributor Information Wan Qin, Email: wanqinhust@hotmail.com. Xianglin Yuan, Email: yuanxianglin@hust.edu.cn. Binghe Xu, Email: xubinghe@medmail.com.cn. References