Tissues and cell lines
The tumorous tissues (T) and paired adjacent normal tissues (N) were collected from NSCLC patients who underwent surgery at Department of Thoracic Surgery in Union Hospital of Tongji Medical College of Huazhong University of Science and Technology from July 2014 to December 2018. Written informed consent was obtained from all patients. Ethical approval was confirmed by Institutional Review Board of Tongji Medical College of Huazhong University of Science and Technology. All NSCLC tissues were snap-frozen in liquid nitrogen and then stored at -80℃ until use. All the surgical samples were confirmed by the pathologists using H&E-stained sections. The clinical characteristics of patients were presented in Table S1. All of the patients were followed up on a regular basis, and overall survival (OS) time was determined from the date of surgery to the date of death or the date of the last follow-up visit for survivors.
Human NSCLC cell lines A549 (RRID:CVCL_0023), NCI-H1299 (RRID:CVCL_0060), NCI-H226 (RRID:CVCL_1544), SPC-A1 (RRID:CVCL_6955), HBE (human bronchial epithelial cell line 1, HBE1(RRID:CVCL_0287)), and LLC (murine Lewis lung cancer cell line, LL/2 (LLC1) (RRID:CVCL_4358)) were purchased from the American Type Culture Collection. H1299, H226, SPC-A1 and HBE cells were cultured in Roswell Park Memorial Institute 1640 (RPMI-1640) culture medium supplemented with 10% FBS. A549 cells were cultured in Ham's F-12K (Kaighn's) medium supplemented with 10% FBS (Gibco, Australia origin). LLC cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% FBS. All cells were cultured at 37 ℃ with 5% CO2.
All cell lines have been authenticated in the past three years according to the manufacturer’s instructions. All experiments were performed with mycoplasma-free cells.
Identification and annotation of human NSCLC circRNAs
Total RNA from four pairs of NSCLC and adjacent normal lung tissues was isolated using the RNeasy Mini Kit (QIAGEN, Germany) according to the manufacturer’s instructions. The total RNA samples were treated with the RiboZero rRNA Removal Kit (Epicentre, WI, USA) for deleting rRNA. Next, RNA was digested with RNase R (Epicentre, Madison, WI, USA) to remove linear RNA and enrich for circular RNAs. The remaining RNAs were fragmented and synthesized cDNA with random primer. Then, the libraries were quality controlled and sequenced by Illumina NovaSeq 6000 (Illumina, San Diego, CA, USA). After that, circRNAs were detected and identified with the tools find_circ26 and CIRI227. The DESeq R package (1.10.1) was used for differential expression analysis between NSCLC and normal lung tissues. The resulting P values were adjusted using the Benjamini and Hochberg’s approach for controlling the false discovery rate. The RNA-seq data was deposited in SRA database (Accession code: PRJNA863919).
RNA preparation, RNase R treatment and qRT-PCR
Total RNA was isolated from tumor tissue, normal tissue or cells by using RNeasy Mini Kit (QIAGEN, Germany). Nuclear and cytoplasmic RNA was extracted using nuclear and cytoplasmic RNA purification kit (Fisher scientific, AM1921). For RNase R treatment, one microgram of total RNA was incubated 15 min at 37°C with or without 3 U of RNase R (Epicentre Technologies, Madison, WI). Then, RNA was reverse-transcribed using the PrimeScript RT Master Mix (Takara, Dalian, China). SYBR Premix Ex Taq II (Takara) was used for real-time PCR. Particularly, the divergent primers annealing at the distal ends of circRNAs were used to determine the abundance of circRNAs. Genomic DNA (gDNA) was extracted using Genomic DNA Isolation Kit (Sangon Biotech, Shanghai, China). The cDNA and gDNA PCR products were observed using 2% agarose gel electrophoresis. The Amplification procedure was performed with the StepOne Plus Real-Time PCR system (Applied Biosystems, Foster City, CA). The Ct thresholds were determined by software. The primers are demonstrated in Table S2.
Actinomycin D treatment and RNA stability assay for RNA lifetime
Cells were placed into six-well plates for actinomycin D (Sigma-Aldrich, USA) experiments. Up to 60% confluency after 24 h, cells were treated with 5 µg/ml Actinomycin D or DMSO (Sigma-Aldrich, USA) and collected at indicated time points. The turnover rate and half-life of RNA was estimated according to a previously published paper28.
RNA Fluorescent in situ hybridization (FISH)
Cy3-labelled circFNDC3B and circFndc3b probes (Table S2) were synthesized by TSINGKE (Wuhan, China). FISH was performed as described with minor modifications29. The cancer cells were fixed and permeabilized, followed by hybridizing with the probes at 37 ℃ overnight in a dark. The coverslips were washed three times 2 × SSC (Solarbio, Beijing, China) for 10 min, before they were sealed with parafilm containing DAPI (Roche Diagnostic, Mannheim, Germany). The images were obtained with a confocal laser scanner (LSM 780, Carl Zeiss).
Cell counting Kit-8 (CCK-8) experiment
The proliferation of cells was measured by CCK-8 kit (Dojindo, Japan) following the manufacturer’s instructions. The optical density at 450 nm was measured using an automatic microplate reader (Synergy4; BioTek, Winooski, VT, USA).
Transwell matrigel invasion assay
The matrigel invasion assay was performed using the transwell precoated matrigel chamber following the manufacturer’s protocol (BD Science, Bedford, MA, USA). The homogeneous single cell suspensions (1×105 cells/well) were added to the upper chambers and incubated for 24 h. The invasion rates were quantified by counting the invaded cells at least three random fields.
Apoptosis assay
The cell apoptosis assay was detected according to the manufacturer’s instructions of FITC Annexin V Apoptosis Detection Kit I (BD Biosciences). The data was then analyzed using the FCS Express 5 software (De Novo Software, Los Angeles, CA).
Cell cycle assay
Cells were fixed in 75% ice-cold ethanol overnight at 4°C. After washing the fixed cells with PBS, propidium iodide (PI) buffer (BD Pharmingen, USA) was applied for staining. Cell cycle analysis was performed by flow cytometer. The data were analyzed using ModFit LT 5.0.
RNA sequencing
Total RNA was isolated from circFNDC3B-overexpressed A549 and H226 cells and the corresponding control cells by using RNeasy Mini Kit (QIAGEN, Germany). Library preparations were conducted using NEBNext® Ultra™ RNA Library Prep Kit for Illumina® (New England Biolabs). The gene expression profiles were determined by Illumina high-throughput RNA sequencing (Novogene, Beijing, China). Significant differentially expressed transcripts were screened by p < 0.05 and log2(fold change) ≥ 1.
Enzyme linked immunosorbent assay (ELISA)
The expression levels of CXCL10 and CXCL11 in the cell culture supernatants were determined with the commercially available ELISA kits from (Biolegend and Raybiotech) following the instructions provided by the manufacturer.
RNA pull-down assays
Biotin-labeled oligonucleotide probes targeting junction site of circRNA (sense) and control (antisense) probes (Table S2) were synthesized by TSINGKE (Wuhan, China). For RNA pull-down assay, 1 × 107 cells were washed in ice-cold phosphate-buffered saline, lysed in 500 µl Co-IP buffer (20mM Tris-HCL, pH 7.5, 150mM NaCl, 1mM EDTA, 0.5% NP-40, and complete protease inhibitors cocktail and RNase inhibitors), and then incubated with 3 µg biotin-labelled circFNDC3B (sense) and control (antisense) probes for 2 h at room temperature. A total of 50 µl washed Streptavidin C1 magnetic beads (Invitrogen) were added to each binding reaction and incubated at room temperature for another hour. After washing with the Co-IP buffer for five times, the proteins bounding to the beads were eluted, and analyzed by mass spectrometry or western blotting.
Silver staining and mass spectrometry analysis
Silver staining was performed using the PAGE Gel Silver Staining Kit (Solarbio, Beijing, China) based on the protocol described. Retrieved proteins were detected by mass spectrometry analysis at Novogene Company (Beijing, China). Afterwards, protein identification and quantification were implemented with Proteome Discoverer software (version 1.4; Thermo Fisher Scientific, USA).
Western blot and Co-immunoprecipitation (Co-IP)
For western blot, whole cell lysates were prepared using RIPA buffer containing protease inhibitors (Beyotime). The total protein lysates from cells were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and transferred onto polyvinylidene difluoride membranes. After blocking with 5% non-fat milk, membranes were incubated with primary antibody overnight at 4°C, followed by an incubation with HRP conjugated secondary antibodies 1 h at room temperature. Immunoreactive bands were detected by using the enhanced chemiluminescence (E412–01, Vazyme, Nanjing, China).
For co-immunoprecipitation, cells were lysed in Co-IP buffer containing protease inhibitor cocktail for 40 min on ice. Cell lysates were incubated with the corresponding antibodies adsorbed to protein A/G Agarose (Thermo Fisher Scientific, #20421) for 4 h at 4°C. Afterwards, the cell lysates were washed three times in Co-IP buffer and eluted at 95°C for 10 min.
Antibodies used included primary antibodies against TFⅡ-I (Santa Cruz Biotechnology, sc-136330, 1:500), AGO2 (Thermo Fisher Scientific, MA5-23515, 1:500), STAT1 (Abcam, ab239360, 1:1000), pSTAT1 (Abcam, ab109461, 1:1000), Flag (Abcam, ab205606, 1:1000), GAPDH (Proteintech, 60004–1-Ig, 1:50000), Histone-H3 (Proteintech, 17168-1-AP, 1:1000). HRP-conjugated secondary goat anti-mouse (Proteintech, SA00001–1, 1:2000), HRP-conjugated secondary goat anti-rabbit (Proteintech, SA00001–2, 1:2000), HRP-mouse anti-rabbit IgG heavy chain specific (Proteintech, SA00001-7H, 1:5000) and HRP-mouse anti-rabbit IgG light chain specific (Proteintech, SA00001-7L, 1:4000) antibodies were used as second antibodies.
RNA Immunoprecipitation (RIP)
RIP experiment was carried out with the Magna RIP RNA-Binding Protein Immunoprecipitation Kit (Millipore, Bedford, MA) according to the manufacturer’s instructions. Cell lysates were incubated with dynabeads coated with AGO2 (Thermo Fisher Scientific, MA5-23515, 8µg), TFⅡ-I (Santa Cruz Biotechnology, sc-136330, 1µg), Flag (Abcam, ab125243, 1µg) or Mouse IgG (Santa Cruz Biotechnology, sc-2025, 1µg) antibody at 4°C overnight, respectively. The co-precipitated RNAs were abstracted using RNeasy MinElute Cleanup Kit (Qiagen), and detected by qPCR.
Immunocytofluorescence
NSCLC cells were grown on the coverslips and fixed with 4% paraformaldehyde. After permeabilizing with 0.1% TritonX-100, cells were washed with PBS, blocked with 5% BSA and incubated with TFⅡ-I (Santa Cruz Biotechnology, sc-136330, 1:500) antibody overnight. Subsequently, cells were washed with PBS again, following by incubating with fluorophore-labeled secondary antibody to mouse IgG (Abcam, ab150113, 1:500). Finally, parafilm containing DAPI was applied for the sealing of cells. Fluorescence images were obtained with a confocal laser scanner (LSM 780, Carl Zeiss).
Vector construction and cell transfection
To establish circFNDC3B, circFndc3b, TFⅡ-I and STAT1 over-expression plasmids, circFNDC3B, circFndc3b, TFⅡ-I and STAT1 cDNA were synthesized by TSINGKE (Wuhan, China). Then they were cloned into pcDNA3.1(+) CircRNA Mini Vector (addgene #60648) and p3XFLAG-CMV-10 vector (Sigma-Aldrich), respectively. The truncations of TFⅡ-I were constructed as region containing 102-399aa (3×Flag_102–399), 400-681aa (3×Flag_400–681), 682-972aa (3×Flag_682–972), 102-681aa (3×Flag_102–681) and 400-972aa (3×Flag_400–972). The Flag-tagged TFⅡ-I truncations were subcloned into p3XFLAG-CMV-10 vector. Oligonucleotides encoding short hairpin RNAs (shRNAs, Table S2) specific for circFNDC3B and STAT1 were cloned into pLKO.1-puro (Sigma-Aldrich).
Transfection was performed using Lipofectamine 2000 (Life Technologies) according to the manufacturer’s instructions. Stable cell lines were screened by administration of neomycin or puromycin (Invitrogen). Scramble shRNA and empty vector were used as controls.
Chromatin immunoprecipitation and qPCR (ChIP-qPCR) assays
The procedure for ChIP experiment was performed with the EZ-Magna ChIP A/G Chromatin Immunoprecipitation kit (Sigma-Aldrich). The DNA fragments were resuspended in PBS and incubated with anti-STAT1 (Abcam, ab239360, 5µg) or Rabbit IgG (Abcam, ab172730, 5µg) at 4°C overnight. The precipitated DNA-chromatin products were determined by qPCR. The data were demonstrated as fold enrichment (normalized to IgG). The primer sequences utilized in ChIP-qPCR were showed in Table S2.
Dual-luciferase reporter assay
A549 cells were co-transfected with a reporter constructs for CXCL10 or CXCL11 promoter (-1000 to 300 bp) in pGL3-Basic vector, and a Renilla luciferase reporter vectors pRL-TK as the internal control. The firefly and Renilla luciferase activities were quantified using Dual-Luciferase® Reporter Assay System (Promega, USA) according to the manufacturer’s protocol, after transfection for 24 h.
Purification of human CD8+ T cells
Ficoll-Hypaque gradient centrifugation was utilized for the isolation of mononuclear cells from healthy volunteers’ peripheral blood. Purified CD8+ T cells were isolated with the human CD8+ T cells isolation kit (Miltenyi Biotec, Teterow, Germany) according to the manufacturer’s protocol. The purity of CD8+ T cells was > 90%, as confirmed by flow cytometry. Purified CD8+ T cells were cultured in RPMI-1640 medium with a 10% FBS concentration. The medium contained recombinant human IL-2 (Abcam, ab119439, 10 ng/ml), soluble anti-human CD28 mAbs (Biolegend, 302937, 1 µg/ml) and plate-bound anti-human CD3 (Biolegend, 317301, 1 µg/ml) mAb. The cells were then cultured for three consecutive days for the subsequent experiments.
Chemotaxis assay
Preactivated CD8+ T cells were resuspended in RPMI 1640 medium with 0.5% FBS, and placed in the upper chamber of Transwell tissue culture inserts (5.0-µm pore diameter) (Corning, New York, USA). Supernatant from A549 cells were added in the lower chamber. RPMI-1640 medium alone as a negative control. When indicated, anti-CXCL11 (R&D Systems, MAB672, 1µg/ml), anti-CXCL10 (R&D Systems, MAB266, 1µg/ml), recombinant human CXCL11 (R&D Systems, 672-IT, 50ng/ml), recombinant human CXCL10 (R&D Systems, 266-IP, 50ng/ml) were added to the lower chamber. The chambers were incubated at 37°C, 5% CO2 for 4 h. Transmigrated cells in the lower chambers were collected and stained with anti-human CD8 mAbs (Biolegend, 980908). After staining, precision count beads (Biolegend, 424902) were added, and the numbers of transmigrated CD8+ T cells were determined using flow cytometry (FACS Canto II; BD Bioscience). The chemotactic index was calculated as the ratio of the number of CD8+ T cells that migrated to corresponding wells divided by the number of CD8+ T cells that migrated to RPMI-1640 medium alone.
In vivo tumorigenesis assays
The NOD-SCID mice or C57BL/6 female mice (4 weeks old) were obtained from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). The mice were bred on a chow diet in a 12-h light/12-h dark environment at 25°C in the Tongji Medical College Animal Care Facility under high health status conditions. All animal studies were approved by the Animal Care and Utilization Committee of Tongji Medical College, Huazhong University of Science and Technology.
For NOD-SCID mice experiments, circFNDC3B-overexpressing and control A549 cells (2 × 106 cells) were subcutaneously injected in the right and left flank of the same NOD-SCID mice, respectively (n = 6). For some experiments, purified human CD8+ T cells (1 × 107 per mice) were injected via the tail vein of NOD-SCID mice (n = 6), beginning a week before inoculation of tumor cells, weekly. After four weeks, mice were euthanatized, the tumor were dissected. The volumes of the dissected tumors were measured using calipers and calculated with the following formula: a2 × b × 0.5 (a indicates the smallest diameter and b represents the diameter perpendicular to a).
For C57BL/6 mice experiments, circFndc3b-overexpressing and control LLC cells (5 × 105 cells) were subcutaneously injected in the right flank of C57BL/6 mice (n = 6 for each group). After two weeks, mice were euthanatized, the tumor were dissected. The volumes of the dissected tumors were determined as mentioned above.
For anti-PD1 antibody treatment, circFndc3b-overexpressing and control LLC cells (5 × 105 cells) were subcutaneously injected in the right flank of C57BL/6 mice (n = 6 for each group) on day 0. Anti-mouse PD-1 monoclonal antibody (Biolegend, 135202) or its isotype control (Biolegend, 400501) were injected into C57BL/6 mice through the tail vein with at 100 µg per dose every four days for four times, beginning on day 6. The growth of the tumor was determined with calipers every 3 days. The volumes of tumor were measured as mentioned above. When tumors reached a maximum of 2000mm3, the mice were euthanized.
CD8+ T cells infiltrating analysis by flow cytometry and immunohistofluorescence
Dissected tumors were treated with DNase I (Sigma, 50 µg/ml) and collagenase type IV (Gibco, 200 U/ml) for 20 minutes at 37°C. To obtain a single cell suspension, cells were passed through a 70-µm filter to remove clumps, diluted in medium. Then cells were surface-stained with anti-mouse CD45 mAbs (Biolegend, 147708), anti-mouse CD3 mAbs (Biolegend, 100206) and anti-mouse CD8 mAbs (Biolegend, 126606) for flow cytometry.
Immunohistofluorescence was performed on formalin-fixed tumor tissues. Five-micrometer paraffin-embedded sections were deparaffinized and rehydrated in graded alcohol series. Staining for human CD8 (Abcam, ab237710, 1/100) or mice CD8 (Abcam, ab217344, 1:500) was carried out with non-labeled primary antibodies and fluorophore-labeled secondary antibodies (Thermo Fisher Scientific, A-11008, 1:1000).
Then the slices were counterstained with DAPI in mounting medium. The images were obtained with a confocal laser scanning microscope (LSM 780, Carl Zeiss) and analyzed using ImageJ software.
Statistical analysis
All analyses of the data in this study were performed using SPSS 26.0 software, and the results were presented as mean ± standard deviation. Two tailed Student’s t-test was carried out between two groups and one-way ANOVA was applied in multiple groups. Overall survival was estimated by the Kaplan-Meier method and compared by the log-rank test. P < 0.05 was considered statistically significant.