RNA sequencing data from FUSCC
High-throughput RNA sequencing was performed in six NT tissues, five PTC tissues and five DDTC tissues from PTC cases undergoing dedifferentiation, which were described in our previous study [22]. A total of fifteen samples were included in the present study because of the bias of the RNA sequencing data from a NT sample. As shown in Fig. S1A, the transcriptome difference among DDTCs, PTCs and NTs was significant via the correlation matrix analyses for the RNA sequencing data of the enrolled fifteen samples. Moreover, nine ATC, five PDTC, sixteen PTC and twenty-one NT samples from FUSCC were used to validate expression of the lncRNAs screened out by RNA sequencing.
Transcriptional profiles from the GEO database and The Cancer Genome Atlas (TCGA) database
As described in our previous study [22], we obtained raw microarray cell intensity files of thyroid cancer including GSE29265, GSE33630 [23, 24], GSE53157 [25], GSE65144 [26] and GSE76039 [5] from the GEO database (http://www.ncbi.nlm.nih.gov/geo/) [27, 28]. The same chip platform (Affymetrix Human Genome U133 Plus 2.0 Array) was utilized by the files of the GEO cohort. The files were background-adjusted and normalized using Robust Multichip Average [29], and the ComBat method was used to remove the batch effects. All probes were mapped to the latest version of the NetAffx Annotation File. If a gene symbol was recorded with multiple probes, the average value was used as its expression level. The combined GEO cohort included the GSE29265 cohort (20 NTs, 20 PTCs and 9 ATCs), the GSE33630 cohort (45 NTs, 49 PTCs and 11 ATCs), the GSE53157 cohort (3 NTs, 15 PTCs and 5 PDTCs), the GSE65144 cohort (13 NTs and 12 ATCs) and the GSE76039 cohort (17 PDTCs and 20 ATCs). TCGA cohort data were available at the website of DATA SETS of University of California Santa Cruz (UCSC) Xena (THCA_22 datasets: https://xenabrowser.net/datapages/; https://www. biorxiv.org/content/early/2018/08/28/326470) and The cBioPortal for Cancer Genomics (http://www.cbioportal.org/). The data from the TCGA cohort and the GEO cohort were described in detail in our previous study [22].
Cell culture and reagents
CAL-62 and B-CPAP cell lines were purchased from the Shanghai Cell Bank Type Culture Collection Committee (CBTCCC, Shanghai, China). We cultured the cells at 37℃ in a humidified incubator with 5% CO2. CAL-62 and B-CPAP were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Invitrogen, Carlsbad, CA, USA) and Roswell Park Memorial Institute (RPMI) 1640 medium (Invitrogen), respectively, supplemented with 10% foetal bovine serum (FBS) (HyClone, Logan, UT, USA), 100 U/ml penicillin and 100 µg/ml streptomycin (Invitrogen), and 8 mg/l antibiotic tylosin tartrate against mycoplasma (Sigma-Aldrich, St. Louis, Missouri, USA). The proteasome inhibitor MG132 and Cycloheximide (CHX) were purchased from Sigma-Aldrich.
Protein-coding potential of LINC00886
The Coding Potential Assessment Tool (CPAT) (http://lilab.research.bcm.edu/cpat/) and Coding Potential Calculator (CPC) (http://cpc.cbi.pku.edu.cn/) were used to predict the protein-coding potential of LINC00886. ACTB and GAPDH were used as coding RNA controls, and NEAT1 and MALAT1 were used as non-coding RNA controls.
5’ and 3’ rapid amplification of the cDNA ends (RACE)
5’and 3’RACE were used to determine the initiation and termination sites of the LINC00886 transcript with a SMARTer RACE cDNA Amplification kit (Clontech, California, USA), according to manufacturer’s instructions. The sequences for the gene-specific PCR primers used for 5’and 3’RACE analysis are listed in Table S1.
Northern blot
LINC00886 was detected using a NorthernMax Kit from Ambion (Thermo Fisher Scientific, Carlsbad, California, USA) and DIG Northern starter Kit (Roche, Indianapolis, Indiana, USA) with Digoxin-labelled RNA probes in the CAL-62 cells. The primers of the LINC00886’s probes are listed in Table S1.
Subcellular distribution
We extracted cytoplasmic and nuclear fractions of the CAL-62 cells using the Nuclear/Cytoplasmic Isolation kit (Thermo Fisher Scientific, Carlsbad, California, USA). β-actin served as the cytoplasmic endogenous control. U2 served as the nuclear endogenous control.
Overexpression and knockdown of LINC00886 and PKR
The human LINC00886 sequence was cloned from CAL-62 cell cDNA and cloned into the expression vector pWPXL to generate pWPXL-LINC00886. Additionally, we cloned the sequence of PKR into the pCMV-N-Flag vector. The CAL-62 cells were transfected with pWPXL-LINC00886 and pCMV-PKR-N-Flag using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s instructions. We used Ribo™ lncRNA Smart Silencer for LINC00886 knockdown, and small interfering RNA (siRNA) oligonucleotides targeting PKR was used for PKR knockdown. The lncRNA smart silencer is a mixture of three anti-sense oligonucleotides (ASO) and three siRNAs. The sequences of lncRNA smart silencer against LINC00886, siRNAs targeting PKR and the negative control (NC) siRNA were provided in Table S1, which were purchased from RiboBio (RiboBio Biotechnology, Guangzhou, China) and Genomeditech (Genomeditech, Shanghai, China). Transfections with lncRNA smart silencer and siRNA were performed with Lipofectamine RNAiMAX (Invitrogen).
Reverse transcription PCR and quantitative real-time PCR (qPCR)
TRIzol reagent (Invitrogen) was used to extract total RNA from the tissue specimens and cell lines. First-strand cDNA was synthesized using the PrimeScript™ Reverse Transcriptase kit (Takara, Dalian, China). We utilized quantitative qPCR to detect relative RNA levels on a 7900 Real-Time PCR System with the SDS 2.3 software sequence detection system (Applied Biosystems, USA) using the SYBR Green (Takara) method. The sequences for the gene-specific primers are listed in Table S1. β-actin is an internal control to quantify LINC00886 and the mRNA levels of other genes. The relative levels of RNA are calculated by the comparative CT (2−ΔΔCT).
Cell proliferation assay and colony formation assay
The CAL-62 cells were seeded in 96-well flat-bottomed plates, with each well containing 2000 cells in 100 µl of cell suspension. Cell viability was measured using Cell Counting Kit-8 (CCK-8) assays (YEASEN, Shanghai, China). We repeated each assay with five replicates for three times and measured continuously for 5 days. Cells were seeded in 6-well culture dishes, and after 1 week-2 weeks, cell colonies were fixed with methanol, stained with crystal violet (Sigma-Aldrich, St. Louis, MO), and counted based on the colony formation number.
Cell invasion assay and migration assay
For the migration assays, the 5 × 104 CAL-62 cells were added to the Millicell chambers with 100 µl of serum-free medium, which were ahead put inside the 24-well culture dishes containing 500 µl of DMEM with 10% FBS. Invasion assays were performed similarly with coating the filters with Matrigel (BD Biosciences, Franklin Lakes, New Jersey, USA). The cell invasion and migration assays were performed in triplicate. After 24 h at 37 °C in an incubator with 5% CO2, migrated cells were fixed with methanol and stained with crystal violet, and cell numbers were counted in five random fields.
RNA sequencing
We extracted total RNA from the CAL-62 cells transfected with LINC00886 smart silencer and NC-siRNA using TRIzol reagent. The RNA samples from si-LINC00886 and NC cells were used for RNA sequencing as mentioned in our previous study [22]. The RNA sequencing data were exhibited in Table S2.
Gene Set Enrichment Analysis (GSEA)
GSEA was performed based on the RNA sequencing data of si-LINC00886 and NC CAL-62 cells and transcriptional profiles of the GEO ATCs and the TCGA PTCs using GSEA software, Version 2.0, which was obtained from the Broad Institute (http://www.broad.mit.edu/gsea), as previously described. Enrichment Map was used for visualization of the GSEA results. Normalized enrichment score (NES) and false discovery rate (FDR) were used to sort the pathways enriched in each phenotype after gene set permutations were performed 1000 times for each analysis.
Western blot
We used lysis buffer (YEASEN, Shanghai, China) with protease inhibitors (Roche, Indianapolis, IN, USA) to lyse CAL-62 cells. Protein concentrations were determined by the BCA method (Pierce, Therrmo Fisher Scientific Inc., Rockford, IL, USA). The protein samples were subjected to SDS/PAGE and transferred to PVDF membranes (Immobilon-P membrane, Millipore, Massachusetts, USA). The membranes were blocked with 5% skimmed milk in TBS plus Tween 20 at room temperature for 1 hour, followed by incubation with target antibodies at 4 °C overnight. Information on the antibodies are provided in Table S3. After incubation with HRP-conjugated secondary antibodies for 1 hour, visualization of the protein bands was achieved by an enhanced chemiluminescent chromogenic substrate using the Enhanced Chemiluminescence Plus Western Blotting Detection System (GE Healthcare, Connecticut, USA) and LAS-4000EPUV mini Luminescent Image Analyzer (GE Healthcare).
RNA pull down
Initially, LINC00886 and antisense-LINC00886 RNAs were labelled by the Biotin RNA Labeling Mix (Roche, USA) and purified with an RNeasy Mini Kit (QIAGEN, USA). Then, the biotinylated RNAs were incubated with streptavidin beads (Invitrogen, USA), and the streptavidin beads combined with and without RNAs were mixed with protein extracts of CAL-62 cells at 4 °C on a rotator overnight. The beads were washed gently three times in 1 × washing buffer (5 mM Tris-HCl, 1M NaCl, 0.5 mM EDTA, and 0.005% Tween 20), followed by mix with DEPC and 5 × SDS buffer. The proteins were separated by gel electrophoresis and visualized by silver staining. Specific bands were identified by mass spectrometry analysis and retrieved in the human RefSeq protein database (National Center for Biotechnology Information), using Mascot version 2.4.01 (Matrix Science, London, UK). The retrieved protein was detected by western blot. The primers of LINC00886 are provided in Table S1.
RNA immunoprecipitation (RIP)
We performed RIP assays using the Magna RIP RNA-binding protein immunoprecipitation kit (Millipore, Massachusetts, USA) according to the manufacturer’s instructions. The CAL-62 cells were lysed in RIP lysis buffer, and the lysate products were immunoprecipitated with PKR or lgG antibody combined with protein magnetic beads. Next, we immobilized the magnetic beads bound complexes and washed unbound materials. The RNAs were extracted using the Total RNA isolation kit (Thermo Fisher Scientific Inc) and analyzed by qPCR and real-time PCR. For the RIP assays of deletion mutants, plasmids with FLAG-full-length and truncated PKR were transiently transfected into the CAL-62 cells, and the cell lysates were immunoprecipitated with the indicated antibodies.
Immunoprecipitation
Immunoprecipitation assays were performed using the Pierce™ Classic Magnetic IP/Co-IP Kit (Thermo Fisher Scientific Inc). The CAL-62 cells were transfected with the indicated plasmids and then lysed in ice-cold IP Lysis/Wash Buffer (pH 7.4, 0.025M Tris, 0.15M NaCl, 0.001M EDTA, 1% NP40, 5% glycerol). Cell lysate were incubated with anti-PKR antibody for 1–2 hours at room temperature or overnight at 4ºC. And then, the antigen/antibody complexes were mixed with protein magnetic beads (Thermo Fisher Scientific Inc.) at room temperature for one hour. The beads were washed twice with IP Lysis/Wash Buffer and once with purified water, and the antigen/antibody complexes were eluted. The immuno-complexes were analysed by SDS/PAGE and immunoblotting with anti-PKR, anti-ubiquitin and anti-β-actin antibody.
Statistical Analysis
Continuous variables were respectively expressed as the mean ± standard error of the mean (SEM) from at least three independent experiments and mean ± standard deviation (SD) in analyses of associations between clinical parameters and molecular variables, and categorical data were summarized with frequencies and percentages. Student’s t-test and one way analysis of variance were performed to evaluate the differences of continuous variables between two groups and among more than two groups, respectively. Associations between continuous variables and categorical variables were evaluated using Mann-Whitney U tests for two groups and Kruskal-Wallis tests for more than two groups. χ2 and Fisher’s exact test were used for categorical variables. Based on the median value of thyroid differentiation score (TDS), we divided the PTC patients from the TCGA cohort into a low-differentiated group and a high-differentiated group. Pearson’s correlation was performed to analyze the correlation of LINC00886 with the TDS. Nonparametric receiver operating characteristic (ROC) analyses were performed to calculate the area under curve (AUC) for LINC00886 that would be predictive of dedifferentiation. Univariate logistic regression analyses were performed to determine associations of LINC00886 with clinicopathological factors in PTC patients from TCGA. Furthermore, LINC00886 was tested for overall survival (OS) analyses in thyroid cancer patients using the GSE76039 cohort [5]. The Kaplan-Meier method was used to construct OS curves, and the univariate survival difference was determined by the log-rank test. A p value < 0.05 was considered significant. Data preparation and statistical analyses were performed using the SPSS for Windows (version 22.0; IBM Corp., Armonk, NY), the R software (version 3.5.1; R Foundation for Statistical Computing, Vienna, Austria) and the GraphPad Prism (version 6.01; GraphPad Software Inc., La Jolla, Calif).