Cell lines and cell culture. Human breast cancer cell lines (MDA-MB-231, MDA-MB-468, HCC-70, MCF-7, T47D, SK-BR-3) and HEK293T cells were originally from the American Type Culture Collection. Luciferase-labeled LM2-4175, a lung metastatic derivative of MDA-MB-231, was kindly provided by Prof. Guo-Hong Hu (University of Chinese Academy of Sciences, Shanghai, China). These human cell lines were cultured in DMEM (Thermo Fisher Scientific) containing 10% fetal bovine serum (Thermo Fisher Scientific) and 1% penicillin/streptomycin (Thermo Fisher Scientific). Two immortalized human mammary epithelial cell lines, HMEC and MCF-10A, were cultured in DMEM/F12 medium supplemented with 5% horse serum, 20 ng/mL epidermal growth factor, 10 µg/mL insulin, 100 ng/mL cholera toxin and 0.5 µg/mL hydrocortis.
Quantitative reverse transcription-PCR (qRT‒PCR). Total RNA was isolated from cell lines using the RNeasy Plus Mini Kit (QIAGEN) and subjected to reverse transcription to cDNA using the PrimeScript RT Reagent Kit with gDNA Eraser (TAKARA). Quantitative PCR was carried out using SYBR Premix Ex Taq (TAKARA) on an ABI 7900HT PCR system (Applied Biosystems). The raw data were processed with SDS v2.1 software in a relative quantification model (-ΔΔC(T)).
Western blot. Immunodetection of each specific protein was carried out according to a standard protocol. Briefly, cultured cells were scraped with lysis buffer (1% SDS, 50 mM Tris-HCl, pH 8.0, 50 mM DTT, protease inhibitor cocktail (Beyotime, China), phosphatase inhibitor cocktail (Roche)) and separated by 8–12% SDS‒PAGE and blotted onto 0.22 µm or 0.45 µm PVDF membranes. The membrane was blocked with 5% nonfat milk in TBS-T buffer and then incubated with specific primary antibody 1:1000 or other dilutions in QuickBlock Primary Antibody Dilution Buffer (Beyotime Biotechnology, China), followed by corresponding HRP-conjugated secondary antibody. After development with SuperSignal West Femto Chemiluminescent Substrate (Thermo Fisher Scientific), images were obtained with a LAS-4000 mini instrument. The following antibodies were used: anti-SNRPE (Proteintech Group, #20407-1-AP), anti-DDX20 (Proteintech Group, #11324-1-AP), anti-ETV3 (Abcam, #ab176717), anti-ACTB (CWbio, China, #CW0098), anti-GAPDH (CWbio, China, # CW0100), anti-Flag (CWbio, China, # CW0287), anti-4E-BP1 (CST, #9452), anti-phospho-4E-BP1 (Ser65) (CST, #9456), and anti-mTOR (7C10) (CST, #2983).
3' rapid amplification of cDNA ends (3’-RACE). The 3’-RACE assay was performed using the SMARTer RACE cDNA Amplification Kit (Clontech Laboratories). Briefly, total RNA from MDA-MB-231 cells was extracted with a RNeasy Mini Kit (QIAGEN), and genomic DNA contamination was removed by on-column RNase-free deoxyribonuclease I (New England Biolabs, USA). The 3'-RACE-ready cDNA of MDA-MB-231 cells was amplified with a SNPRE gene-specific primer and a nested primer GSP with corresponding special oligo (dT) primers. The products were cloned into the pGEM-T easy vector (Promega) for Sanger sequencing.
SNRPE overexpression construct. The CDS of the full-length SNRPE gene (NM_003094) with the Flag sequence and Kozak tag attached at the N-terminal end was synthesized by the Synbio-tech company (Suzhou, China) and cloned into the pcDNA3.1(+) vector at the NheI/BamHI sites. pcDNA3.1-Flag-HA (Addgene #52535) was used as a negative control.
siRNA transfection. Small interfering RNA (siRNA) duplexes with a 3’-terminal dTdT overhang that target human DDX20 or SNRPE mRNA were designed using BLOCK-iT RNAi Designer software (Thermo Fisher Scientific) and synthesized by RiboBio Co., Ltd. (Gungzhou, China). MDA-MB-231 cells were placed in 6-well plates and transfected with 50 nM siRNAs on the following day using Lipofectamine RNAiMAX Reagent (Thermo Fisher Scientific) following the manufacturer's instructions. At 48 h after transfection, the cells were subjected to immunoblotting or other assays.
shRNA plasmid construction. For shRNA vector construction, two validated sequences with high knockdown efficiency that target human SNPRE mRNA were chosen from the Broad TRC lentiviral shRNA library. The annealed synthesized oligonucleotides were ligated into the pLKO.1 plasmid using EcoRI/AgeI restriction enzymes. A human genome nontargeting sequence served as a negative control, and its target sequence was 5’-CAACAAGATGAAGAGCACCAA-3’.
Construction of tetracycline-activated (tet-on) inducible plasmid. The SNRPE shRNA pLKO.1 #1 target sequence was subcloned into a GV307 vector (a pTRIPZ-derived vector, Genechem, China) by digestion with XhoI and EcoRI restriction enzymes and ligation with T4 DNA ligase (New England Biolabs, USA).
Lentivirus packaging and cell infection. For lentivirus particle production, 1.5×106 293FT cells were cultured in 10-cm dishes overnight and cotransfected with 6.0 µg recombinant vector (shRNA or tet-on shRNA) and 1.5 µg pMD2. G and 4.5 µg pspAX2 using Lipofectamine 2000. At 48 h posttransfection, virus-containing supernatant was collected, filtered (0.45 µm filter), aliquoted and stored at -80 ℃ until use. The cells were infected with virus supernatant with 6 µg/ml polybrene for 8 h and selected with puromycin for more than 3 days.
SNRPE inducible knockdown cells. MDA-MB-231 or LM2-4175 cells were infected with shSNRPE tet-on lentivirus, followed by 1.0 µg/ml puromycin (Thermo Fisher Scientific) selection in DMEM containing 10% tetracycline-free FBS for 3 days. To induce RNA interference with SNPRE, gradient concentrations of doxycycline (Sigma) were added to the culture medium for 3 days. Fluorescence microscopy observes TurboRFP fluorescence to verify that the induction is working. Anti-SNPRE immunoblotting to optimize the optimal induction concentration of doxycycline. The puromycin-resistant cells were expanded for subsequent in vivo assays.
Cell proliferation assay. A total of 1 x 103 or 2 x 103 cells in 150 µL DMEM were seeded in 96-well plates and imaged using the IncuCyte ZOOM system (Essen BioScience). Frames were captured at 12 h or 24 h intervals from four separate regions per well, and the growth rate was determined using IncuCyte software (2013A Rev2).
SILAC-based identification of interacting partners for SNRPE. 293T cells were cultured in SILAC DMEM (Thermo Fisher Scientific) (supplemented with 10% dialyzed FBS), where half of the 293T cells were labeled with heavy media containing 42 mg/L lysine (U-13C6, 99%; U-15N2, 99%) (Lys8) and 73 mg/L arginine (U-13C6, 99%; U-15N4, 99%) (Arg10) and the other half were labeled with light media containing unlabeled amino acids purchased from Sigma‒Aldrich. Then, 200 mg/L proline (Sigma‒Aldrich) was added to prevent the arginine to proline conversion. After seven passages in the corresponding media, the heavy or light amino acid-labeled cells were transfected with pcDNA3.1-Flag-SNRPE plasmid or control plasmid, respectively, using Lipofectamine 2000 according to the instruction manuals. Two days after transfection, 293T cells were washed with prechilled PBS and scraped with RIPA buffer (Beyotime Biotechnology, China). The lysate from heavy- and light-labeled cells was immunoprecipitated by using anti-Flag magnetic beads (Thermo Fisher Scientific). The beads were washed three times with RIPA buffer and eluted with buffer containing 200 mM Tris-HCl (pH 7.9), 0.2 mg/ml Flag peptide (Sigma), and protease inhibitor cocktail. The eluted proteins were denatured and separated with 10% SDS‒PAGE. A few elutes were used for silver staining analysis. The majority of the eluates were mixed at a 1:1 ratio and stained with Coomassie Brilliant Blue. The combined mixture gel lanes were excised, and in-gel trypsin digestion was performed. The peptides were collected, and LC‒MS/MS was performed on an LTQ-Orbitrap XL mass spectrometer (Thermo Fisher Scientific). The resulting raw data files were analyzed with MaxQuant software against the SWISSPROT database for humans. The peptide/protein ratios were calculated using the SILAC function module.
DDX20 knockout stable cell clones. For genetic knockout of DDX20, two sgRNAs localized in DDX20 exon 1 were independently cloned into BbsI-digested px459 v2.0 (Addgene #62988). Eight micrograms of each sgRNA recombinant vector was then transiently transfected into 1.5 x 106 MDA-MB-231 cells seeded in 10-cm dishes using reduced serum media (Opti-MEM) and Lipofectamine 2000 (Life Technologies). At 24 h posttransfection, 1.5 µg/mL puromycin was added to the cells for two days of selection, and parental cells without plasmids served as the negative control. A portion of these puromycin-resistant enriched cells was harvested for DDX20 detection by Western blotting, and the remaining cells were counted with a Vi-Cell XR Cell Viability Analyzer (Beckman Coulter) and plated at dilutions of 1.5 cells/well in 96-well plates to facilitate the isolation of single-cell clones. After 2–3 weeks, clones were expanded, and DDX20 knockout-positive clone cells were picked and validated by Western blotting.
Generation of MTOR promoter constructs. Genomic DNA isolated from MDA-MB-231 cells was used as a PCR template. A total of 725 bp of the human MTOR promoter [-600, + 107] fragment was amplified by PCR. The fragment was subcloned into a pGL3-basic vector (Promega) at the MluI/XhoI sites. The MTOR promoter reporter with ETV3 binding site mutation was created by directly synthesizing the template by the Synbio-tech company (Suzhou, China) and then cloning it into the pGL3-basic vector.
Splicing reporter. For the splicing reporter plasmid, an intron-containing luciferase reporter, CMV-LUC2CP/ARE (Addgene plasmid #62857), was generated by inserting a 133-nucleotide chimeric intron (pCI-neo, Promega).
Dual luciferase assay. Luciferase-containing reporter plasmids were cotransfected into MDA-MB-231 or HCC-70 cells with the Renilla luciferase vector pRL-SV40 using Lipofectamine 2000. The activity of the reporter was measured using the Dual-Luciferase Reporter Assay System (Promega) after 24 h of transfection and normalized using Renilla luciferase activity in a Molecular Devices Microplate Reader (Spectra Max M5). The results were obtained from three independent experiments.
Chromatin immunoprecipitation assays. Chromatin immunoprecipitation (ChIP)-qPCR assays were performed using an EZ-Magna ChIP A/G kit (Millipore) according to the standard protocol using rabbit polyclonal anti-ETV3 and anti-DDX20. The precipitated DNA samples were analyzed by a quantitative PCR assay, and the data are shown as percentages of the input DNA. Anti-rabbit IgG (Santa Cruz Biotechnology) was used as a control.
In vivo studies. For orthotopic primary tumor formation, shSNRPE #1 tet-on MDA-MB-231 cells were washed with PBS and resuspended in PBS buffer with an equal amount of Matrigel (BD Biosciences, San Diego, CA, United States). Six- to eight-week-old female BALB/c nude mice were injected into the 4th mammary fat pad with 100 µl suspension containing 5.0 × 106 cancer cells. Tumor volume was monitored at 2 weeks after implantation. The volumes were calculated with the formula (mm3) = width × width × length × 0.5, and animals were sacrificed if tumor sizes reached 2000 mm3 for animal ethics. For tail vein injection, luciferase-expressing LM2-4175 cells with tet-on shSNRPE #1 (1.0 × 105 cells in 100 µL PBS) were injected per mouse. The formation of lung metastasis was monitored by bioluminescence imaging using the NightOwl LB981 imaging system. On day 14, to induce SNRPE silencing in vivo, 2 g/L doxycycline was added to the drinking water of mice after grouping according to the normalized photon flux. All animal studies were conducted according to the guidelines of the ethics committee.
RNA-seq. Total RNA was extracted using the RNeasy Mini Kit (QIAGEN) following the instructions. One microgram of total RNA was treated with VAHTS mRNA Capture Beads (Vazyme, Nanjing, China) for the enrichment of polyA + RNA. RNA-seq libraries were performed using a VAHTS mRNA-seq v2 Library Prep Kit for Illumina (Vazyme, Nanjing, China) according to the manufacturer’s instructions. Briefly, enriched RNA samples were fragmented and reverse copied into first- and second-strand cDNA with random hexamer primers. After treatment with end repair and dA-tailing, the dsDNA was ligated with an adapter. The dsDNA was purified and subjected to 12 cycles of PCR amplification, and the libraries were constructed and sequenced on an Illumina sequencing platform. The RNA-seq raw data of sequencing reads were aligned using the spliced read aligner HISAT2 and then mapped to the human reference genome. For alternative splicing (AS) analysis, human genome references of AS events were downloaded from the MISO website and operated according to MISO manuals. The output data were filtered on the basis of both PSI (percent spliced in) differences and PSI distribution plots followed a sashimi plot.
Statistical analysis. Student’s t test was used to compare continuous variables. We used a chi-square test for categorical variables and a chi-square or Fisher’s exact test for contingency tables. The Kaplan‒Meier method was used to compare the clinical outcome between the high-risk and low-risk groups, and the P value was calculated by the log-rank test. Adjusted hazard ratios (HRs) with 95% confidence intervals (CIs) were calculated using Cox proportional hazards modeling. All other statistical tests were two-sided. Statistical significance is shown with NS (not significant) (p > 0.05), *p value ≤ 0.05, **p value ≤ 0.01, *** p value ≤ 0.001. The statistical analyses were performed using SPSS 22.0 (SPSS Inc., Chicago, IL, USA) or R 3.5.0 (R Foundation for Statistical Computing, Vienna, Austria) software programs.