Databases used and bioinformatics
To identify differential expressed genes (DEGs), we resort to the cancer genome atlas (TCGA) dataset (http://genome-cancer.ucsc.edu) to screen all genes positively correlated with poor prognosis of RCC. Gene ontology and KEGG pathway enrichment analysis revealed most of these differential expressed genes are associated with cell cycle control. After tumor survival analysis of these hub genes, we found cell-division cycle-associated 5 overexpressed in ccRCC and predicted a poor prognosis in patients.
Patients and tumor specimen
From October 2012 to May 2016, total 137 patients underwent RCC resection at Chinese People’s Liberation Army (PLA) General Hospital were enrolled in this research with median followed-up period 76.93 months (range 4.43-86.06 months). Resected samples were immediately snap-frozen in liquid nitrogen, and then stored at -80°C. Paired tumor tissue and adjacent normal tissue were taken for further analysis. None of them received chemoradiotherapy or molecular targeted therapy prior to nephrectomy. Written informed consent was obtained from all participants. The study was conducted in accordance with protocol approved by the Protection of Human Subjects Committee of Chinese People’s Liberation Army (PLA) General Hospital.
All RCC specimens were staged according to the 2011 Union for International Cancer Control (UICC) TNM classification of malignant tumors. The Fuhrman nuclear grading system was used to determine the nuclear grade.
Antibodies and reagents
The following primary antibodies were applied in this experiment:
Anti-CDCA5(1:2000, ab192237, Abcam), β-tubulin(1:3000, BE0025, Easybio), Akt (11E7, 1:2000, 4685S, CST), phosphate Akt (Ser473, 1:2000, 4685S, CST), Stat3 (D3Z2G, 1:2000, 12640S, CST), Phospho-Stat3 (Tyr705, 1:2000, 9145S, CST), NF-κB (1:1000 BE3154, Easybio), Phospho-NF-κB (1:2000, 3033S, CST), Phospho-mTOR (Ser2448, 1:2000, 5536, CST), P53 (Ser2448, 1:1000, 10442-1-AP, proteintech), Phospho-Histone H2A.X (Ser139, 1:1000, 9718T, CST), Phospho-BRCA1 (Ser1524, 1:1000, 9009, CST), Cyclin B1 (1:1000, 55004-1-AP, proteintech), PARP (46D11, 1:1000, 9532, CST), BCL2 (Ser2448, 1:1000, 12789-1-AP, proteintech), Ki67(D3B5, 1:400, 9129S, CST),
Cell lines
Human normal cell line, HKC, 293T, and RCC cell lines like SN12-PM6, ACHN, Caki-1, A498, OSRC-2, 769-P, and 786-O, were purchased from the National Platform of Experimental Cell Resources for Sci-Tech (Beijing, China). All cell lines were cultured respectively in Dulbecco's Modified Eagle's Medium (DMEM), Minimum Essential Media (MEM), Roswell Park Memorial Institute (RPMI) 1640 medium or McCoy’s 5A (Gibco, ThermoFisher Scientific, Shanghai, China) supplemented with 10% fetal bovine serum (FBS) (EVERY GREEN, TIANHANG biotechnology, Zhejiang, China) and 1% penicillin G sodium/streptomycin with humidified 37°C incubator containing 5% CO2. Cells were authenticated by short tandem repeats (STR) typing and tested to exclude mycoplasma contamination throughout experiments.
Plasmid construction
To generate stable expression of CDCA5 knockdown cell lines, SN12-PM6 and 786-O were infected with lentiviral vector. phU6-MCS-Ubiquitin-EGFP-IRES-puromycin plasmid (GV280, Genechem, China) was used to colon shRNA for CDCA5. Based on the sequence of CDCA5, two interference sequences were designed:
5’-CCGGCCAAAGTACCATAGCCAGTTTCTCGAGAAACTGGCTATGGTACTTTGGTTTTTG-3’ (named KD1), and 5’-CCGGGAGCAGTTTGATCTCCTGGTTCTCGAGAACCAGGAGATCAAACTGCTCTTTTTG-3’ (named KD2).
EcoR I and BamH I restriction sites were introduced. Vector and synthesized interference sequence were double-enzyme linearized by T4 DNA. The lentiviruses were incubated in 293T cells with the pSPAX2 packaging plasmid and VSVG envelope plasmid together with reconstructed plasmid. After 48h and 72h transfection, virus supernatants were filtered through a 0.45 µm syringe filter. Then it was concentrated for immediate use or frozen at −80℃.
Quantitative reverse-transcription polymerase chain reaction (qRT-PCR)
Cells and tissues were lysed with TRIzol Reagent (Invitrogen, USA). Total RNA was extracted using chloroform extraction as well as isopropanol precipitation. Reverse transcription was then conducted by a cDNA synthesis kit (E6560S, New England Biolabs) at 42℃ for 15 min. Subsequent quantitative polymerase chain reaction(qPCR) analyses for each cDNA genes were quantified under specific conditions as follows: Pre-denaturation at 94℃ for 30 seconds; 40 cycles of denaturation (94℃ for 5 seconds), annealing (50℃ for 15 seconds, 72℃ for 10 seconds). PPIA was used as an internal control. They were performed on an ABI 7500 Fast Real-Time PCR System (Applied Biosystems, USA). The data were calculated with the comparative Ct ( ) method. Primers of respective gene are listed in detail (Additional file 1: Table S1).
Cell proliferation assay
Cell proliferation assay was evaluated by crystal violet assay for determining cell viability. We start to seed 786-O and SN12-PM6 cells separately in a 6-well plate. After transfection of lentivirus for 72h, cells were fixed with 4% paraformaldehyde for 20 min and stained with 0.1% crystal violet for 20 min. Wash the cells gently in water, and then dissolve the dye into acetic acid. Measure the optical density of collecting solution at 570nm (OD570) with a plate reader. Calculate the mean value of three independent experiments.
Cell migration assay
In cell migration assay, the Boyden chamber was available for 24-well plates. Inserts were coated with polycarbonate Transwell membrane (Corning, NY) with an pore size. Followed by refilling with 500 high concentration medium (10% FBS) into the 24-well plate, approximately cells were adding into compartments with 150 medium containing 1% FBS. After incubation for 12 hours or overnight, cells remaining on the surface above were gently removed by cotton swab. Then the chamber with migrating cells attached to the bottom surface were fixed with methanol, then stained with 0.1% violet dye. Subsequently, chambers were screened, imaged, and quantified in five random fields per well under an electronic upright microscope. All assays were performed independently three times.
Cell cycle detection
Transfected 786-O and SN12-PM6 cell lines with plasmid for 48hs, collecting cells of both CDCA5 knockdown and negative control groups were washed in PBS and fixed in ice-cold 70% ethanol overnight at -20℃. Then cells were resuspended and treated with propidium iodide (PI) staining solution (P04D17, Gene-Protein Link, China) for 24 h consistent with manufacturer’s instructions. BD FACSCalibur (BectonDickinson, Franklin Lakes, NJ, USA) flow cytometer was used to analyze percentage of the cells in cell cycle phases. Samples was assayed in triplicate and repeated thrice.
Apoptosis analysis
Cell apoptosis determination was analyzed using apoptosis detection kit (P04D03, Gene-Protein Link, China) following manufacturer’s instructions. The treated RCC cells were stained with Annexin V-fluorescein isothiocyanate (FITC) and prodidium iodide (PI) for 15 minutes at room temperature under light-proof condition. Subsequently, cells were evaluated by flow cytometry as mentioned above.
Orthotopic RCC tumorigenicity in nude mice
Animal experiments were carried out following Animal Research Advisory Committee Guidelines of NIH and protocols approved by the Institutional Animal Care and Use Committee of Chinese PLA General Hospital. 18 four-six-week-old male BALB/c nude immunodeficient mice, weighing 15-18 g, were obtained from Vital River Laboratory Animal Technology Co. Ltd (Beijing, China) and bred under a special pathogen-free (SPF) grade laboratory (Cyagen, Heibei, China). After transfected with lentivirus to build stable expression of CDCA5 shRNAs and control shRNA, those cells ( ) were suspended in PBS with Matrigel (Corning, Australia). Mice were anesthetized (Pentobarbital sodium, 30 mg/kg) and then orthopedically implanted into the right subrenal capsule (n=9 in each group). At 28 days post tumor engraftment, mice were euthanized followed by injectable anesthetics. Tumor volume calculations were obtained using the formula: V = , where ‘length’ represents the longest dimension and ‘width’ represents the longest dimension[21]. Tumor size was thereafter measured with standard calipers. Tumor weight, imaging, and histological staining were conducted extracorporeally for further analysis.
Western blot
Total protein from tissues and cells were prepared in RIPA lysis buffer containing protease inhibitor cocktail (#5871, CST). Quantified protein (30 µg) was separated by 8-12% SDS/PAGE gel electrophoresis, and then transferred to 0.45 polyvinylidene fluoride membranes (Direct-Q, Millipore, MA). After blocking the membrane with 5% non-fat milk for 1 hour, they were incubated with respective primary antibody overnight. Afterwards, the membrane was incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies for 1 h and visualized using the ECL system (Thermo Fisher Scientific, Waltham, MA).
Tissue microarray construction and immunohistochemistry of tissue
Seven slides of TMA containing 137 paired formalin-fixed paraffin-embedded (FFPE) ccRCC and adjacent normal tissue samples on each slide was constructed. IHC for CDCA5 expression in TMA was carried out by standard protocol. TMA slides were deparaffinized and rehydrated through a series of graded xylene and ethanol washes. Antigen retrieval was conducted in a 0.01 M citrate buffer for 15 min with microwave heat Induction. Endogenous peroxidase activity was blocked with 3% hydrogen peroxide. Then sides were applied by incubation of rabbit anti-CDCA5 antibody overnight at 1:200, followed with incubation of secondary anti-rabbit antibody (PV-9001, ZSGB-BIO, China). After utilizing horseradish peroxidase (HRP) as the detection system, staining was visualized with chromogen 3,3’-diaminobenzidine (DAB) and hematoxylin (ZIL-9018, ZSGB-BIO, China). Each TMA spot was evaluated independently and blindly by two researchers and assigned a score on its intensity of staining: 0 (no staining, negative), 1 (<10% of malignant cells staining, weak positive), 2 (10%-50% of malignant cells staining, moderate positive), or 3 (>50% of malignant cells staining, strong positive) within carcinomatous areas. Normal mouse lung tissues were used as negative control [22].
Immunofluence analysis with confocal microscope
786-O and SN12-PM6 cells were each cultured in 3 wells of 24-well plate with coverslips. After transfection of lentivirus for 24h and screening by puromycin for another 24h, cells were fixed with 4% paraformaldehyde for 15 min followed by permeabilization with 0.3% Triton X-100 for 10 min on ice. Then, cells were blocked in 3% goat serum albumin-PBS for 30 min. Primary antibody against γH2AX (1:100, 9718s, CST) incubated with cells overnight at 4℃. Cells were then washed and incubated in secondary antibodies Alex Fluor 594 Goat Anti-Rabbit IgG (1:500, A32740, Invitrogen) for 1h at room temperature. Place the coverslip on the fluorescent mounting medium with DAPI (ZLI-9557, ZSGB-BIO, China). Slides were imaged by Leica SP5 confocal microscopy with X100 amplification and analyzed by software.
Statistical analysis.
Data are generally expressed as the mean±standard deviation (SD) from experiments independently performed at least three times. Measurement data between groups were compared by one-way ANOVA, while the chi-square test or Fisher’s exact test were applicable to categorical variables. Comparisons of continuous values or abnormally distributed data were carried out either using Student’s t test or the Mann-Whitney U test. The overall survival analysis was calculated using Kaplan-Meier and further compared by log-rank tests. The cox proportional hazards regression model was applied to identify the risk factors. Value of P <0.05 was indicative of statistical significance. Statistical calculation was performed using SPSS 23.0 (SPSS Inc., Chicago, IL, USA)