Abstract Objective This study aimed to elucidate the expression profile and biological implications of peroxidase 5 (PRDX5) in bladder cancer (BC), specifically investigating its influence on BC progression through modulation of reactive oxygen species (ROS) levels and activation of ferroptosis pathways. Methods We employed urine proteomics data and transcriptomic information from the Cancer Genome Atlas (TCGA) to identify differentially expressed genes in BC tissues, focusing on PRDX5. Using single-cell RNA sequencing (scRNA-seq), we assessed PRDX5 distribution across various cell types in the tumor microenvironment. We conducted in vitro experiments to analyze the impact of PRDX5 on BC cell proliferation, migration, and invasion, while exploring its mechanisms of modulating ROS levels and ferroptosis. In vivo experiments were performed to observe PRDX5's influence on ferroptosis signaling in tissue contexts. Results We found significant upregulation of PRDX5 in BC tissues, with scRNA-seq revealing its enrichment in bladder epithelial cells, correlating with disease advancement and established BC markers. In vitro analyses showed that overexpressed PRDX5 enhanced proliferation, migration, and invasion of BC cells, while PRDX5 knockout produced opposing effects. Additionally, PRDX5 modulated ROS levels and impacted ferroptosis pathways. In vivo experiments confirmed that PRDX5 knockout inhibited tumor growth and activated ferroptosis signaling pathways in tissues. Conclusion Our study highlights the elevated expression of PRDX5 in BC and its role in promoting tumor progression through regulation of ROS levels and ferroptosis. PRDX5 may serve as a promising target for BC treatment, supporting further exploration of its potential in clinical applications. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-025-13881-y. Keywords: Bladder cancer, PRDX5, Ferroptosis, Multi-omics, Urine, Progression, Single cell Introduction Bladder cancer (BC) is one of the most common malignancies affecting the urinary system [[42]1, [43]2]. Global statistics indicate that approximately 540,000 new cases of BC are diagnosed each year, resulting in around 190,000 deaths attributed to the disease [[44]3]. Clinically, BC is classified into two main types based on the depth of invasion into the bladder wall: muscle-invasive bladder cancer (MIBC) and non-muscle-invasive bladder cancer (NMIBC) [[45]4]. Patients with MIBC continue to face a high risk of distant metastasis, even after undergoing radical cystectomy and systemic chemotherapy, with an approximate 5-year survival rate of 32%[[46]5]. Despite the fact that the combination of gemcitabine and cisplatin is considered the first-line chemotherapy regimen for this disease [[47]6], the objective response rate for patients with advanced BC who are undergoing this treatment is only 40–60%, with an increase in overall survival of merely 5% [[48]7, [49]8]. Therefore, it is crucial to identify new therapeutic targets to develop precision treatment strategies. Recent studies have increasingly focused on the molecular mechanisms and therapeutic targets of BC [[50]9]. Redox processes have been observed in cells, and reactive oxygen species (ROS)-byproducts of metabolic activity [[51]10], play a critical role in cellular physiology [[52]11]. Ferroptosis, a form of programmed cell death, is initiated by iron-induced lipid hydro-peroxidation [[53]12, [54]13]. Ferroptosis is regulated by oxidative damage to the cell membrane [[55]14], where abnormal accumulation of phospholipid hydroperoxides (lipid ROS or lipid LOOH) is regarded as a hallmark feature of ferroptosis [[56]15]. Considering that a key characteristic of ferroptosis is the accumulation of ROS leading to cell death, and given that the core function of the Peroxiredoxins (PRDX1-PRDX6, PRDXs) are the regulation of peroxides [[57]16], we hypothesize that a member of the PRDXs may modulate ROS levels, thereby regulating ferroptosis and ultimately influencing the progression of BC. PRDX5 is particularly significant due to its unique subcellular localization and its role in regulating ROS [[58]17]. It is widely accepted that the accumulation of iron ions, which results from lipid peroxidation, along with the subsequent increase in ROS levels, serves as a critical trigger for ferroptosis [[59]18]. A recent study by Fujita et al. [[60]19] highlighted that the deletion of PRDX6 can lead to decreased selenoprotein expression and enhance ferroptosis by inhibiting the expression of glutathione peroxidase 4 (GPX4). Although there is currently a lack of direct evidence linking PRDX5 to the regulation of ferroptosis, emerging research suggests that PRDX5 may play a crucial role in this process due to its known antioxidant properties and involvement in ROS regulation. Given this potential link, this study aims to investigate how PRDX5 modulates ferroptosis and assess its impact on the development and progression of BC. Materials and methods Multi-omics integration for differential gene identification This study collected urine samples from five patients diagnosed with BC and fresh urine samples from five healthy control individuals, in adherence to the Declaration of Helsinki and ethical guidelines set by the Institutional Medical Ethics Committee of Lanzhou University Second Hospital, all participants provided signed written informed consent prior to their inclusion in the study. The pathological diagnoses for each BC patient were independently confirmed by at least two certified pathologists. Furthermore, this investigation utilized the BC dataset from the Cancer Genome Atlas (TCGA, [61]https://portal.gdc.cancer.gov/), which includes gene expression profiles and survival data for 414 BC patients surviving more than 60 days post-diagnosis, along with 19 adjacent normal tissue samples. Transcriptome RNA sequencing and single-cell RNA-Seq data analysis UMUC-3 cells with a knockout of PRDX5, along with control UMUC-3 cells, were treated with TRIzol reagent (Invitrogen) and immediately to liquid nitrogen for cryopreservation. All subsequent procedures were conducted by Majorbio Bio-Pharm Technology Co., Ltd. in Shanghai, China. To analyze the single-cell dataset, we performed dimensionality reduction and clustering on samples from two datasets: [62]GSE135337, which included seven tumor samples and one normal sample, and [63]GSE129845, which encompassed three normal samples. Cell type annotations were assigned based on cell type-specific marker genes previously identified [[64]20]. The single-cell RNA sequencing (scRNA-seq) data underwent preprocessing utilizing the 'Seurat' R package, following detailed methodological protocols outlined in prior research. Principal component analysis was used to the dataset to reduce dimensionality, and the top 15 principal components were selected for further analysis and visualization. To refine the classification of cell populations, utilized the UMAP algorithm for dimensionality reduction and visualization, building upon the PCA results. Finally, we utilized the k-nearest neighbor algorithm to subdivide the cell populations, setting the resolution parameter to 0.1. Tissue and cell lines Five pairs of fresh-frozen BC tissues, along with their corresponding adjacent normal tissues, were surgically obtained and immediately preserved in liquid nitrogen for subsequent analysis of PRDX5 protein expression. Additionally, 16 normal tissue samples and 16 BC tissue samples were collected to assess PRDX5 RNA expression levels. All clinical samples were collected with informed consent from the patients and the collection was approved by the Medical Ethics Committee of the Lanzhou University Second Hospital. SV-HUC-1 cells, which are immortalized human ureteral epithelial cells, were cultured in DMEM (L110KJ; BasalMedia, Shanghai, China). In contrast, BC cell lines, including UMUC-3, 5637, and J82, were maintained in RPMI-1640 medium (L210KJ; BasalMedia, Shanghai, China). All cell lines were procured from the Key Laboratory of Urinary System Diseases at the Lanzhou University Second Hospital, located in Lanzhou City, Gansu Province, China. Each culture medium was supplemented with 10% fetal bovine serum (FBS) (ST30-3302; PAN-Biotech, Germany). All cultures were maintained at 37 °C in a humidified incubator containing 5% CO[2]. Quantitative reverse transcription PCR (qRT-PCR) Total RNA was extracted following the manufacturer's instructions using TRIzol Reagent (AcuracyBio). After RNA extraction, reverse transcription was carried out, and real-time quantitative PCR (qPCR) analysis was conducted using Prime-Script RT Master Mix (Takara Bio) and the SYBR Premix Ex Taq kit (Takara Bio). The primer sequences used in this study are provided in Supplementary Table 1. Western blotting Total protein was extracted using RIPA lysis buffer (Solarbio). Subsequently, 20 μg of protein was subjected to electrophoresis and transferred to polyvinylidene difluoride (PVDF) membranes, and the blots were cut prior to incubation with antibodies during blotting. Following transfer, the membranes were incubated with the appropriate primary and secondary antibodies, and protein signals were visualized using chemiluminescence on an ECL detection system. Densitometric analysis of protein band intensities was performed using ImageJ software. The antibodies utilized in this study included PRDX5 (ab180587; Abcam, USA), GAPDH (HY-P80137; MedChemExpress, China), and the Ferroptosis Essentials Antibody Kit (PK30002; Proteintech, China). CRISPR/Cas9 gene editing technology To establish the PRDX5 knockout cell line, two single-guide RNAs (sgRNAs) were designed: sg1-PRDX5 (5′-CACCGGGCTATATACTCGTCGGTGGTTT-3′) and sg2-PRDX5 (5′-CACCATTACTAGATGATTCGCGTTT-3′). These sgRNAs were synthesized using the Q5 Site-Directed Mutagenesis Kit (E0552S; New England Biolabs, USA) and subsequently cloned into the lenti-Rv2 plasmid (Addgene). After co-transfecting HEK293T cells with the psPAX2 and pMD2.G packaging plasmids, viral supernatants were harvested to infect UMUC-3 and J82 cells. Monoclonal cell lines were generated by limiting dilution in 96-well plates following selection with puromycin. The successful PRDX5 knockout was confirmed by Sanger sequencing and Western blot analysis. Construction of overexpression cell line and lentiviral transfection The full-length coding sequence of human PRDX5 was cloned into the pLV3-CMV-3 × FLAG-mCherry-Puro vector (Miaoling Biological, China). HEK293T cells were co-transfected with the psPAX2 and pMD2.G vectors, and viral supernatants were collected to infect UMUC-3, J82, and T24 cells. Stable cell lines overexpressing PRDX5 were generated after puromycin selection. Cell transfection was performed using Polybrene (Solaibao, China) following the manufacturer's protocol. Cell counting kit (CCK) −8 and colony formation assay We initially seeded UMUC-3 and J82 cells, which were in the logarithmic growth phase, at a density of 2000 cells per well in 96-well plates. After allowing the cells to adhere, we added CCK-8 reagent (Biosharp) diluted in serum-free medium at a ratio of 9:1 to each well. Following a 2-h incubation period, the absorbance was measured at 450 nm (OD450) using a microplate reader. This process was repeated at regular intervals up to 96 h to comprehensively assess the cell proliferation rate. For the negative control, we used cell-free medium and similarly incubated it with the CCK-8 reagent. At the end of the incubation period, the absorbance was measured at 450 nm using a microplate reader. To determine the percentage of cell proliferation relative to the negative control, the absorbance values of each experimental group were divided by the absorbance value of the negative control (NC group) and then multiplied by 100. This calculation provided the cell proliferation rate as a percentage, thereby enabling the quantitative analysis of the proliferation rates of UMUC-3 and J82 cells. To further investigate long-term proliferation capability, we performed colony formation assays. Cells were seeded at low densities (500 cells per well) in 6-well plates and allowed to grow for 10–14 days until visible colonies formed. During this period, the culture medium was refreshed every three days to ensure optimal growth conditions. After the incubation period, the colonies were fixed with 4% paraformaldehyde for 30 min and stained with 0.1% crystal violet for 15 min. Colonies containing more than 50 cells were counted under a microscope. The number of colonies provided an indication of the cells' ability to proliferate and form colonies, thereby assessing their long-term proliferative potential. Wound healing, migration, and invasion assays In the wound healing experiments, BC cells were cultured to confluence in 6-well plates. A sterile pipette tip was then used to create a uniform scratch in the center of each well, which served as a marker for subsequent analysis. After that, unattached cells were removed by washing with phosphate-buffered saline, and the remaining cells were incubated in serum-free medium to facilitate continued cultivation. The migratory capacity of the cells was evaluated by measuring the closure of the scratch at 0, 24, and 48 h after the injury. ImageJ software was employed to calculate the area of the scratch at both 0, 24, and 48 h after scratching. The migration rate of cells was then represented by the ratio of the reduced area over time. For the migration assays, 4 × 10^4 BC cells were seeded in the upper chamber of a transwell plate with 8 µm pores, and 700 µL of complete medium was added to the lower chamber. After a 24-h incubation period, the cells that migrated through the membrane were fixed with 4% paraformaldehyde and subsequently stained with crystal violet for visualization. In the invasion experiments, transwell plates with 8 µm pores were first coated with a layer of Matrigel (Corning, USA) to simulate the extracellular matrix environment. After the Matrigel solidified, 8 × 10^4 BC cells were inoculated into the upper chamber, while the lower chamber was filled with complete medium. After a 24-h incubation, the invading cells that crossed the membrane were fixed in 4% paraformaldehyde and stained with crystal violet for quantification. 3D matrigel drop invasion assay and flow cytometry analysis BC cells (5 × 10^4) were resuspended in 10 μL of Matrigel (Corning, USA) and carefully added to each well of 24-well plates using a vertical dropwise technique. The plates were left undisturbed for 20 min to allow the Matrigel to solidify into a droplet structure. After solidification, complete medium was added to each well. The invasive capacity of the cells was assessed by measuring the distance of migration from the edge of the Matrigel drops after 6 days of culture. To evaluate apoptosis, an Annexin V-FITC/PI Apoptosis Detection Kit (AT101; Multi Sciences) was employed. Following the manufacturer's protocol, cell aliquots were stained with FITC-Annexin V and propidium iodide (PI) to identify apoptotic cells, enabling a comprehensive analysis of cell viability and apoptosis. MitoSOX, TMRE, JC-1, ROS assays BC cells were cultured in confocal dishes and treated with MitoSOX probes (MX4313, Mkbio) in addition to Hoechst 33,342 (C0031, Solaibao) for a duration of 30 min. After the treatment, the cells were examined using a Zeiss LSM 510 confocal microscope (Germany) equipped with a 60 × 1.3 NA oil immersion objective. Initially, 1 × 10^6 cells were seeded into the confocal dishes. The following day, 0.5 mL of working solutions of JC-1 dye (C2005, Beyotime) and Hoechst 33,342 were added to each dish and incubated for 25 min at a temperature of 37 °C. Subsequently, the cells were observed under confocal microscopy. Furthermore, BC cells in confocal dishes were treated with TMRE probes (C2001S, Beyotime) and Hoechst 33,342 for an additional 30 min, after which imaging was conducted to assess mitochondrial membrane potential. Intracellular levels of ROS were evaluated using the DCFH-DA fluorescent probe (HY-D0940, MedChemExpress). For this analysis, BC cells were plated in six-well plates and treated with DCFH-DA at a final concentration of 25 μM, followed by incubation in the dark for 30 min. Fluorescence intensity was quantified using a Beckman CytoFLEX flow cytometer. Xenograft model All animal experiments and procedures were conducted with the approval of the Animal Welfare and Ethics Committee of the Second Hospital of Lanzhou University. Ten 4-week-old male nude mice (Huachuang Sino, China) were randomly allocated into two groups (KO-PRDX5 and NC-PRDX5), with five mice in each group. The cells for both the KO-PRDX5 and NC groups were collected and suspended in phosphate-buffered saline at a concentration of 2 × 10^7 cells/mL, then mixed with Matrigel (Corning, USA) at a 1:1 ratio. A total of 100 μL of this cell suspension was subcutaneously injected into the axillary region of the nude mice. Throughout the experimental period, the body weight and tumor volume of the mice were measured every 5 days. On day 15, the mice were euthanized via intraperitoneal injection of an overdose of pentobarbital sodium. Following euthanasia, the tumors were excised, weighed, their dimensions recorded, and photographed. Tumor volume was assessed using caliper measurements and calculated using the following formula: Volume = 0.5 × length × width^2. Statistical analysis Data are presented as mean ± standard deviation from a minimum of three independent experiments, each conducted in triplicate. The following statistical tests were performed with Prism 9 software (GraphPad). The following statistical analyses were used: t test (unpaired and two-tailed) and one-way analysis of variance (ANOVA) with Dunnett’s multiple comparisons test. P-values < 0.05 were considered significant, represented by "*", P values less than 0.01 were denoted as "**", P values less than 0.001 were indicated as "***", while P values less than 0.0001 were represented by "#". Results Integrated multi-omics approach for identifying differentially expressed genes in BC Our previous study successfully executed RNA extraction and LC–MS detection of urinary samples from BC patients and healthy controls [[65]21], Building upon this foundation, our objective was to identify noninvasive diagnostic markers for BC and identify highly differentially expressed genes. We initially screened for differentially expressed genes in TCGA using volcano plots (Fig. [66]1A). To enhance the reliability of the selected markers, we also incorporated data from urine proteomics. Using the urine proteomics volcano plot (Fig. [67]1B) and the combined multi-omics differential distribution plot (Fig. [68]1C), we identified differential molecules that were highly expressed in BC. Our focus was on molecules that are readily secreted into urine for early diagnosis. Thus, we prioritized markers showing elevated expression levels across multiple omics platforms. A Venn diagram (Fig. [69]1D) highlighted the overlap between the 3,895 highly expressed DEGs in TCGA-BC transcriptomics (left circle) and the 352 in urine proteomics (right circle), revealing 32 overlapping molecules (center intersection). Incorporating the 32 hub genes into a lasso regression analysis (Fig. [70]1E), we found that the model achieved its best fit when the Beta coefficient was set to 7. Subsequently, seven molecules were chosen for inclusion in the prognostic model. Among these, PRDX5 emerged as the most significant (Fig. [71]1F), with higher expression levels associated with more accurate prognostic predictions. Further exploration of PRDX5 expression in various cancers, derived from the TCGA database, indicated a high expression level in the majority of cancer types (Fig. [72]1G). Fig. 1. [73]Fig. 1 [74]Open in a new tab Selection of differential genes in BC through multi-omics integration. A Volcano plot of differentially expressed genes in the TCGA-BLCA gene expression profile; (B) Volcano plot of differentially expressed genes in the urinary proteomics gene expression profile; (C) Rank versus expression abundance curve for differentially expressed genes combining TCGA-BLCA and urinary proteomics data; (D) A Venn diagram illustrating the intersection of highly expressed differential genes from urine proteomics data and the TCGA-BLCA gene expression profile, along with their respective top-ranked differential genes; (E) Screening of Lasso regression coefficients for 32 hub genes; (F) 1-year, 3-year, and 5-year nomograms used to predict overall survival in BC; (G) Expression levels of PRDX5 across different cancer types Expression characteristics of PRDX5 in BC and its correlation with clinical and pathological features To investigate the expression profile of PRDX5 in BC and its potential biological relevance, we initially analyzed urine samples from five healthy volunteers and five BC patients. The analysis revealed a significant upregulation of PRDX5 in the BC cohort (Fig. [75]2A). This observation suggests that PRDX5 may serve as a noninvasive early diagnostic biomarker for BC. Fig. 2. [76]Fig. 2 [77]Open in a new tab Differences in PRDX5 expression levels in BC and their correlation with clinical pathological features. A PRDX5 expression levels in urine proteomics from 5 BC patients and 5 normal individuals; (B) Comparison of PRDX5 expression levels between BC and normal tissues; (C) Paired comparison of PRDX5 expression levels between BC and normal tissues; (D) Comparison of PRDX5 expression levels among different histological grades; (E) Comparison of PRDX5 expression levels among different subtypes; (F) Relationship between PRDX5 expression levels and overall survival of BC patients; (G) Immunohistochemical staining of PRDX5 in adjacent non-cancerous tissues; (H) Immunohistochemical staining of PRDX5 in BC tissues To validate these preliminary findings, we used data from the TCGA-BLCA dataset. The analysis of this dataset demonstrated a marked increase in PRDX5 expression in BC tissues compared to normal tissue controls (Fig. [78]2B). Importantly, this significant difference persisted when cancer samples were matched to their adjacent non-tumor tissues (Fig. [79]2C). We further explored the expression patterns of PRDX5 across different pathological grades of BC. Notably, we observed a significant upward trend in PRDX5 expression in both low-grade and high-grade tumors (Fig. [80]2D). In cases of papillary BC, PRDX5 expression reached its highest level (Fig. [81]2E), highlighting its potential role in BC progression. While there were no significant differences observed in overall survival (Fig. [82]2F) between the groups classified based on high and low PRDX5 expression, it is worth noting that patients with high levels of PRDX5 exhibited a trend towards a poorer prognosis. Taking this cumulative evidence into account, we believe that PRDX5 is a promising candidate biomarker for the early noninvasive diagnosis of BC. In addition, immunohistochemical data obtained from the Human Protein Atlas (HPA) database confirmed that the staining intensity of PRDX5 in adjacent normal tissues (H-score: 25.6 ± 5.2) (Fig. [83]2G) was significantly lower than that in BC tissues (H-score: 188.3 ± 46.2) (Fig. [84]2H), and the staining intensity and the proportion of positive cells in tumor tissues were significantly higher than those in normal tissues. Single-cell sequencing reveals the role of PRDX5 in the bladder tumor microenvironment In this study, we explored the functional characteristics of PRDX5 across different cell types within the tumor microenvironment of BC using scRNA-seq technology. Subsequently, we applied dimensionality reduction techniques to analyze the single-cell BC data, allowing us to visualize spatial structural information (Figs. [85]3A, B). We further examined the levels of PRDX5 enrichment across different subcellular types (Figs. [86]3C-H). Notably, our results indicated significant enrichment of PRDX5 in bladder urothelial cells (Fig. [87]3D), a trend that was corroborated by findings from the independent [88]GSE130001 single-cell dataset (Figs. [89]3G, H). Fig. 3. [90]Fig. 3 [91]Open in a new tab ScRNA-seq analysis of cell type distribution in the bladder tumor microenvironment and its relationship with PRDX5 expression. A t-SNE plot showing the distribution of different cell types in the bladder tumor microenvironment; (B) UMAP plot illustrating the distribution of various cell types in the bladder tumor microenvironment; (C) PRDX5 expression levels among different cell types within the bladder tumor microenvironment; (D) PRDX5 expression levels across different cell clusters in the bladder tumor microenvironment; (E) Dimensionality reduction analysis of different cell types in the bladder tumor microenvironment; (F) Cluster analysis of PRDX5 expression levels among various cell types in the BC microenvironment; (G) Cluster analysis of different cell types in the bladder tumor microenvironment from the single-cell dataset [92]GSE130001; (H) PRDX5 expression levels in different cell types within the bladder tumor microenvironment from the single-cell dataset [93]GSE130001 These observations indicate that the expression pattern of PRDX5 within the BC microenvironment is specific to certain cells, with notably elevated expression levels in bladder epithelial cells. This suggests that PRDX5 may play a crucial role in the development of BC and provides valuable insights for a more comprehensive understanding of BC pathogenesis, as well as the potential advancement of targeted treatment strategies. Single-cell sequencing investigates the localization and role of PRDX5 in bladder tumors To investigate the function of PRDX5 in bladder epithelial cells, we used scRNA-seq data for cluster analysis (Fig. [94]4A). We identified six distinct cell types: endothelial cells, epithelial cells, fibroblasts, macrophages, and plasma cells (Fig. [95]4B) based on previously established cell type-specific marker genes from our research group [[96]20]. The results indicated that PRDX5 was more prevalent and abundant in tumor cells compared to normal tissue cells (Fig. [97]4C). Importantly, PRDX5 showed significant enrichment in bladder epithelial cells (Fig. [98]4D). Fig. 4. [99]Fig. 4 [100]Open in a new tab Analysis of scRNA-seq data in BC tissues. A Eight clusters were annotated based on cell type-specific marker genes following scRNA-seq of BC samples; (B) Cells were classified into five main types based on transcriptomic characteristics: endothelial cells, epithelial cells, fibroblasts, macrophages, and plasma cells; (C) Differences in PRDX5 expression between single cells from normal tissues and tumor tissues; (D) Expression levels of PRDX5 in various cell types, with notably higher expression in bladder epithelial cells; (E) Relationship between tumor T staging and PRDX5 expression in single-cell BC tissues; (F) Positive correlation of PRDX5 with common epithelial malignancy-related genes in BC, such as GATA3, KRT7, S100P, TP63, and UPK2 Additionally, we examined the relationship between PRDX5 expression and bladder tumor T stage at the single-cell level. Our findings revealed that PRDX5 expression significantly increases with advancing T stage (Fig. [101]4E). This suggests that PRDX5 may play a crucial role in the progression of BC. Further analysis demonstrated that in BC samples, PRDX5 shows a positive correlation with several epithelial malignancy-associated genes (GATA3, KRT7, S100P, TP63, and UPK2) (Fig. [102]4F). This correlation indicates that PRDX5 might also be involved in regulating the development of BC. In summary, our scRNA-seq analysis has uncovered a significant upregulation of PRDX5 expression in bladder epithelial cells as the disease progresses. More importantly, the strong correlation between PRDX5 and specific BC markers suggests that PRDX5 not only plays a pivotal role in the onset of BC but could also serve as an important indicator for assessing patient prognosis. These findings provide new insights into the mechanisms by which PRDX5 contributes to BC and lay the groundwork for developing diagnostic or therapeutic strategies based on PRDX5. Analysis of PRDX5 expression in BC cell lines and tissues To investigate the differential expression of PRDX5 between BC cell lines (UMUC-3, J82, T24, 5637) and normal bladder epithelial cells (SV-HUC-1), we employed two validation methods: RT-PCR (Fig. [103]5A) and Western blot analysis (Fig. [104]5B). The results indicated that PRDX5 expression levels were significantly higher in BC cells (UMUC-3, J82, T24) compared to normal bladder epithelial cells (SV-HUC-1). Furthermore, analysis of data from the CELL database revealed that PRDX5 exhibited high expression levels in most BC cell lines, although the selected lines (UMUC-3, J82, T24) had relatively lower expression levels (Fig. [105]5C). Fig. 5. [106]Fig. 5 [107]Open in a new tab Expression of PRDX5 in BC cell lines and clinical patients. A mRNA expression levels of PRDX5 in normal bladder epithelial cell line SV-HUC-1 and BC cell lines UMUC-3, J82, T24, and 5637; (B) Protein expression levels of PRDX5 in normal bladder epithelial cell line SV-HUC-1 and BC cell lines UMUC-3, J82, T24, and 5637; (C) mRNA expression profile of PRDX5 in BC cell lines; (D) mRNA expression levels of PRDX5 in BC tissues and adjacent non-cancerous tissues; (E) Protein expression levels of PRDX5 in BC tissues and adjacent non-cancerous tissues; (F) mRNA expression levels following PRDX5 overexpression in BC cell lines; (G) Protein expression levels following PRDX5 overexpression in BC cell lines; (H) mRNA expression level assessment of KO-PRDX5 efficiency in BC cell lines; (I) Protein expression level assessment of KO-PRDX5 efficiency in BC cell lines. Pa, adjacent non-cancerous tissue; Ca, BC tissue; ns, not significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; #, p < 0.0001 Additionally, RT-PCR validation was performed using 16 pairs of BC and adjacent tissue samples collected from the Lanzhou University Second Hospital. This analysis confirmed that the expression of PRDX5 was significantly higher in BC tissues compared to adjacent normal tissues (Fig. [108]5D). Furthermore, examination of tissue proteins from five BC patients showed a similar trend, with significantly higher levels of PRDX5 expression in cancerous tissue compared to adjacent normal tissues (Fig. [109]5E). Taken together, these findings support our hypothesis that PRDX5 may have a role in the development of BC. To further investigate the potential impact of PRDX5 in BC, we introduced lentiviral vectors overexpressing PRDX5 into BC cell lines (UMUC-3, J82, T24). The efficacy of overexpression was assessed through RT-PCR (Fig. [110]5F) and Western blot analysis (Fig. [111]5G), both confirming high levels of PRDX5 overexpression. Conversely, we employed CRISPR-Cas9 gene editing technology to knockout PRDX5 in the BC cell lines (UMUC-3, J82) that had relatively high expression levels. Monoclonal cell lines were selected from these cultures, and two clonal knockout fragments were validated. Analysis through RT-PCR (Fig. [112]5H) and Western blot (Fig. [113]5I) confirmed the successful establishment of KO-PRDX5 cell lines from BC cell lines UMUC-3 and J82. Impact of PRDX5 on the biological behavior of BC cells To systematically explore the potential role of PRDX5 in the biological behavior of BC cell lines (UMUC-3, J82, T24), we employed a series of in vitro experimental methods for comprehensive assessment. Initially, a CCK-8 cell proliferation assay (Fig. [114]6A) revealed a significant association between PRDX5 overexpression and an increased proliferation rate of BC cells. This finding was further supported by colony formation assays, which showed that the PRDX5-overexpressing group exhibited denser and larger clonal colonies compared to the control group (Fig. [115]6B), highlighting the role of PRDX5 in promoting BC cell proliferation and growth. Fig. 6. [116]Fig. 6 [117]Open in a new tab Effects of PRDX5 overexpression in BC cell lines (UMUC-3, J82, and T24). A Comparison of cell proliferation ability over time in BC cell lines overexpressing PRDX5; (B) Cloning formation assays demonstrating enhanced growth and proliferative capacity in BC cell lines with PRDX5 overexpression; (C) Scratch migration assays indicating increased migration ability in cell lines overexpressing PRDX5; (D) Transwell migration assays showing enhanced migration capability in cell lines with PRDX5 overexpression; (E) Transwell invasion assays revealing increased invasive ability in cell lines with PRDX5 overexpression; (F) Enhanced invasive capability in 3D spheroid models of PRDX5-overexpressing UMUC-3; (G) Enhanced invasive capability in 3D spheroid models of PRDX5-overexpressing J82 Subsequent experiments aimed to evaluate alterations in cell migration and invasion. Cell scratch assays (Fig. [118]6C) demonstrated that the overexpression of PRDX5 considerably augmented the migratory activity of BC cells. Transwell migration assays (Fig. [119]6D) not only validated this enhancement but also revealed that PRDX5 overexpression was associated with a noteworthy rise in cell invasion (Fig. [120]6E), thereby offering more profound insights into the invasive capability of BC cells. To assess the long-term effects, we employed a 3D sphere-forming invasion model (Figs. [121]6F, 6G), which consistently demonstrated that PRDX5-overexpressing cells exhibited a sustained increase in invasive capacity over time, thereby supporting the idea that PRDX5 functions as a positive regulator of BC progression. In contrast, we examined the impact of PRDX5 deletion on BC cell behavior using CRISPR-Cas9 knockout technology. The CCK-8 proliferation assay (Fig. [122]7A) revealed that KO-PRDX5 led to a decreased proliferation rate in BC cells. Colony formation assays (Fig. [123]7B) further confirmed this finding, indicating that both the number and size of colonies in the knockout group were smaller than those in the control group. This suggests that PRDX5 is crucial for maintaining proliferative activity in BC cells. Fig. 7. [124]Fig. 7 [125]Open in a new tab Effects of KO-PRDX5 in BC cell lines (UMUC-3 and J82). A Comparison of cell proliferation ability over time in BC cell lines with KO-PRDX5; (B) Cloning formation assays indicating reduced growth and proliferative capacity in BC cell lines with KO-PRDX5; (C) Scratch migration assays demonstrating decreased migration ability in cell lines with KO-PRDX5; (D) Transwell migration assays showing reduced migration capability in cell lines with KO-PRDX5; (E) Transwell invasion assays revealing decreased invasive ability in cell lines with KO-PRDX5; (F) Reduced invasive capability in 3D spheroid models of KO-PRDX5 UMUC-3; (G) Reduced invasive capability in 3D spheroid models of KO-PRDX5 J82 Cell scratch assays (Fig. [126]7C) and transwell assays (Figs. [127]7D, E) demonstrated significant decreases in cell migration and invasion following PRDX5 knockout, thereby providing additional evidence of its involvement in facilitating invasive behavior. Ultimately, the results from the 3D sphere-forming invasion model (Figs. [128]7F, G) further supported these findings, indicating a gradual decline in invasion over time in KO-PRDX5 BC cells. PRDX5 regulates cell death in BC cell lines and its potential mechanisms In the UMUC-3 cell line, PRDX5 overexpression was significantly associated with a reduction in cell death compared to the control cells (2.15% vs. 3.82%). This phenomenon was also observed in the J82 cell line (2.13% vs. 4.84%) (Fig. [129]8A). Conversely, PRDX5 knockout led to a significant increase in cell death within the UMUC-3 cell line (3.19% vs. 10.48%), with a similar trend observed in the J82 cell line (3.67% vs. 10.47%) (Fig. [130]8B). These findings highlight the crucial role of PRDX5 in modulating cell death in BC. Fig. 8. [131]Fig. 8 [132]Open in a new tab Impact of PRDX5 expression levels on apoptosis, gene expression, and ROS levels in BC cell lines. A Comparison of apoptosis rates between PRDX5 overexpressing cell lines and control groups; (B) Comparison of apoptosis rates between KO-PRDX5 cell lines and control groups; (C) Differences in the expression of classic apoptosis pathway genes between PRDX5 overexpressing and control groups in UMUC-3; (D) Differences in the expression of classic apoptosis pathway genes between PRDX5 overexpressing and control groups in J82; (E) Differences in the expression of classic apoptosis pathway genes between KO-PRDX5 and control groups in UMUC-3; (F) Differences in the expression of classic apoptosis pathway genes between KO-PRDX5 and control groups in J82; (G) Comparison of ROS fluorescence levels between PRDX5 overexpressing cell lines and control groups; (H) Comparison of ROS fluorescence levels between KO-PRDX5 cell lines and control groups; (I) Flow cytometry analysis comparing ROS levels between KO-PRDX5 cell lines and control groups To further elucidate the mechanisms underlying PRDX5's regulatory effects on apoptosis, we assessed the mRNA expression levels of three key genes involved in classical apoptotic pathways in cell lines overexpressing PRDX5 and KO-PRDX5 cell lines. Notably, there were no significant alterations in the expression of markers associated with the mitochondrial apoptotic pathway or the death receptor pathway. In contrast, we observed a marked upregulation of markers related to the endoplasmic reticulum stress pathway in cell lines overexpressing PRDX5 (Figs. [133]8C, D). Meanwhile, while the expression levels of markers for the mitochondrial and death receptor pathways remained largely unchanged in KO-PRDX5 cell lines, we noted a pronounced downregulation of markers related to the ER stress pathway (Figs. [134]8E, F). Collectively, these results suggest that PRDX5-mediated cell death may function independently of classical apoptotic pathways. Considering that the primary function of PRDX5 is the regulation of ROS, and given that ROS accumulation is a pivotal feature of ferroptosis [[135]15], we hypothesize that PRDX5 may influence cell death by modulating ROS levels and impacting ferroptosis signaling pathways. To test this hypothesis, we first evaluated ROS levels in both PRDX5-overexpressing and knockout cell lines. Our results indicated that overexpression of PRDX5 significantly decreased ROS levels in BC cells (Fig. [136]8G), whereas knockout of PRDX5 resulted in a substantial increase in ROS levels (Fig. [137]8H). Flow cytometry assays further confirmed that KO-PRDX5 enhanced ROS production (Fig. [138]8I). Taken together, these findings suggest that PRDX5 regulates cell death through the modulation of ROS levels rather than through conventional apoptotic pathways. This discovery provides valuable insights into the mechanisms by which PRDX5 influences BC biology. PRDX5 influences ferroptosis in BC by regulating lipid metabolism and mitochondrial function To thoroughly investigate the role of PRDX5 in BC, we conducted a transcriptome sequencing analysis comparing KO-PRDX5 lines with control strains in UMUC-3 cells. The findings revealed that the functional enrichment of differentially expressed genes between PRDX5-depleted cell lines and their controls predominantly associated with cancer-related pathways (Fig. [139]9A). Additionally, KEGG pathway enrichment analysis highlighted significant involvement of lipid metabolism and immune system pathways (Fig. [140]9B). Given the critical link between aberrant lipid accumulation and ferroptosis [[141]15], we hypothesize that PRDX5 may influence ferroptosis through its regulatory effects on lipid metabolism, thereby modulating BC cell death. Fig. 9. [142]Fig. 9 [143]Open in a new tab Impact of KO-PRDX5 on gene expression profiles and mitochondrial function in BC cell lines. A Disease-related functional enrichment analysis of differentially expressed genes between KO-PRDX5 cell lines and control groups; (B) KEGG pathway enrichment analysis of differentially expressed genes between KO-PRDX5 cell lines and control groups; (C) Comparison of mitochondrial superoxide (MitoSOX) levels between KO-PRDX5 and control groups in the UMUC-3 cell line; (D) Comparison of mitochondrial superoxide (MitoSOX) levels between KO-PRDX5 and control groups in the J82 cell line; (E) Comparison of mitochondrial membrane potential between KO-PRDX5 and control groups in the UMUC-3 cell line; (F) Comparison of mitochondrial membrane potential between KO-PRDX5 and control groups in the J82 cell line; (G) Mitochondrial fluorescent probe (TMRE) staining showing KO-PRDX5 and control groups in UMUC-3 cells; (H) Mitochondrial fluorescent probe (TMRE) staining showing KO-PRDX5 and control groups in J82 cells To validate this hypothesis, we first assessed the impact of KO-PRDX5 on mitochondrial superoxide levels (as measured by MitoSOX) in the BC cell lines UMUC-3 and J82. Our results indicated a substantial increase in mitochondrial superoxide levels in the KO-PRDX5 cell lines compared to controls (Fig. [144]9C, D). Furthermore, the evaluation of mitochondrial membrane potential using the JC-1 assay demonstrated a marked decline in potential (as indicated by diminished red fluorescence and increased green fluorescence) in the PRDX5-depleted cell lines (Fig. [145]9E, F), suggesting compromised mitochondrial functionality. Complementary experiments employing TMRE fluorescent probes further substantiated these findings, revealing a significant decrease in mitochondrial membrane potential (as indicated by reduced red fluorescence) following KO-PRDX5 in both UMUC-3 and J82 cell lines (Fig. [146]9G, [147]H). In summary, our data suggest that PRDX5 plays a pivotal role in regulating ferroptosis and, consequently, cell death in BC by modulating lipid metabolism and mitochondrial functions. These findings offer valuable insights into mechanisms underlying PRDX5's contribution to the progression of BC. The role of PRDX5 in regulating ferroptosis during BC progression and its in vivo validation To investigate the involvement of PRDX5 in the regulation of the ferroptosis pathway, we initially compared the transcriptomic profiles of control and KO-PRDX5 group (Fig. [148]10A). Notably, our analysis revealed alterations in signaling molecules essential for ferroptosis that were activated following KO-PRDX5 (Fig. [149]10B-E). To further evaluate this hypothesis, we examined the impact of KO-PRDX5 on both UMUC-3 and J82. Our findings showed that PRDX5 depletion led to the activation of the ferroptosis signaling pathway, as evidenced by elevated levels of acyl-CoA synthetase long-chain family member 4 (ACSL4) and kelch-like ECH associated protein 1 (KEAP1) proteins, along with a corresponding decrease in the levels of NRF2 (Nuclear Factor Erythroid 2–related Factor 2), GPX4, and SLC7A11 proteins (Fig. [150]10F). Fig. 10. [151]Fig. 10 [152]Open in a new tab The function of PRDX5 in BC progression. A Volcano plot showing differential gene expression between the NC group and the KO-PRDX5 group; (B) Distribution of relative expression TPM values for ACSL4 in the NC group versus the KO-PRDX5 group; (C) Distribution of relative expression TPM values for KEAP1 in the NC group versus the KO-PRDX5 group; (D) Distribution of relative expression TPM values for NRF2 in the NC group versus the KO-PRDX5 group; (E) Distribution of relative expression TPM values for GPX4 in the NC group versus the KO-PRDX5 group; (F) Changes in the levels of ferroptosis marker proteins due to PRDX5 knock-out in cell lines; (G) Curve graph illustrating changes in tumor volume over time, highlighting differences in tumor growth rates between the NC-PRDX5 and KO-PRDX5 groups; (H) Comparison of tumor weights, demonstrating differences between the NC-PRDX5 and KO-PRDX5 groups; (I) Appearance of tumors between the NC-PRDX5 and KO-PRDX5 groups; (J) Size comparison of tumors between the NC-PRDX5 and KO-PRDX5 groups; (K) Changes in the levels of ferroptosis marker proteins due to PRDX5 knock-out in tumor tissues from nude mice To corroborate the in vivo function of PRDX5 in BC progression, we conducted a subcutaneous xenograft model using nude mice. The results demonstrated that the KO-PRDX5 group exhibited a significantly slower increase in tumor volume over time compared to the control group (Fig. [153]10G). Moreover, the final tumor weight in the KO-PRDX5 group was significantly reduced relative to the control group (Fig. [154]10H). Visual observations of the subcutaneous tumors in the nude mice (Fig. [155]10I) and the quantification of tumor volumes (Fig. [156]10J) further confirmed that KO-PRDX5 effectively inhibited tumor growth. Additionally, analysis of subcutaneous tumor tissues from the xenograft model verified the activation of ferroptosis signaling pathways induced by KO-PRDX5 (Fig. [157]10K). In conclusion, our study elucidates a critical role for PRDX5 in the progression of BC. The knockout of PRDX5 in BC cell lines activated the ferroptosis pathway, as evidenced by increased ACSL4 and KEAP1 protein levels and decreased levels of NRF2, GPX4, and SLC7A11. Moreover, PRDX5 depletion significantly diminished tumor volume and weight, effectively inhibiting tumor growth in the subcutaneous xenograft model. These findings highlight the potential of PRDX5 as a therapeutic target for BC treatment. Discussion In our study, we observed that PRDX5 was significantly upregulated in BC tissues, with the highest expression levels detected specifically in cases of papillary BC. Although our analysis did not reveal a statistically significant correlation between elevated PRDX5 expression and overall or disease-specific survival rates in patients, a discernible trend suggestive of poorer prognosis emerged among those with high PRDX5 levels. This observation aligns with prior research that identifies PRDX5 as a negative prognostic marker in various cancer types, further highlighting its potential relevance in the context of BC outcomes [[158]22, [159]23]. Additionally, our study represents the first report of PRDX5 expression levels in urine samples from BC patients, suggesting its potential as a noninvasive diagnostic biomarker for future clinical applications. Through the application of scRNA-seq, we identified a significant enrichment of PRDX5 in the epidermal cells of BC, indicating its potential critical role in the disease’s pathogenesis. This cell-specific expression pattern offers a fresh perspective on the functional implications of PRDX5 within the context of BC. Moreover, we observed that as BC progresses, PRDX5 expression levels are notably elevated and closely correlated with specific markers of BC, such as GATA3, KRT7, S100P, TP63, and UPK2, all of which are implicated in the malignant transformation of bladder epidermal cells. This underscores the relevance of PRDX5 in the disease's trajectory and its potential role in facilitating BC progression. Through a comprehensive series of in vitro and in vivo experiments, our study demonstrated that the overexpression of PRDX5 in BC cell lines markedly enhanced cellular proliferation, migration, and invasion. Conversely, silencing PRDX5 expression resulted in a significant reduction in these tumorigenic behaviors. These results corroborate earlier research highlighting the role of PRDX5 in cancer biology, reinforcing its influence as a pivotal factor in BC progression dynamics [[160]22]. These findings further underscore the critical role of PRDX5 in the development of BC. Prior research has established that PRDX5 acts as an oncogene across various cancer types. For instance, Lee et al. [[161]24] reported elevated levels of PRDX5 in oral cancer, highlighting its involvement in promoting tumor cell proliferation. Likewise, Xie et al.[[162]23] found that PRDX5 expression was significantly increased in hepatocellular carcinoma, where it was associated with tumor invasion and metastasis. Our study builds upon this existing body of knowledge, specifically illuminating the mechanisms by which PRDX5 exerts its effects in the context of BC progression. Further studies revealed that PRDX5 impacts cell death by regulating ROS levels rather than through traditional apoptotic pathways. This finding aligns with existing literature on the role of PRDX5 in redox balance [[163]25]. Given that members of the PRDX family play significant roles in ferroptosis progression, Cui et al.[[164]26] revealed that PRDX3 undergoes specific hyperoxidation during ferroptosis, establishing it as a novel biomarker for ferroptosis. Additionally, a recent study by Fujita et al. [[165]27] indicated that loss of PRDX6 could promote ferroptosis onset. Considering that ROS accumulation is a hallmark of ferroptosis and PRDX5 primarily regulates ROS levels[[166]28], our study provides the first evidence that PRDX5 influences the ferroptosis pathway by regulating lipid metabolism. This mechanism significantly affects the fate of BC cells, highlighting the intricate relationship between PRDX5, lipid metabolism, and ferroptosis in BC. We observed alterations in ferroptosis-associated markers following PRDX5 knockout, indicating its influence on this pathway. Additionally, our in vivo experiments demonstrated that the activation of ferroptosis effectively inhibited tumor growth. These findings are consistent with literature highlighting the role of ferroptosis in BC and other malignancies, further underscoring the potential therapeutic implications of targeting this pathway in cancer [[167]29, [168]30]. The role of PRDX5 in BC indicates its potential as a therapeutic target. By influencing cell fate through the regulation of the ferroptosis pathway, targeting PRDX5 may represent a novel therapeutic strategy. Research has shown that modifications to ferroptosis regulators can significantly affect tumor responses to therapy. Repurposing drugs and administering various vitamins, such as vitamin D, as prophylactics can have a modulatory effect and positively impact redox potential in cancer [[169]31]. For instance, the pharmacological inhibition of GPX4 activity has been shown to induce ferroptosis, leading to reduced tumor growth [[170]32]. This finding enhances the understanding of ferroptosis in cancer, offering valuable insights into the mechanisms by which PRDX5 influences BC development. It is noteworthy that the expression of PRDX5, as an antioxidant enzyme, is often influenced by various proteins, particularly NRF2 [[171]25]. NRF2 is a key regulatory factor in cellular antioxidant responses, initiating the expression of a series of antioxidant genes by binding to antioxidant response elements. Existing studies have shown that NRF2 can directly regulate the expression of multiple antioxidant enzymes, including PRDX5 [[172]33]. Evidence indicates that NRF2 not only influences the expression of antioxidant enzymes through transcriptional regulation but can also interact directly with PRDX5, thereby enhancing their activity and stability[[173]34]. Although there is currently no direct evidence to demonstrate a similar interaction between PRDX5 and NRF2 in BC, understanding the potential interaction between PRDX5 and NRF2 could not only elucidate the reasons behind the high expression of PRDX5 in BC but also provide a basis for developing new therapeutic strategies. Inhibitors targeting the NRF2 signaling pathway may represent a potential method for reducing PRDX5 expression and enhancing chemotherapy sensitivity. This study represents the first systematic investigation into the expression characteristics of PRDX5 in BC and its specific mechanisms in disease progression, while also establishing a relationship between PRDX5 and other tumor markers, thereby paving new avenues for BC treatment. Future research should focus on exploring the relationship between PRDX5 and NRF2 to provide theoretical support for targeted therapy strategies. Conclusion In this study, we demonstrated that PRDX5 is highly expressed in BC and confirmed its role in promoting tumor progression by regulating ROS levels and the ferroptosis pathway. These findings not only enhance our understanding of BC pathogenesis but also establish a theoretical basis for developing novel therapeutic strategies targeting PRDX5. Future research should investigate the efficacy and safety of PRDX5 as a potential therapeutic target in the context of BC, paving the way for innovative treatment approaches to improve patient outcomes. Supplementary Information [174]Supplementary Material 1^ (4.4MB, docx) [175]Supplementary Material 2^ (14.2KB, docx) Abbreviations BC Bladder cancer PRDX5 Peroxidase 5 ROS Reactive oxygen species TCGA The Cancer Genome Atlas scRNA-seq Single-cell RNA sequencing MIBC Muscle-invasive bladder cancer NMIBC Non-muscle-invasive bladder cancer GPX4 Glutathione peroxidase 4 CCK-8 Cell Counting Kit-8 NRF2 Nuclear Factor Erythroid 2–related Factor 2 ACSL4 Acyl-CoA synthetase long-chain family member 4 KEAP1 Kelch-like ECH associated protein 1 Authors' contributions Shun Wan, Kun-Peng Li, and Si-Yu Chen are mainly responsible for writing manuscripts, later revision of language and grammar, and article format. Shan-hui Liu and Li Yang are mainly responsible for writing manuscripts and article format. Chen-Yang Wang and Kun Cheng are accountable for this article's leading writing and structure arrangement. Jian-Wei Yang, Li-yun Ding, and Tuan-jie Che are mainly responsible for data access. They have made corresponding contributions to this article. All authors have read and approved the final manuscript. Funding This study was supported by Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital (Grant numbers CY2021-MS-A12); Investigation of the Effects and Mechanisms of HTRA1 on Bladder Tumors Through Combined Non-invasive Urinary Metabolomics (Grant numbers 22YF7FA090); Exploring the Accuracy and Mechanisms of Non-Invasive Urinary HTRA1 Molecular Detection in BC Diagnosis (Grant numbers 23JRRA1006); Multimodal Urinary Analysis for the Identification of Bladder Tumor Biomarkers and Investigation into the Molecular Mechanisms of HTRA1 (Grant numbers 2023–4-34). Data availability Data were download from the TCGA database- BLCA ([176]https://portal.gdc.cancer.gov). All data generated or analyzed during this study are included in this published article. Declarations Ethics approval and consent to participate Our study adhered to the Declaration of Helsinki and ethical guidelines established by the Institutional Medical Ethics Committee of Lanzhou University Second Hospital. All clinical samples were collected with informed consent from the patients and the collection was approved by the Medical Ethics Committee of the Lanzhou University Second Hospital (D2024-774). Consent for publication Not applicable. Competing Interest 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. Shun Wan, Kun-peng Li and Si-yu Chen contributed equally to this work. Contributor Information Shan-hui Liu, Email: 1106074594@qq.com. Li Yang, Email: ery_yangli@lzu.edu.cn. References