Abstract Background CSMD2 has been reported as a potential prognostic factor in several cancers. However, whether CSMD2 affects bladder cancer (BC) remains unclear. Methods Public data were obtained from the TCGA ([33]https://cancergenome.nih.gov) databases. CSMD2expression and its prognostic value were analyzed using bioinformatics methods. CSMD2 mRNA level in patients with BC and BC cell lines was evaluated via quantitative reverse transcriptase polymerase chain reaction. CSMD2 protein level in patients with BC was evaluated via immunohistochemistry. BC cell lines T24 and UMUC-3 were selected for loss-of-function assays targeting CSMD2. Cell viability was determined by CCK8 and clone formation experiments. Cell migration and invasion were evaluated using Transwell assays. Furthermore, the transcriptome of UMUC-3 with CSMD2 knockdown was sequenced to analyze potential signaling network pathways. Finally, the TIMER2.0 database was employed to identify the correlation between CSMD2 and immune cells in the tumor microenvironment. Results CSMD2 expression was up-regulated in BC tissues compared to adjacent tissues. High CSMD2 expression was associated with poor survival and could serve as an independent predictor for survival in patients with BC. Furthermore, down-regulation of CSMD2 notably restrained the viability, migration, and invasion abilities of T24 and UMUC-3 cells. Moreover, transcriptomic sequencing after CSMD2 knockdown in UMUC-3 cells revealed its involvement in the regulation of the malignant phenotype in BC. Finally, public databases suggest a connection between CSMD2 and immune cell infiltration in BC. Conclusions These findings suggest that CSMD2 may promote proliferation and tumorigenicity, and could represent a potential target for improving the prognosis of BC. Keywords: CSMD2, Prognostic marker, Bladder cancer, Biomarker, Immune cells 1. INTRODUCTION In the United States, bladder cancer (BC) is the second most prevalent cancer of the genitourinary system, with an estimated 82,290 new cases and 16,710deaths are estimated in 2023 [[34]1]. Urothelial cell carcinoma accounts for ∼90 % of BC cases, while ∼5 % are squamous cell carcinoma. Other pathological types, such as adenocarcinoma, are rare [[35]2]. Of the new cases, 75 % are classified as non-muscle invasive BC (NMIBC), while ∼25 % of the new cases involve BC that has invaded the bladder muscle layer or deeper, known as muscle-invasive BC (MIBC) [[36]3]. After treatment, 10–15 % of patients with NMIBC progress to MIBC [[37]4]. Despite the proven benefits of neoadjuvant chemotherapy and adjuvant immunotherapy in improving the survival of patients with MIBC, their overall 5-year overall survival (OS) rate following surgery is ∼50 % [[38]5,[39]6,[40]7]. Hence, thereit is imperativeto identify new biomarkers that can pave the way for innovative approaches to diagnose and target BC. CUB and sushi multiple domains 2 (CSMD2), located on the short arm of human chromosome 1 (1p35.1), encodes a single-pass transmembrane protein with a large extracellular domain consisting of CUB and Sushi domain repeats. CSMD2 is highly expressed in the developing and mature brain and has been linked to schizophrenia [[41]8,[42]9]. Previous reports have suggested associations between CSMD2 and the progression of various cancers, including small-cell lung cancer [[43]10], hepatocellular carcinoma [[44]11], colorectal cancer [[45]12], pancreatic cancer [[46]13], and melanoma [[47]14]. In a recent pan-cancer analysis study, it was revealed that CSMD2 is upregulated in most tumors and exhibits moderate to high diagnostic efficiency, with high expression closely associated with poor prognosis in patients with tumors [[48]15]. However, these findings have not been experimentally validated, and the biological functions of CSMD2 in BC remain largely unexplored. Immunotherapy is currently considered an effective adjuvant therapy for cancer treatment, especially for advanced metastatic cancer [[49]16]. CSMD2 has the potential to participate in regulating the complement cascade reaction of the immune system [[50]15], but its role in BC remains unknown. This study aims to provide a preliminary exploration of the role of CSMD2 in BC, including its expression in BC, its predictive value for clinical prognosis, and the relationship between CSMD2 expression and anti-tumor immunity as well as immune evasion in the tumor microenvironment. 2. Materials and methods 2.1. Bioinformatics analysis First, we downloaded RNA-sequencing data for BC from The Cancer. Genome Atlas (TCGA; [51]https://cancergenome.nih.gov) database, which included 412 BCE samples and 19 adjacent tissues. We then employed R packages [[52]17], such as “ggplot” [[53]18], “rms” v6.3-0, and “survival” v3.3.1, to analyze the clinical characteristics and the prognostic value of CSMD2 in patients with BC within the TCGA database. Additionally, we utilized the R package "DESeq2″ v1.26.0 [[54]19] with R v4.2.1 to investigate the correlations between CSMD2 mRNA level and all genes in BC. Subsequently, we conducted gene set enrichment analysis (GSEA) using the “cluster Profiler” package within the MSigDB Collections [[55]20,[56]21]. In this analysis, statistically significant results were defined as having a normalized enrichment score (|NES|) > 1, a false discovery rate (FDR) < 0.25, and an adjusted p-value <0.05. 2.2. Clinical samples The 20 pairs of adjacent and cancerous tissue specimens, which were employed for quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) analysis, were obtained from the First Affiliated Hospital of Nanchang University (China). The collection and utilization of all biological samples in this study were granted approval by the Ethics Committee of the First Affiliated Hospital of Nanchang University (#[2022] CDYFYYLK [11–031]5), and were conducted in compliance with the informed consent of the participating patients. 2.3. Cell culture The human BC cell lines (BIU, EJ, T24, 5637, and UMUC-3) and the non-malignanturothelial cell line (SV-HUC-1) were generously provided by the Institute of Urology at The First Affiliated Hospital of Nanchang University. SV-HUC-1 cells were cultivated in F–12K complete medium; T24 cells were grown in medium composed of DMEM (Gibco, Billings, MT, USA); whereas UMUC3, BIU, and EJ cells were cultured in medium containing MEM (Procell, Wuhan, China). Cell line 5637 was maintained in medium containing 1640 (Gibco). All of these culture media were supplemented with 10 % fetal calf serum, 0.1 mg/mL streptomycin, and 100 U/mL penicillin. The cells were incubated at 37 °C in an atmosphere with 5 % CO[2]. 2.4. Cell transfection All of the siRNA sequences in S1 were custom-designed and synthesized by RiboBio (Guangzhou, China). A day prior to siRNA transfection, 2·10^5 cells were seeded in the culture medium without antibiotics in six-well plates. Following the manufacturer's protocol, once the cell concentration had reached 80 %, the Lipofectamine 2000 kit (Invitrogen, Carlsbad, CA, USA) was utilized for transfection, and the efficiency of gene knockdown was assessed 24–48h later. 2.5. RNA extraction and qRT-PCR Total RNA was extracted using the TRIzol reagent (Invitrogen), and cDNA was synthesized using the PrimeScript RT Reagent Kit (Transgen, Beijing, China). mRNA levels of CSMD2 were assessed using the SYBR Premix Ex Taq system (TaKaRa, Dalian, China), and qRT-PCR analysis was conducted using the ABI Prism 7500 Fast RT-PCR System (Applied Biosystems, Waltham, MA, USA). Relative expression levels were calculated using the comparative (2-^ΔΔCT) method and expressed as fold-change. The expression of β-actin was utilized as an internal control. The specific primer sequences used are provided in [57]Supplementary Table S1. Subsequently, three pairs of BC cell RNAs, which were transfected with the CSMD2-knockdown small interfering RNAs (siRNAs; si-1, si-2,and the negative control si-nc), were extracted and sent to OE Biotech (Shanghai, China) for transcriptome sequencing. 2.6. Western blot assays For the extraction of total protein lysates, RIPA lysis buffer (Beyotime, Nantong, China) containing protease inhibitor was used. Protein concentrations were measured using a BCA Protein Assay Kit (Thermo Fisher Scientific, Waltham, MA, USA). Protein samples were separated by sodium dodecyl sulfate‐polyacrylamide gel electrophoresis. Subsequently, the samples were transferred onto a polyvinylidene fluoride membrane, which was then blocked with 5 % BSA for 1.5 h. The membrane was subsequently probed with primary antibodies, including anti‐CSMD2 (NBP2-92141; Novus, Centennial, CO, USA), β-Tubulin (#10094-1-AP; Proteintech, Wuhan, China), anti‐phospho‐AKT (phosphorylated at Ser473; Abcam, Cambridge, UK), anti‐AKT (#4685; CST, Danvers, MA, USA), anti‐PI3K (#3011S; CST), and anti‐phospho‐PI3K (phosphorylated at Y607; Abcam). The samples were incubated overnight at 4 °C, followed by washing in Tris‐buffered saline buffer with Tween‐20 four times for 8 min. Then, horseradish peroxidase-conjugated secondary antibodies were applied for 1.5 h. After washing the membrane, protein level bands were visualized using an enhanced Electro‐Chemi‐Luminescence kit (Beyotime) on a ChemiDoc MP imaging system (Bio‐Rad, Hercules, CA, USA). The relative quantity of proteins was determined as the ratio of the protein of interest to an internal reference. 2.7. IHC analysis The IHC procedure and scoring for CSMD2 level were performed following previously established methods [[58]22,[59]23]. Briefly, tissue sections affixed to glass slides were first deparaffinized and rehydrated using xylene and then submerged in EDTA antigen retrieval buffer. They were treated with 3 % hydrogen peroxide and subsequently incubated with 1 % bovine serum albumin. Subsequently, they were left to incubate overnight at 4 °C with an anti-CSMD2 antibody (1:200 dilution, BS-8187R; Bioss, Beijing, China). Normal goat serum served as a negative control. After washing with phosphor-buffered saline (PBS)-Tween (PBS + 1 % Tween), a secondary antibody from Zymed (San Francisco, CA, USA) was applied, followed by incubation with the avidin-biotin peroxidase complex (Zymed). The tissue sections were then immersed in 3,3′-diaminobenzidine, counterstained with 10 % Mayer's hematoxylin, dehydrated, and mounted on slides. Quantification of CSMD2 for each sample was determined by a pathologist who was unaware of the clinical and molecular data, using a modified H-score formula: H-SCORE = ∑(pi × i) = (percentage of weak intensity × 1) + (percentage of moderate intensity × 2) + (percentage of strong intensity × 3) [[60]24,[61]25]. 2.8. CCK-8 cell proliferation assays T24 and UMUC-3 cells were seeded in 96-well cell plates at a density of 3000 cells per well and cultured under standard conditions. Cell viability was assessed every 24 h by adding 10 μL CCK-8reagent to each well and incubating them at 37 °C for 2 h. Subsequently, the optical density value at a wavelength of 450 nm was measured and recorded. 2.9. Plate clone formation assays To prepare a cell suspension, exponential T24 and UMUC-3 cells were digested using trypsin. This cell suspension, containing 800 cells per dish, was then transferred to 6-well cell plates, each containing 3 mL pre-culture medium. The cells were cultured at 37 °C with 5 % CO[2] for 1–2 weeks, and the culture process was concluded once visible colonies had formed. After rinsing with PBS, the cells were fixed with 4 % paraformaldehyde for 20 min and stained with 0.1 % crystal violet for 20 min. Finally, the size and number of colonies were evaluated. 2.10. Transwell chambers assays In the cell invasion and migration assays, 8 μm pore-size Transwell inserts were employed. For the invasion assay, following a 24-h transfection period, 2.10^5 cells were resuspended in 200 μL serum-free medium and placed into the upper chamber of Transwell plates, which were pre-coated with Matrigel diluted at 1:8. The lower chamber was filled with 600 μL medium containing 20 % fetal bovine serum. After 24 h incubation, the cells in the lower chamber were collected, fixed, and stained. The experimental steps for the Transwell migration assay are similar to the invasion assays, except that Matrigel is not used in the migration assay. 2.11. Statistical analysis Differential expression analysis was performed using the Wilcoxon signed-rank test in GraphPad Prism v9.2 (GraphPad Software, La Jolla, CA, USA) for statistical computations. For sequencing data and mining of datasets, such as TCGA, automated statistical analysis was performed using statistical packages in the R programming language. 3. RESULTS 3.1. CSMD2 is up-regulated in BC First, we analyzed CSMD2 expression using the TCGA database. As shown in [62]Fig. 1A and B, CSMD2 exhibits significantly higher expression levels in BC tissues in comparison to normal tissues (p < 0.01). Furthermore, the data from our 20 pairs of adjacent and cancerous tissues revealed that the CSMD2 mRNA level ([63]Fig. 1C) and protein expression ([64]Fig. 2A–E) are notably higher in the BC group when compared to the normal group. These findings suggest that CSMD2 is up-regulated in BC, and may be associated with the progression of this condition. Fig. 1. [65]Fig. 1 [66]Open in a new tab CSMD2 mRNA level is upregulated in bladder cancer. (A–B) Analysis of CSMD2 expression in unpaired and paired bladder cancer tissues from the TCGA database. (C) CSMD2 mRNA level in 20 paired bladder tumor tissues and their adjacent normal tissues. (D) Real-time PCR analysis of CSMD2 expression in SV-HCV-1 and bladder cancer cell lines. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001. Fig. 2. [67]Fig. 2 [68]Open in a new tab CSMD2 protein level is upregulated in bladder cancer tissues. (A–D) Representative images of CSMD2 protein level in normal bladder tissues and bladder cancer (upper: magnification × 200; lower: magnification × 400). (C) CSMD2 protein level in 20 paired bladder tumor tissues and their adjacent normal tissues. 3.2. CSMD2 expression in BC cell lines in vitro To assess mRNA and protein levels of CSMD2 in vitro, we selected two BC cell lines, i.e., T24 and UMUC-3, with the normal bladder cell line SV-HUC-1 serving as the control. Real-time PCR analysis indicated that CSMD2 mRNA level was upregulated in BC cells when compared to the control cells ([69]Fig. 1D). We selected the T24 and UMUC-3 cells that exhibited high CSMD2 expression to conduct the knockdown experiments. 3.3. Clinical value of CSMD2 We used a chi-squared test to determine the associations between CSMD2 expression and clinical symptoms in patients with BC. As illustrated in [70]Supplementary Table S2, the expression of CSMD2 exhibited positive correlations with pathologic T stage, pathologic stage, and histologic grade (p < 0.01). Interestingly, univariate analysis revealed that CSMD2 expression, TNM stage, T stage, N stage, M stage, and patient age had statistically significant prognostic value for overall survival (OS; [71]Table 1). Additionally, multivariate analysis demonstrated that CSMD2 expression and N stage are independent prognostic factors for the OS of patients with BC ([72]Table 1). Moreover, we utilized a Kaplan–Meier plot to directly visualize the relationship between CSMD2 expression and OS. As depicted in [73]Fig. 3A, the results suggested that patients with high CSMD2 expression had a poorer OS rate compared to patients with low CSMD2 expression (p = 0.024). These findings indicate that CSMD2 could serve as an independent predictor for patients with BC. Patients with higher CSMD2 expression patients were more likely to have higher pathologic stage ([74]Fig. 3B), higher T stage ([75]Fig. 3C), higher pathologic N stage ([76]Fig. 3D), and higher histologic grade (p < 0.0001; [77]Fig. 3E). Furthermore, we utilized the R packages “rms” and “survival” to construct a nomogram that incorporated pathologic T, N, and M stages, age, gender, and CSMD2 expression of patients with BC into a nomogram to predict patient survival outcomes at 1, 3, and 5 years ([78]Fig. 3F). The nomogram exhibited strong predictive performance for OS rates in BC, and calibration plots confirmed its reliability in predicting model performance ([79]Fig. 3G). Table 1. Univariate and multivariate analysis of prognostic parameters in bladder cancer. Characteristics Univariate analysis __________________________________________________________________ Multivariate analysis __________________________________________________________________ p-value HR 95 % CI p-value HR 95 % CI CSMD2 (low/high) 0.039 1.37 1.02–1.84 0.003 2.191 1.317–3.642 TNM stage (I + II/III + IV) <0.001 2.267 1.567–3.281 0.265 0.512 0.158–1.660 T stage (T1 + T2/T3 + T4) <0.001 2.157 1.485–3.132 0.081 2.602 0.889–7.609 N stage (N0/N1 + N2 + N3) <0.001 2.25 1.649–3.072 0.008 2.065 1.205–3.538 M stage (M0/M1) 0.002 3.112 1.491–6.493 0.648 1.25 0.479–3.262 Age (≤70/>70 years) 0.018 1.424 1.064–1.906 0.45 1.199 0.748–1.922 Gender (female/male) 0.39 0.868 0.629–1.198 [80]Open in a new tab I, II, III, and IV represent different stages. T, size or direct extent of the primary tumor; N, degree of spread to regional lymph nodes; M, presence of distant metastasis; CI, confidence interval; HR, hazard ratio. Fig. 3. [81]Fig. 3 [82]Open in a new tab The clinical value of CSMD2 in bladder cancer. (A) Association between CSMD2 mRNA level and overall survival in patients with bladder cancer in the TCGA database (p < 0.05). (B–E) Associations between CSMD2 expression and pathologic stage, T stage, N stage, and histologic grade in the TCGA database. (F–G) Nomogram depicting the impact of CSMD2 expression on outcomes of patients with bladder cancer and performance calibration graph. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001. 3.4. Silencing CSMD2 inhibits BC progression We used qRT-PCR to detect the efficiency of CSMD2 knockdown in BC cells. Following transfection with the siRNAs si-1 and si-2, the data clearly demonstrated a significant downregulation of CSMD2 mRNA levels when compared to the si-nc groups in T24 and UMUC-3 cells (p < 0.01; [83]Fig. 4A). The results of CCK-8 assays ([84]Fig. 4B and C) indicated that the optical density values in the si-1 and si-2 groups were significantly lower than those in the si-nc group (p < 0.01). In clone formation assays, it was observed that the number of visible colonies in the si-1 and si-2 groups was significantly fewer compared to the si-nc groups in T24 and UMUC-3 cells (p < 0.01; [85]Fig. 4D and E). The findings from invasion and migration assays demonstrated that, in both T24 and UMUC-3 cells, the numbers of cells involved in invasion and migration were significantly reduced in the si-1 and si-2 groups compared to the si-nc group (p < 0.01; [86]Fig. 4F–H). Fig. 4. [87]Fig. 4 [88]Open in a new tab Knockdown of CSMD2 inhibits the tumorigenicity of bladder cancer. (A) Knockdown efficiency of siRNA targeting CSMD2 in the bladder cancer cell lines UMUC-3 and T24. nc, negative control. (B–C) CCK-8 assays reveal significantly reduced cell proliferation in the si-csmd2-1 and si-csmd2-2 groups compared to the si-nc group in bladder cancer cells (**p < 0.01). (D–E) Clone formation assays demonstrate a remarkable decrease in cell clone formation efficiency in the si-csmd2-1 and si-csmd2-2 groups compared to the si-nc group in bladder cancer cells (**p < 0.01). (F–H) Transwell assays indicate that fewer UMUC-3 and T24 cells migrate and invade in the si-csmd2-1 and si-csmd2-2 groups compared to the si-nc group (**p < 0.01). 3.5. CSMD2 expression affects the PI3K/AKT signaling pathway To determine which pathway might be involved in CSMD2-mediated BC progression, we conducted GSEA analysis in the TCGA BC dataset (n = 412). As shown in [89]Fig. 5A, our analysis revealed a correlation between high CSMD2 expression and the PI3K/AKT pathway. The phosphorylation of PI3K and AKT decreased in CSMD2-silenced cells. However, it's noteworthy that the down-‐regulation of CSMD2 did not significantly impact the protein levels of PI3K and AKT ([90]Fig. 5B). These results collectively suggest that the downregulation of CSMD2 is associated with decreased BC cell proliferation and invasive capacities through the regulation of the PI3K/AKT signaling pathway ([91]Fig. 5C). Fig. 5. [92]Fig. 5 [93]Open in a new tab Down‐regulation of CSMD2 and its association with p‐PI3K and p‐AKT. (A) GSEA plot illustrating the positive correlation between CSMD2 expression and PI3K-AKT gene signatures. (B) Western blotting shows that the knockdown of CSMD2 in UMUC-3 and T24 cells significantly reduces p‐PI3K and p‐AKT levels compared to the si‐1, si-2, and si‐NC groups (Full non-adjusted images of WB shown in S4). (C) The downregulation of CSMD2 leads to a decrease in phosphorylation of PI3K and AKT, resulting in reduced proliferation, migration, and invasive abilities of bladder cancer cells. 3.6. Sequencing analysis after knockdown of CSMD2 We extracted total RNA from three groups of UMUC-3 cells: the CSMD2 knockdown group (si-csmd2-1), the negative control group (NC), and a control group with no treatment. Transcriptional profiling analysis was performed using RNA sequencing to gain comprehensive insights into the biological functions of CSMD2 in BC. [94]Supplementary Fig. S3Adisplays a clustered heatmap illustrating differentially expressed genes between the two groups. Furthermore, a volcano plot depicting the distribution of differentially expressed genes between the two groups is shown in [95]Supplementary Fig. S3B. Compared to the control group, the CSMD2 knockdown group exhibited significant upregulation of 615 genes and significant downregulation of 455 genes. The CSMD2 gene appears to be implicated in immune response through multiple pathways and is associated with biological processes such as cell adhesion and growth. These insights were derived from gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses performed on the differentially expressed genes, as indicated in [96]Fig. 6A and B. [97]Fig. 6C shows the protein–protein interaction (PPI) network analysis of differential gene expression between the two groups based on sequencing data. Fig. 6. [98]Fig. 6 [99]Open in a new tab Analysis of CSMD2 knockdown sequencing results. (A–B) GO and KEGG enrichment analysis plots for genes that differ between the two groups. BP, biological process; CC, cellular component; MF, molecular function. (C) PPI network analysis of the 30 most differentially expressed genes from sequencing data. (E) Pathway enrichment analysis comparing CSMD2 knockdown to control cells. NC, negative control. Moreover, according to the GSEA results shown in [100]Fig. 6D, the genes in the NC group are primarily enriched in DNA response, DNA replication, cell cycle, transcriptional misregulation in cancer, and other related signaling pathways. This suggests that CSMD2 may regulate tumor development by modulating DNA alterations and the cell cycle. 3.7. Correlation of CSMD2 with immunomarkers in BC The positive correlation observed with the infiltration of immune evasion-related cells, such as Tregs and cancer-associated fibroblasts ([101]Fig. 7A and B) suggests that CSMD2 may be associated with immune evasion. We extended our analysis by comparing the infiltration of various immune cell subtypes in patients with BC with varying copy numbers of the CSMD2 gene, using the “SCNA” module of the TIMER database. As shown in [102]Fig. 7C, statistically significant differences were noted in the changes in infiltration of CD4^+ T cells, macrophages, neutrophils, and dendritic cells across various copy number states. This implies that the copy number of CSMD2 might influence the infiltration of immune cells. Furthermore, we evaluated the impact of immune cell infiltration on the clinical outcomes of patients with BC using the TIMER platform ([103]http://timer.cistrome.org/). Our findings revealed that lower levels of CD8^+ T cells were associated with poorer prognosis in patients with BC (p < 0.05; [104]Fig. 7D). Fig. 7. [105]Fig. 7 [106]Open in a new tab CSMD2's association with the immune microenvironment. (A–B) Correlations of CSMD2 expression with cancer-associated fibroblasts and Tregs based on XCELL and QUANTISEQ. Correlations are depicted with partial Spearman's correlation, and statistical significance is based on TIMER2.0. (C) Changes in the infiltration of immune subpopulations at each copy number status of CSMD2. (D) Clinical survival outcomes of patients with bladder cancer in the CD8^+ T cells group. 4. Discussion Despite advancements in surgical and anesthesia techniques, as well as the widespread use of perioperative chemotherapy, the long-term survival rate of patients with BC has remained stagnant for decades [[107]6,[108]7]. Simultaneously, through advanced molecular research, our understanding of disease biology has significantly expanded. A recent study on the prognosis of patients with occult hepatitis B virus infection indicated that the development of cirrhosis and subsequent hepatocellular carcinoma may be associated with CSMD2 [[109]11]. Another recent report on a pan-cancer study emphasized that CSMD2 is upregulated in most tumors and demonstrates moderate to high diagnostic efficiency. High expression of CSMD2 is also closely linked to a poor prognosis in cancer patients [[110]15]. However, the additional functions of CSMD2 and its mechanisms in cancers have yet to be fully elucidated. Through our research, we identified abnormal expression of CSMD2 in BC using various online resources, datasets, and tissue samples, as well as cell lines that we collected. By integrating this data with clinical and pathological parameters, we determined that high CSMD2 expression is associated with a poorer prognosis in patients with BC, and that the expression of CSMD2 holds significant diagnostic value. To further validate our bioinformatics analysis results, we used siRNAs to knock down CSMD2 and experimentally confirmed its impact on the proliferation, migration, and invasion of BC cell lines. Additionally, we conducted transcriptomic sequencing to analyze the differentially expressed genes and relevant signaling pathway networks associated with the immune microenvironment in the CSMD2 knockdown group compared to the control group. The PI3K/Akt signaling pathway is an intracellular signaling pathway that plays a pivotal role in various cellular processes, including cell cycle progression, cell proliferation, survival, lifespan, and even cancer. Akt, as the central mediator of the PI3K pathway, is a serine/threonine kinase that plays a crucial role in multiple cellular processes such as cell proliferation, apoptosis, cell migration, glucose metabolism, and transcription [[111]26]. The JAK/STAT signaling pathway is involved in diverse cellular processes, including immune response, cell growth, differentiation, inflammation, and hematopoiesis. Dysregulation of this pathway can contribute to the development of various diseases, making it an important target for therapeutic interventions [[112]27]. In our study, we observed that the knockdown of CSMD2 led to significant alterations in the PI3K/AKT and JAK/STAT pathways ([113]Fig. 5B), suggesting that CSMD2 may primarily regulate tumor functional changes in tumors through the modulation of these two signaling pathways. However, further experimental validation is required to confirm this hypothesis. The tumor microenvironment encompasses the tissues surrounding a tumor, including the extracellular matrix, immune cells, and stromal cells [[114]28]. In recent years, the tumor microenvironment, particularly the immune microenvironment, has received significant attention in research. Within the tumor immune microenvironment, various molecules and cells can influence tumor development and treatment outcomes [[115]29]. CD8^+ T cells play critical roles in immune responses, including cytotoxic activity, immune memory, cytokine production, regulation of other immune cells, and immune modulation, making them vital for anti-tumor immunity. Tregs, on the other hand, exert negative regulation on CD8^+ T cells, contributing to immune tolerance, immune homeostasis, and the prevention of excessive immune responses [[116]30]. In our study, we identified a positive correlation between CSMD2 and Tregs infiltration. Additionally, we observed that patients with low CD8^+ T cell infiltration had worse prognoses. These findings suggest that CSMD2 may regulate Tregs and subsequently impact CD8^+ T cells. We also noted a positive correlation between CSMD2 expression and immune escape-related tumor-associated fibroblasts. Therefore, we hypothesize that CSMD2 may promote tumor progression through immune evasion or immune suppression. However, further experimental validation is needed to confirm these observations. Although we conducted a comprehensive analysis of CSMD2 in BC, there are still certain limitations to be addressed. First, we sourced the transcriptomic data and clinicopathological information from public databases, which introduces data heterogeneity, platform differences, and potential gaps in clinical treatment information. Second, the immune-related mechanisms of CSMD2 in BC are based on bioinformatics predictions and require further experimental validation. In the future, we aim to address these limitations and further validate the bioinformatics predictions. In conclusion, our study demonstrates that CSMD2 is highly expressed in BC and is associated with poor prognosis. In addition, we have shown for the first time that the downregulation of CSMD2 inhibits proliferation, invasion, and migration. These findings suggest that CSMD2 may be involved in the development of BC by influencing cell proliferation, migration, and the immune microenvironment. Our study identifies the pathogenic role of CSMD2 and provides a potential therapeutic target for patients with BC. 5. Consent for publication All authors consent to the publication of this study. Data availability Publicly available datasets were analyzed in this study. These data can be found freely from the TCGA data portal ([117]https://portal.gdc.cancer.gov/). The RNA sequencing data (BioProject accession number PRJNA 1030890) reported in this paper have been deposited into Sequence Read Archive (SRA) database. These data will be available in the date of publication. Funding This study was supported by the National Natural Science Foundation of P.R. China (Grants #81560419, #81960512, and #81760457), Key Project of Natural Science Foundation of Jiangxi Province (#20212ACB206013), and Youth Project of Natural Science Foundation of Jiangxi Province (#20212BAB216037). CRediT authorship contribution statement Zhijun Yao: Writing – original draft. Hailang Yang: Visualization, Software. Xiaoqiang Liu: Investigation, Funding acquisition. Ming Jiang: Visualization. Wen Deng: Visualization, Software, Conceptualization. Bin Fu: Writing – review & editing, Funding acquisition. Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Acknowledgements