2.1. Cell lines and cell culture
The malignant human invasive bladder cancer cell line YTS-1 and the normal human bladder mucosal epithelial cell line HCV29 were obtained from Dr. Sen-itiroh Hakomori (The Biomembrane Institute, Seattle, WA, USA). Both cell lines were grown in RPMI 1640 medium (HyClone, Provo, UT, USA) supplemented with 10% fetal bovine serum (FBS; Biological Industries, Beit Haemek, Israel) at 37°C in a 5% CO2 atmosphere.
2.2. sEV isolation
sEVs were isolated using differential ultracentrifugation. Briefly, when bladder cells reached 80% confluence in a 15-cm Petri dish after approximately 48 h, the growth medium was replaced with a sEV-depleted FBS medium. After 48 h of incubation, culture supernatants were collected and underwent a series of centrifugation steps: 300 × g for 10 min to pellet cellular debris, 2,000 × g for 10 min to pellet apoptotic bodies and microparticles, 10,000 × g for 30 min, and 100,000 × g for 70 min at 4°C. The resulting pellet was resuspended in 100 µL phosphate-buffered saline (PBS). The resulting supernatant was then centrifuged at 10,000 × g for 15 min at room temperature (RT; 25°C) and filtered through a 0.22 µm-diameter membrane. The final sEV pellet was stored at − 80°C until use.
2.3. Transmission electron microscopy (TEM)
Purified sEV samples were pipetted onto carbon-coated 400 mesh grids, incubated for 5 min, and washed twice with PBS. Uranyl acetate (2%) was pipetted onto the copper mesh and incubated for 30 s. Images were acquired using a transmission electron microscope (model H-7650; Hitachi, Tokyo, Japan) at 80 kV.
2.4. Nanoparticle tracking analysis
sEVs were loaded into a NanoSight LM10 instrument (Malvern Panalytical, Malvern, UK), and particles tracked for 60 s using the NanoSight nanoparticle tracking analysis software.
2.5. Cellular sEV uptake
When cells reached 80% confluence in a well, the ExoTracker probe was incubated with sEVs at a 1:500 volume ratio at 25°C for at least 40 min in the dark. After incubation, unlabeled sEVs were centrifuged at 1,000 × g for 5 min at 4°C through a 10 KD filter membrane, with this process repeated thrice. The probes containing sEVs were then incubated with cells for 1 h. After trypsin digestion, the cells were collected, centrifuged at 1,000 × g for 5 min at room temperature (RT; 25°C), washed with PBS, suspended in 500 µL PBS, and analyzed using flow cytometry.
2.6. Total protein extraction
Cells were detached using trypsin, washed three times with cold PBS, and lysed in RIPA buffer (50 mM Tris, pH 7.2, 1% Triton X-100, 0.5% sodium deoxycholate, 0.1% SDS, 150 mM NaCl, 10 mM MgCl2, and 5% glycerol) containing protease inhibitors. The lysate was centrifuged at 14,000 × g for 15 min at 4°C, and the supernatant was collected. Protein concentration was determined using the bicinchoninic acid assay (Beyotime Biotechnology, Jiangsu, China), following the manufacturer’s instructions.
2.7. Western blotting
Proteins were separated using sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred onto polyvinylidene difluoride membranes (Bio-Rad, Hercules, CA, USA). Membranes were blocked with 5% skim milk in TBST (20 mmol/L Tris-HCl, 150 mmol/L NaCl, 0.05% Tween 20, pH 8.0) for 1 h at 37°C, probed with primary antibodies overnight at 4°C, and incubated with the appropriate HRP-conjugated secondary antibodies (Beyotime Biotechnology, Jiangsu, China). The bands were visualized using enhanced chemiluminescence (Vazyme Biotech, Nanjing, China) and photographed using a gel documentation system (Tanon Science & Technology Co., Shanghai, China).
Antibodies used in this study CTNND1 (A11399) was from ABclonal Technology (ABclonal, Wuhan, China). AKT (4685s), phosphorylated-AKT (4060s), erk (4696s), phosphorylated-erk (4370s), Alix (2171s), Calnexin (2679s), STAT3 (9139s) and phosphorylated-STAT3 (p-STAT3) (9145s) were from Cell Signaling Technology (Beverly, MA, USA). E-Cadherin (sc-8426) and N-Cadherin (sc-59987) were from Santa Cruz Biotechnology (Santa Cruz, CA, USA). GAPDH (G-9545) was from Sigma-Aldrich (St. Louis, MO, USA). CD63 (ab134045) and TSG101 (ab125011) were from Abcam (Cambridge, MA, USA).
2.8. CTNND1 knockdown
A short hairpin RNA (shRNA) targeting CTNND1 was subcloned into a lentivirus vector (pLKO-puro vector; Addgene, Cambridge, MA, USA) to construct the sh-CTNND1 vector, and shRNA sequences were listed. Lentiviral particles were generated in HEK293T cells using a packaging system with two assistant vectors (pMD2.G and psPAX2) with PEI MAX 40000 (Polysciences, Warrington, PA, USA). These lentiviral particles were collected from the supernatant after 48 h and used to infect YTS-1 cells. Stably transfected cells were selected using puromycin.
List of shRNA primers
Primer name | Primer sequences (5′-3′) |
shRNA1-F | CCGGTCACCGGAGGTGCCATCGGATCAATACGA ATATTGATCCGATGGCACCTCC |
shRNA1-R | AATTCAAAAGGAGGTGCCATCGGATCAATATTC GTATTGATCCGATGGCACCTCC |
shRNA2-F | CCGGTCACCGCATGAGCGAGGAAGTTTAGCCGA AGCTAAACTTCCTCGCTCATGC |
shRNA2-R | AATTCAAAAGCATGAGCGAGGAAGTTTAGCTTC GGCTAAACTTCCTCGCTCATGC |
2.9. CTNND1 overexpression
CTNND1 was amplified from the cDNA of the YTS-1 cells and was cloned into a pLVX-IRES-Hyg lentiviral vector (Takara, Shiga, Japan). Lentiviruses were packaged in HEK293T cells and collected. HCV29 cells were infected with these lentiviruses, and stable transfectants were selected using puromycin.
2.10. Cell apoptosis
Cells were stained with annexin V-conjugated FITC and 7-AAD-conjugated APC (BioLegend, San Diego, CA, USA) for 15 min at RT in annexin V binding buffer. Flow cytometry (ACEA Biosciences, San Diego, CA, USA) was used to quantify cells in early (annexin V-positive) and late apoptosis (annexin V-positive and 7-AAD-positive).
2.11. Cell cycle detection
Cells were fixed with 70% ethanol, stained with propidium iodide (BioLegend), and treated with RNase (CoWin, Suzhou, China) in PBS. A flow cytometer (ACEA Biosciences) was employed to evaluate the cell cycle.
2.12. Cell proliferation assay
For the Cell Counting kit-8 (CCK-8) assay, 1500 cells were plated in a 96-well plate and incubated with 10 µL CCK-8 solution (Beyotime Biotechnology) for 4 h. Absorbance at 450 nm was measured using a microplate reader. For the EdU cell proliferation assay, cells were incubated with 10 µM EdU for 12 h, fixed with 2% paraformaldehyde, permeabilized with 0.2% Triton X-100, stained with iClick EdU solution (Genecopoeia, Rockville, MD, USA) for 30 min, rinsed with 1x iClick permeabilization wash reagent, and finally evaluated using flow cytometry.
2.13. Transwell assay
Transwell chambers (24-mm diameter, 8-µm pore size; Corning Life Sciences, Corning, NY, USA) were used for this assay. Cells at a density of 2 × 104 were seeded into the upper chamber and starved overnight in a serum-free medium. Complete medium was added to the bottom chamber, and cells were grown for 24 h. Migrated cells were fixed with 4% paraformaldehyde(Sigma-Aldrich, St. Louis, MO, USA)for 15 min, stained with 0.1% crystal violet for 15 min, and photographed under a microscope ༈Ningbo Shunyu Optical Technology, Ningbo, China༉.
2.14. Wound-healing assay
The wound-healing assay was performed as previously described (Tan et al. 2020). Briefly, confluent cells in 6-wells plates were pretreated with 0.4 µg/mL mitomycin C (Sigma-Aldrich) for 30 min and scratched with a pipette tip. Cells were then rinsed with PBS, placed in RPMI 1640 medium supplemented with 0.04 µg/mL mitomycin C, and photographed under a microscope after 24 h. Wound tracks were marked, and relative migration distances were calculated using Image Pro Plus software (Media Cybernetics, Silver Spring, MD, USA).
2.15. Statistical analyses
All experiments were independently repeated at least three times. Results are presented as the mean ± standard deviation (SD). A normality test was applied, and Student's t-test was used to assess differences between the two groups. Statistical analyses were conducted using GraphPad Prism software (GraphPad Software, San Diego, CA, USA). Differences were considered statistically significant at p < 0.05.