2.1 PMEPA1 is highly expressed in bladder cancer and correlates with disease progression
Analysis of publicly available RNA-seq data from TCGA-BLCA (Bladder Urothelial Carcinoma) using thresholds of |Log2FC| > 1 and adjusted p-value < 0.05 reveals that 3825 genes are significantly upregulated and 2495 genes are downregulated in tumor tissues compared to adjacent non-cancerous tissues (Figure 1A). Specifically, PMEPA1 is significantly upregulated in tumor tissues compared to adjacent non-cancerous tissues, both in paired and unpaired samples (Figure 1B, C). Analysis of PMEPA1 expression across different clinical stages shows a significant increase from T2 to T3 stages, while other stages exhibit an increasing trend without statistical significance (Figure 1D). Furthermore, high expression of PMEPA1 is associated with poor prognosis in bladder cancer (Figure 1E).
Previous studies have shown that PMEPA1 can promote the progression of various tumors. To further elucidate the expression of PMEPA1 in bladder cancer, we conducted RT-qPCR to measure the mRNA levels of PMEPA1 in cell lines. Compared to SV-HUC-1 cells, PMEPA1 is significantly upregulated in 5637 cells (Figure 2A).
2.2 PMEPA1 Promotes Proliferation, Migration, and Inhibits Apoptosis of Bladder Cancer Cells
Given the significant impact of PMEPA1 on the prognosis of bladder cancer patients, we aimed to understand its involvement in regulating biological behaviors and its potential as a novel target for clinical therapy. To investigate the effect of PMEPA1 on bladder cancer cells, we transfected 5637 cells with siRNA targeting PMEPA1 and measured PMEPA1 expression levels after 24 hours.The RT-qPCR results show that compared to the NC group, siPMEPA1 significantly reduces the mRNA expression levels of the PMEPA1 gene (Figure 2B). Western blot results indicate that compared to the NC group, the expression level of PMEPA1 protein is also reduced in the siPMEPA1 group (**P < 0.01) (Figure 2C, D).Cell viability was assessed using the CCK-8 assay, and the results indicate that knocking down pmepa1 significantly inhibits the growth of 5637 cells compared to the siNC group (Figure 2E).In wound healing experiments, it was observed that knocking down pmepa1 significantly reduces the migration ability of 5637 cells (Figure 2F, G).Additionally, we used flow cytometry to assess the impact of knocking down PMEPA1 on apoptosis in 5637 cells. Compared to the NC group, knocking down PMEPA1 significantly increases apoptosis in 5637 cells (Figure 2H, I).
2.3 hsa-mir-1323 targets PMEPA1 in bladder cancer.
MicroRNAs (miRNAs) are small endogenous RNAs that regulate gene expression post-transcriptionally by binding to the 3' untranslated region (UTR) of target genes. They are important regulators of gene expression [23] .To study the upstream regulation of PMEPA1 by microRNAs, we utilized a miRNA binding prediction database established using four different methods. This approach identified 8 potential microRNAs that may interact with PMEPA1 (Figure 3A).
We conducted correlation analysis of PMEPA1 expression and the expression of 8 potential interacting microRNAs in bladder cancer samples from the TCGA-BLCA dataset. Among them, miR-421, miR-1323, and miR-760 showed a significant negative correlation with PMEPA1 expression (p < 0.001) (Figure 3B, Supplementary Figure 1). Considering the expression levels of miRNAs in tumor tissue and the context++ score from the TargetScan_8.0 database, we selected hsa-miR-1323 for further investigation (Supplementary Table 1).
To confirm whether hsa-miR-1323 directly targets PMEPA1 through its 3' untranslated region (UTR) to regulate cell activity, we conducted dual-luciferase reporter assays in 293T cells. After co-transfection with hsa-mir-1323 mimics, the luciferase activity in cells containing PMEPA1-3'UTR-WT was reduced by 33% compared to the control group (P < 0.01). When the PMEPA1-3'UTR region was mutated (PMEPA1-3'UTR-MUT), the luciferase activity in these cells increased by 56.7% compared to PMEPA1-3'UTR-WT (P < 0.01) (Figure 3C). These results demonstrate that miR-1323 directly targets PMEPA1 through its 3'UTR.
Additionally, transfection of miR-1323 mimics into human bladder cancer cells 5637 resulted in significantly downregulated protein expression of PMEPA1 compared to the control group and NC (negative control) group. This further demonstrates that miR-1323 can influence the protein expression levels of PMEPA1 in bladder cancer (Figure 3D-E).
2.4 hsa-miR-1323 inhibits bladder cancer cell proliferation and migration, and promotes apoptosis by targeting PMEPA1.
Cell viability was assessed using the CCK-8 assay, and the results indicate that compared to the control group, inhibition of hsa-miR-1323 significantly enhances the activity of 5637 cells (Figure 4A). Concurrently inhibiting hsa-miR-1323 while knocking down PMEPA1 reduces the enhanced cell activity caused by hsa-miR-1323 inhibition.
In wound healing experiments, it was observed that inhibition of hsa-miR-1323 significantly enhances the migration ability of 5637 cells compared to the control group (Figure 4B). Concurrently inhibiting hsa-miR-1323 while knocking down PMEPA1 reduces the enhanced cell migration ability caused by hsa-miR-1323 inhibition (Figure 4C).
Additionally, we used flow cytometry to assess the effect of inhibiting hsa-miR-1323 on apoptosis in 5637 cells. As shown in the figures, compared to the NC group, inhibition of hsa-miR-1323 significantly reduces apoptosis in 5637 cells (Figure 4D-E). Concurrently inhibiting hsa-miR-1323 while knocking down PMEPA1 partially rescues the apoptosis induced by hsa-miR-1323 inhibition (Figure 4D-E).
2.5 hsa-miR-1323 activates the Smads-dependent TGF-β signaling pathway by inhibiting PMEPA1
To further explore the specific signaling pathways through which PMEPA1 functions in bladder cancer, we conducted differential gene enrichment analysis using KEGG and GO terms on TCGA-BLCA data. The results indicate that the TGF-β signaling pathway is the most likely pathway PMEPA1 is involved in (Figure 5A-B). Protein-protein interaction (PPI) network analysis using the STRING database reveals interactions between PMEPA1 and SMAD2, SMAD3 (Figure 5C). Additionally, Gene Set Enrichment Analysis (GSEA) indicates that the pathway "PATHWAY RESTRICTED SMAD PROTEIN PHOSPHORYLATION" is upregulated in bladder cancer patients with high PMEPA1 expression, showing significant differences (P < 0.01) (Figure 5D).
Additionally, previous studies have shown that PMEPA1 can promote bone metastasis in prostate cancer by blocking TGF-β signaling [24]. PMEPA1 can also interfere with the phosphorylation of R-Smad proteins to inhibit activation of the TGF-β signaling pathway[25].
Combining the above information, PMEPA1 likely functions in bladder cancer by regulating the TGF-β signaling pathway.
In our experiments, we explored how hsa-miR-1323 and PMEPA1 influence the TGF-β signaling pathway. Upon inhibiting hsa-miR-1323, we observed a significant decrease in the protein expression levels of Smad2, Smad3, p-Smad2, and p-Smad3 compared to the control group. Conversely, when simultaneously inhibiting hsa-miR-1323 and knocking down PMEPA1, the protein expression levels of Smad2, Smad3, p-Smad2, and p-Smad3 were significantly elevated compared to the control group (Figure 5E-F).
In summary, in bladder cancer, PMEPA1 exerts an inhibitory effect on the Smad-dependent TGF-β signaling pathway, while hsa-miR-1323 can relieve this inhibition by affecting the expression of PMEPA1.