Data preparation and processing
Available mRNA sequencing (mRNA-seq) data and miRNA sequencing (miRNA-seq) data from 408 BLCA samples were gained from TCGA database(https://portal.gdc.cancer.gov/). All raw RNA-seq data (miRNAs and mRNAs) were normalized as fragments per kilobase of exon model per million mapped fragment reads. Transformation of miRNA sequences into human mature miRNA names was accomplished using the miRBase database (http://www.mirbase.org).
Screening of DEGs
When performing the differential expression analysis in CEP55high and CEP55low BLCA samples, we determined the differentially expressed genes (DEGs) with thresholds of |logFC| >1 and p < 0.05. Volcano plots of the DERNAs (including DEmiRNAs and DEmRNAs) were visualized using R language (version 4.1.2).
Establishment of the ceRNA network in BC
The ceRNA network was constructed by the following steps: (1) ENCORI (StarBase, http://starbase.sysu.edu.cn/) was used to predict the potential miRNAs targeted by mRNA interaction pairs; (2) TargetScan (version 7.2, http://www.targetscan.org/) were used to forecast the target genes of the DEmiRNAs and build the miRNA-mRNA interaction pairs; (3)The VennDiagram package in R software was utilized to compare the target genes with DEmRNAs, and the target genes that overlapped with DEmRNAs in this study were selected for the next analysis, thus building the ceRNA-miRNA-mRNA triple regulatory network.
The Cytoscape plug-in cytoHubba was performed to identify the hub ceRNA-miRNA-mRNA triple regulatory network. The generated networks were visualized by Cytoscape software (version 3.7.0, https://www.cytoscape.org/).
Cell culture
Three BC-derived cell lines (TCCSUP, T24, and 5637) and a normal bladder cell line (SV-HUC-1) were obtained from American Tissue Type Culture Collection (ATCC, Manassas, VA, USA), while the EJ bladder cancer cell line was obtained from Japanese Cancer Research Resources Bank (JCRB, Tokyo, Japan) and the BIU-87 bladder cancer cell line from China Center for Type Culture Collection (CCTCC, Wuhan, China). The TCCSUP line was cultured in the Dulbecco’s modified Eagle’s medium (DMEM, Gibco, Grand Island, NY) supplemented with 10% fetal bovine serum (FBS, Gibco) and 1% penicillin at 37°C under a humidified atmosphere containing 5% CO2. The 5637, BIU-87, EJ, T24, and SV-HUC-1 lines were cultured in RPMI 1640 medium (Gibco) supplemented with 10% FBS and 1% penicillin at 37°C under a humidified atmosphere containing 5% CO2.
Patients and tumor specimens
Bladder tumor samples and normal adjacent tissues were obtained from biopsy or surgical resection and analyzed in accordance with institutional guidelines on the use of human tissue. All patients provided informed written consent, and the study was approved by the Institute Research Ethics Committee, General Hospital of Southern Theatre Command, China. After biopsy or resection, all tissue samples were immediately frozen in liquid nitrogen and stored at -80°C for subsequent analysis. Samples were histologically confirmed as non-muscle-invasive bladder tumors (NMIBCs, n = 40), muscle-invasive bladder tumors (MIBCs, n = 30), and normal adjacent tissues (n = 70). Of these tumor samples, 35 were classified as low grade and 35 as high grade according to the TNM classification system and WHO criteria. In addition, a total of 59 fresh bladder tumor samples and normal adjacent tissues were used to detect the expression of CEP55 by qRT-PCR (Table 1, Fig. 1A,B).
Table 1
Correlation of the expression of CEP55 with clinicopathologic features.
Clinicopathologic features | n (%) | Relative expression of CEP55 | P-value |
Sex | | | 0.573 |
Male | 50(85) | 5.52 ± 3.55 | |
Female | 9(15) | 4.81 ± 2.67 | |
Age (Year) | | | 0.092 |
≤ 60 | 30(51) | 4.68 ± 3.18 | |
༞60 | 29(49) | 6.18 ± 3.55 | |
Tumor stage | | | 0.001 |
CIS, Ta, T1(NMIBC) | 38(64) | 3.71 ± 2.18 | |
T2-T4(MIBC) | 21(36) | 8.49 ± 3.12 | |
Tumor grade | | | 0.072 |
Low | 35(59) | 4.70 ± 2.80 | |
High | 24(61) | 6.46 ± 4.01 | |
Plasmid construction and transfection
To construct a CEP55 3’UTR overexpression plasmid, the human CEP55 mRNA 3’UTR cDNA sequence was amplified by polymerase chain reaction (PCR) using the forward primer 5'-CCGGAATTCCAAAATAAGTATTTGTTTTGATATTA-3' and reverse primer 5'-ATAAGAATGCGGCCGCTAAAACATTAAATAATTTTATTCT-3'. The amplification product was then digested with BamHI and Hind III, and cloned into the pcDNA3.1(+) plasmid (Invitrogen, Waltham, MA, USA). Human specific CEP55 siRNA, miR-497-5p mimic, miR-497-5p inhibitor, and normal control (NC) sequences were synthesized by RiboBio Biotechnology (RiboBio, Guangzhou, China) (Supplementary Table S1). All constructs were verified by DNA sequencing (Sangon Biotech, Shanghai, China).
For transfection, cells maintained as described at 5×105/well were harvested, reseeded in 6-well plates, and treated with 4 µg cDNA or 50 nM RNA plus Lipofectamine 2000 reagent (Invitrogen) according to the manufacturer’s protocol. The efficiency of transfection was confirmed by quantitative real-time PCR (qRT-PCR) and western blot as described below.
RNA extraction and quantitative real-time PCR
Total RNA was extracted from patient bladder samples or cell lines using the RNA Universal Tissue Kit (Qiagen, Valencia, CA, USA) according to the manufacturer’s instructions. A 2 µg total RNA sample was then reverse transcribed into cDNAs using ImProm-IITM Reverse Transcription System (Promega) according to the manufacturer’s manual. Quantitative RT-PCR for cDNA amplification and quantitation was performed using SYBR Green qPCR SuperMix (Invitrogen) according to the manufacturer’s instructions on an ABI PRISM® 7500 Sequence Detection System (Applied Biosystems, Waltham, MA, USA). The thermocycle was 2 min at 50°C, 2 min at 95°C, and then 40 cycles of 95°C for 15 s and 60°C for 32 s. Primer sequences used for RT-PCR are listed in Supplementary Table S2. Expression levels of CEP55, HMGA2, and PTHLH were calculated using the 2−ΔΔCt method with GAPDH as the internal control, while expression of miR-497-5p was measured with U6 snRNA as the internal control.
Wound healing assays
Cells were seeded into 6-well plates and transiently transfected as described. After 24 h, cells were allowed to grow to 90% confluence in complete medium. Cell monolayers were then wounded with a 200 µL sterile pipette tip and washed several times with phosphate buffered saline (PBS) to remove cell debris. The completed medium was then replaced with serum-free medium and incubation continued. Images of the scratch area (wound) were captured immediately and at 24 h post-scratch under an Olympus microscope (40×). Cells migrating into the wound surface and the average distance of migration were determined at designated time points (0 h and 24 h).
Transwell migration and invasion assays
Cells were transfected for 48 h with the indicated plasmid, harvested, and reseeded with serum-free media in the upper chambers of transwell culture dishes. The transwell inserts (8-µm pore size, Millipore, Burlington, MA, USA) were left untreated for migration assays or coated with Matrigel (BD Biosciences, Franklin Lakes, NJ) for invasion assays. Medium containing 10% FBS was added to the lower chambers as the chemoattractant and plates incubated at 37°C under a 5% CO2 atmosphere for 48 h. The cells that had migrated or invaded through the membrane were fixed with methanol, stained with 0.1% crystal violet, and counted under an inverted microscope (Olympus, Tokyo, Japan).
Flow cytometry analysis of apoptosis
After transfection, cells were grown in serum-free conditions for 48 h. EJ cells stably expressing CEP55 3’-UTR and control cells transfected with empty plasmid were treated with 8 µmol/L cisplatin for 48 h, collected, washed with PBS, and labeled with 10 µL propidium iodide (PI) and 5 µL Annexin V/FITC (BD Biosciences) per 100 µL cell suspension (1×106 cells/mL). Cells were incubated in the dark for 15 min at room temperature and apoptosis assessed using a FACS Calibur system. The total number of apoptotic cells was determined by adding AnnexinV+/PI- and AnnexinV+/PI + cell fractions.
Stable cell lines
EJ cells stably overexpressing CEP55 3’UTR were obtained by transfection with pLVX-mCMV-ZsGreen-puro lentiviral vector containing full-length CEP55 3’UTR cDNA (produced according to the manufacturers' manual) using Lipofectamine 2000 (Invitrogen). Cells were selected by sustained incubation in 1 µg/mL puromycin for 2 weeks. The empty pLVX-mCMV-ZsGreen-puro vector was used to generate control cell lines.
Tumor xenograft model
EJ cells stably expressing CEP55 3’-UTR and control cells transfected with empty vector were collected and injected subcutaneously at 2×106 per 200 µL normal saline into 4-week-old female BALB/c nude mice obtained from the Laboratory Animal Center of Guangdong Medical College. All animal experiments were approved by the Institutional Animal Care and Use Committee of the General Hospital of Southern Theater Command. After 9 days, all mice exhibited palpable tumors. Mice were then randomly divided into two groups receiving either intraperitoneal cisplatin (4 mg/kg body weight in 0.5 mL saline) or equal-volume normal saline every 3 days. Tumor dimensions were measured weekly using a vernier caliper, and the volume calculated according to the formula
$$V=\frac{\pi L{W}^{2}}{6}$$
,
where L is length and W is width. All mice were sacrificed after 33 days and the final tumor weights were recorded.
Luciferase reporter assay
For miRNA target validation studies, HEK 293T cells were seeded in 24-well plates at 2×104 cells/well and transfected with hluct/hRluc luciferase reporter psiCHECK-2 vectors containing the wild-type (WT) or mutant (MUT) miR-497-5p binding sites of the CEP55 gene 3’UTR (psiCHECK-2/CEP55 3’UTR or psiCHECK-2/CEP55 MUT 3’UTR), HMGA2 gene 3’UTR (psiCHECK-2/HMGA2 3’UTR or psiCHECK-2/HMGA2 MUT 3’UTR), or PTHLH gene 3’UTR (psiCHECK-2 /PTHLH 3’UTR or psiCHECK-2 /PTHLH MUT 3’UTR) plus miR-497-5p mimic or control sequence. For the validation of ceRNA interaction, HEK 293T cells were co-transfected with psiCHECK-2 vectors containing the 3’UTR of HMGA2 or PTHLH and miR-497-5p, empty psiCHECK-2 vectors or vectors containing CEP55 3’UTR. Firefly luciferase activity relative to cell number was used to control for transfection efficiency. Forty-eight hours after transfection, luciferase activities were measured with the dual luciferase reporter assay system (Promega, Madison, WI, USA).
Western blot
Cells were lysed in RIPA buffer (50 mM Tris-Cl [pH 7.4], 150 mM NaCl, 1% TritonX-100, 1% sodium deoxycholate, 0.1% SDS, 1 mM EDTA) supplemented with protease and phosphatase inhibitor cocktail (Sigma, St. Louis, MO, USA). Protein concentrations were equalized across samples by measuring the total sample concentration using a BCA Protein Assay Kit (KEYGEN, China) and adding appropriate volumes of RIPA buffer. Total proteins were separated on 12% SDS-polyacrylamide gels and transferred onto PVDF membranes. Membranes were blocked with 5% skim milk for 1 h at room temperature and probed with the primary antibodies against the following proteins overnight at 4°C: CEP55 (1:100, sc-374051, Santa Cruz, Dallas TX, USA), PTHLH (1:200, sc-9680, Santa Cruz), HMGA2 (1:100, 8179, Cell Signaling Technology, Danvers, MA, USA), TWIST (1:200, sc-15393, Santa Cruz,), E-cadherin (1:500, AF0131, Affbiotech, Cincinnati, OH, USA), Vimentin (1:500, AF7013, Affbiotech), Snail (1:500, AF6032, Affbiotech), ERK1/2 (1:200, sc-135900), p-ERK1/2 (Thr202/Tyr204) (1:200, Santa Cruz, sc-16982), MEK1/2 (1:200, sc-436, Santa Cruz),, p-MEK1/2 (Ser217/221) (1:1000, 9154P, Cell Signaling Technology), P38 (1:1000, 8690, Cell Signaling Technology), p-P38(Thy180/Tyr182) (1:200, sc-17852-R, Santa Cruz), and NF-κB p65 (Bioworld Technology, St. Louis Park, MN, USA). After primary antibody incubation, blots were washed three times with Tris-buffered saline containing 0.1% Triton X (TBST), incubated with appropriate secondary antibodies for 1 h at room temperature, and washed three times in TBST. Immunolabeling was visualized using Electro-Chemi-Luminescence (ECL) reagent.
Cell proliferation and colony forming assays
The MTT (3, 4, 5-dimethylthiazol-2, 5 biphenyl tetrazolium bromide, Invitrogen) cell viability assay was used to estimate proliferation rates in culture following the indicated treatments. After 48 h of transfection, cells were seeded on 96-well plates at 4 × 103 cells/well for 0, 24, 48, 72, and 96 h. Then, 20 µL MTT solution (5 mg/mL in Dulbecco’s PBS) was added to each well and incubated for 4 h at 37°C. The culture medium was carefully removed and 200 µL of dimethyl sulfoxide added to dissolve the formazan generated by viable cells. Cell number was estimated by measuring the absorbance at 570 nm using a spectrophotometer. In other experiments, the MTT assay was conducted following treatment of 5× 103 cells/well with cisplatin (0, 10, 20, 30, 40, and 50 µmol/L).
For colony-forming assays, cells were transfected for 48 h as described and seeded on 6-cm plates at 300 cells/well for 10 days at 37°C under a 5% CO2. Then, the culture medium was carefully removed and remaining cells fixed with methanol and stained with 0.1% crystal violet. Culture wells were photographed under an inverted microscope and the number of colonies counted. Three independent experiments were performed with triplicate treatments.
Morphological assessment for endothelial–mesenchymal transition
Cells were seeded into 6-well plates, transiently transfected as described above, and incubated for 48 h, a time sufficient to allow EMT. Plates were washed with fresh medium to remove dead cells and examined at 100× magnification under a phase contrast microscope. Cells presenting mesenchymal characteristics, with a more fibroblast-like and long shuttle morphology, were considered as having undergone EMT.
Immunofluorescence staining
Cells were grown on glass chamber slides, fixed with 4% paraformaldehyde in PBS for 30 min, permeabilized in 0.1% Triton X-100 for 30 min, blocked with 3% bovine serum albumin in PBS for 1 h at room temperature, and then incubated sequentially with rabbit anti-human E-cadherin (AF0131, Affbiotech) and anti-vimentin antibody (AF7013, Affbiotech) overnight at 4°C and FITC-conjugated secondary antibody (Bioworld, Atlanta, Georgia, USA) for 1 h. Cells were washed with PBS between each step, then examined by laser scanning confocal microscopy.
In situ hybridization
For detection of miRNA expression in situ hybridization, 4-µm thick paraffin-embedded sections were mounted on tissue microarray (TMA) slides, incubated at 60°C for 1 h, deparaffinized in xylene, rehydrated with graded alcohol, washed three times with RNase-free PBS, digested with 8 mg/mL pepsin at 3°C for 10 min, washed, and then dehydrated in graded alcohol. Tissues were then hybridized with 50 nm locked nucleic acid (LNA)-modified DIG-labeled probes for miR-497-5p (Exiqon) at 40°C overnight. After stringency washes (5×, 1×, 0.2 × SSC), sections were incubated in blocking buffer for 30 min at room temperature and then overnight at 4°C in alkaline phosphatase-conjugated anti-DIG Fab fragment solution. Antibody signals were colorized with NBT and BCIP substrate and nuclei counterstained with Nuclear Fast Red.
Immunohistochemical staining
Immunohistochemistry was conducted on 5-mm formalin-fixed, paraffin-embedded tissue sections from BC patients (n = 70; General Hospital of Southern Theater Command, China) using antibodies against CEP55 (1:200, ab214302, Abcam, Cambridge, MA, USA) and HMGA2 (1:100, 8179, Cell Signaling Technology). Immunostaining was performed using the ChemMateTMDAKO EnVisionTM Detection Kit (DakoCytomation, Glostrup, Denmark). Subsequently, sections were counterstained with hematoxylin (Zymed Laboratories, South San Francisco, CA, USA) and mounted in nonaqueous mounting medium. The primary antibody was omitted for negative controls.
Immunohistochemical scoring
Immunohistochemical staining was assessed according to an immunoreactive score (IRS) that considered both staining intensity and the proportion of positive cells. Staining intensity was graded as 0 (no staining), 1 (weak), 2 (moderate), or 3 (strong), and the proportion of positive cells as 0 (negative), 1 (< 10%), 2 (10–50%), or 3 (> 50%). Intensity and positivity scores were multiplied to yield the IRS. An IRS ≥ 3 were defined as cytoplasmic expression positive and < 3 as negative.
Statistical analysis
All data were analyzed using SPSS (version 20.0). Results are presented as mean ± standard deviation (SD) of at least three independent experiments. Two group means were compared by two-sided Student’s t-test and more than two group means by analysis of variance (ANOVA) with Bonferroni correction for pair-wise comparisons. P < 0.05 was considered significant for all tests.