Cell culture and reagents
The tumor cell lines and HEK293T were originally obtained from the American Type Culture Collection (ATCC) or Japanese Collection of Research Bioresources Cell Bank (JCRB), where mycoplasma contamination was routinely tested and cell identity was monitored by short tandem repeat (STR) profiling. Drug-tolerant persister cells were generated through continuous treatment with 1 µM of erlotinib for 10 days, according to protocols described previously 6. Drug-resistant cells were generated by overexpressing the EGFRT790M mutant in PC9 cells, and then maintained in 1 µM erlotinib. Cells were cultured in RPMI1640 (Life Technologies) supplemented with 10% fetal bovine serum (Gibco), l-glutamine (2 mM), penicillin (100 units/mL), and streptomycin (100 µg/mL). Small-molecule inhibitors were purchased from Selleck Chemicals or MedChemExpress, and reconstituted in DMSO (Sigma-Aldrich) at a stock concentration of 10 mM. The following inhibitors were used at these final concentrations unless otherwise indicated: erlotinib (1 µM), afatinib (4 µM), osimertinib (4 µM), trametinib (5 µM), ulixertinib (1 µM), pictilisib (1 µM), ipatasertib (1 µM), alectinib (5 µM), capmatinib (4 µM), PLX8349 (4 µM), erdafitinib (2 µM), sotorasib (1 µM), chloroquine (50 µM). For visualization, cells were fixed with formalin and stained with crystal violet.
Plasmids and sgRNAs
Plasmids for gene overexpression were constructed using the Gibson Assembly Cloning Kit (New England Biolabs) and Gateway Cloning System (Invitrogen). EGFR mutations were generated using the Q5 Site-Directed Mutagenesis Kit (New England Biolabs) and verified by Sanger sequencing. The CRISPR-Cas9 system was employed to knock out indicated genes. The primers for cloning and sgRNA sequences were provided in Supplementary Table 4.
Virus production and cell infection
For virus production, HEK293T cells in a 10-cm dish were co-transfected with 5 µg of lentiviral constructs, 5 µg of plasmid Δ8.9, and 3 µg of plasmid VSVG using Lipofectamine 2000. Cells were incubated at 37°C and the medium was replaced with fresh complete medium after 12 h. Virus-containing medium was collected at 48–72 h after transfection and supplemented with 8 µg/mL polybrene (Fluka) to infect target cells in 6-well dishes. Infected cells were selected with 2–5 µg/mL puromycin or blasticidin for one week.
Western blotting analysis
Cells were lysed in RIPA buffer (50 mM Tris pH 7.4, 150 mM NaCl, 1% NP-40, 0.1% SDS, 2 µM EDTA) containing protease inhibitors (Roche) and phosphatase inhibitors (Roche). Protein concentrations were quantified using Pierce BCA Protein Assay Kit (Thermo Fisher Scientific). The cell lysates (~ 20 µg protein) were subjected to SDS-PAGE (Invitrogen) and Western blot. The following primary antibodies were used: YY1 (ab109237, Abcam), phospho-EGFR (Y1068) (#48576, Cell Signaling Technology), EGFR (#4267, Cell Signaling Technology), phospho-ERK (T202/Y204) (#9106, Cell Signaling Technology), ERK (#4695, Cell Signaling Technology), phospho-AKT (T308) (#2965, Cell Signaling Technology), AKT (#2966, Cell Signaling Technology), LC3-II (#12741, Cell Signaling Technology), β-actin (#5125, Cell Signaling Technology).
Immunofluorescence staining
Cells were fixed with 4% paraformaldehyde for 15 min, and permeabilized with 0.1% Triton X-100 in PBS for 10 min. After three PBS washes, cells were blocked with 2% BSA in PBS for 30 min at room temperature (RT), and incubated with primary antibodies against YY1 (#66281-1-Ig, Proteintech) diluted in 2% BSA at 4°C overnight. Cells were incubated with Alexa Fluor 488-labeled anti-mouse IgG (A11029, Invitrogen) and Alexa Fluor 594-labeled anti-rabbit IgG (A11037, Invitrogen) for 1 h at RT in the dark, followed by 4',6-diamidino-2-phenylindole (DAPI) (Invitrogen) counterstaining for 5 min. The staining images were acquired using a confocal laser scanning microscope (Leica). For RFP-GFP-LC3 analysis, PC9 cells were transfected with 2µg of tandem monomeric RFP-GFP-LC3 plasmid for 48 h and subjected to the indicated treatment. For evaluating tandem fluorescent LC3 puncta, the cells were rinsed once with PBS followed by confocal microscopy (Leica) analysis. The number of LC3 puncta per cell was counted using Image J (v 2.1.0).
EdU incorporation assay
The EdU (5-ethynyl-2'-deoxyuridine) incorporation assay was performed with the Cell-Light™ EdU DNA Cell Proliferation Kit (RiboBio) according to the manufacturer’s protocol. Briefly, cells were treated as indicated, incubated with EdU (50 µM) for 2 h, and fixed in 4% paraformaldehyde at RT for 30 min. The cells were stained with ApolloGreen fluorescent dye, followed by incubation with Hoechst reaction solution. Stained samples were viewed under a fluorescence microscope (Leica).
Patient samples
Human samples were obtained in accordance with ethical guidelines of U.S. Common Rule, and the study was approved by the Ethics Committee of Ren Ji Hospital. Written informed consent was acquired from all patients. Paired lung adenocarcinoma samples were collected before and after neoadjuvant targeted therapy, or at baseline and upon disease recurrence following first-line EGFR inhibitor treatment. In addition, we assembled two cohorts of treatment-naïve lung adenocarcinomas. One cohort contained 18 patients, whose tissue specimens were subjected to Western blotting analysis. The other cohort contained 186 patients with driver gene mutations for immunohistochemistry staining of the formalin-fixed and paraffin-embedded (FFPE) sections.
Immunohistochemistry staining
The tissue slides were baked, dewaxed with xylene, passed through graded alcohols, and antigen retrieved with 10 mM citric sodium (pH 6.0) in a steam pressure cooker for 20 min. The slides were then treated with 3% hydrogen peroxide solution in methanol for 10 min to quench endogenous peroxidase activity, blocked with goat serum and incubated with primary antibodies against YY1 (ab81552, Abcam), followed by incubation with horseradish peroxidase-conjugated secondary antibody for 1 h at RT. Antigen visualization was performed using 3,3’-diaminobenzidine (DAB) chromogen (Vector Laboratories). Slides were counterstained with hematoxylin, dehydrated and cover-slipped with mounting solution (Invitrogen). Whole slides were scanned with a Leica Aperio CS2 slide scanner system (Leica Biosystems). The staining was assessed independently by two pathologists without knowledge of patient characteristics based on the H-score method. The H-score was calculated by adding the percentage of positive cells multiplied by an ordinal value corresponding to the intensity level (none = 0, weak = 1, moderate = 2, strong = 3). H-scores ranged from 0 to 300, and tissue samples were further defined as negative (0–49), weak (50–99), medium (100–199), or positive (200–300).
Cell cycle analysis
The cell cycle was measured using iodide (PI) staining and flow cytometry analysis. Briefly, genetically edited PC9 cells were collected and washed once with PBS. The cells were then fixed with cold 70% ethanol for 30 minutes and resuspended in Propidium Iodide (PI)/RNase Staining Solution (Cell Signaling Technology). FACS AriaII cytometer (BD Biosciences) was used for the flow cytometry analysis and the data was processed with Flowjo software.
Transmission electron microscopy
For transmission electron microscopy (TEM), PC9 cells were treated as indicated, washed twice with PBS, and then fixed for 6 h at RT in 2.5% ultrapure glutaraldehyde in PBS. Postfixation was done by 1% osmium tetroxide for 90 min at 4°C, before washing four times in PBS and dehydrating with gradient ethanol solutions (Sinopharm Chemical Reagent) from 50–100% in a 10% graded series. Infiltration was carried out using ethanol with propylene oxide (1:1 ratio) for 30 min. The specimens were embedded in Epon 812 resins (Ted Pella), followed by polymerization at 60°C for 48 h. Ultrathin sections were cut to a thickness of 70 nm by Leica EM UC7 ultramicrotome and stained with saturated uranyl acetate in 50% ethanol, followed by Reynolds lead citrate. Electron microscopy images were obtained using a TEM system (FEI Tecnai G2 Spirit BioTwin).
RNA extraction and quantitative PCR assay
Total RNA from cells was extracted with RNA Isolation Kit (Vazyme Biotech) and converted to cDNA using the High-Capacity cDNA Reverse Transcription Kit (Invitrogen). Quantitative real-time PCR was performed on a ViiA™ 7 Real-Time PCR System (Applied Biosystems) using ChamQ Universal SYBR qPCR Master Mix (Vazyme Biotech). All samples were normalized to β-actin expression as the endogenous control. At least three biological replicates were included for each condition. The primer sequences for PCR were provided in Supplementary Table 4.
RNA sequencing and analysis
PC9 cells were genetically edited or treated as indicated. Total RNA was purified using the RNeasy Plus kit (Qiagen) following the manufacturer's instructions. RNA quality was assessed by NanoDrop 8000 (Thermo Fisher Scientific) and agarose gel electrophoresis. Sequencing libraries were prepared using the NEBNext Ultra RNA Library Prep Kit for Illumina (NEB). Clustering of the index-coded libraries on a cBot Cluster Generation System using TruSeq PE Cluster Kit v3-cBot-HS (Illumina), and the library preparations were sequenced on an Illumina Novaseq platform to generate 30 million 150 bp paired-end reads (Novogene). All the downstream analyses were based on the clean data which were obtained by removing low quality reads and reads containing adapters or ploy-N sequences. The index of the reference genome was built using Bowtie (v2.2.3) 34, and clean reads were aligned to the reference genome using HISAT2 (v2.0.5) 35. FeatureCounts (v1.5.0-p3) was used to count the reads numbers mapped to each gene 36. Differential expression analysis was performed using the DESeq2 (v1.20.0) R package 37. Transcripts with adjusted P values of < 0.05 were assigned as differentially expressed genes.
ATAC sequencing
The assay for transposase-accessible chromatin using sequencing (ATAC-seq) was performed as previously described 38. First, a total of 5 × 104 cells were harvested in ice-cold washing buffer (10 mM Tris-HCl pH 7.4, 10 mM NaCl, 3 mM MgCl2, 0.1% IGEPAL CA-630). Cell pellets were resuspended in Tn5 transposition reaction mix containing 25 µL 2× TD Buffer, 2.5 µL Tn5 Transposes (Illumina) and 22.5 µL nuclease-free water, and incubated at 37°C for 30 min. DNA was purified with MinElute PCR Purification Kit (Qiagen). Eluted DNA was barcoded and amplified using NEBNext Q5 Hot Start HiFi PCR Master Mix (#M0543L, New England Labs) in a total volume of 50 µL with the following PCR program: 72°C for 3 min; 98°C for 30 s; 12 cycles of 98°C for 30 s, 63°C for 30 s and 72°C for 3 min; 72°C for 5 min. The libraries were purified with QIAquick PCR Purification Kit (Qiagen) and sequenced on the Illumina HiSeq with 50-bp paired-ends. Reads were aligned to the human reference genome (GRCh37/hg19) using BWA with standard parameters 39, and filtered for high-quality (MAPQ ≥ 13), non-mitochondrial, and properly paired reads (longer than 18 nt). MACS2 (v 2.2.6) was used to call peaks using the parameters "-q 0.05 -g hs --nomodel --shift − 75 --extsize 150" 40. Peaks with an FDR of lower than 5% were saved to detect chromosomal regions for further analyses. Bigwig files were generated from the filtered BAM files using deepTools2 bamCoverage with the options "--normalizeUsing RPKM" 41. Peak annotation was performed with HOMER annotatePeaks.pl 42. Heatmaps and average profile plots were generated using deepTools plotHeatmap. Coverage tracks were visualized using the trackViewer R package 43.
CUT&Tag analysis
Cleavage under targets & tagmentation (CUT&Tag) was performed using the Hyperactive In-Situ ChIP Library Prep Kit for Illumina (TD901, Vazyme Biotech) following the manufacturer’s instructions. Briefly, cells were collected into a low absorbing tube, washed twice in 500 µL Wash Buffer, and incubated with concanavalin A-coated magnetic beads for 10 min at RT. The bead-bound cells were resuspended in 50 µL Antibody Buffer, followed by incubation with 1 µg of primary antibody at 4°C overnight with slow rotation. The supernatant was removed, and cells were incubated with secondary antibody in Dig-wash Buffer for 1 h at room temperature. Cells were washed three times with Dig-wash Buffer and incubated with 0.04 µM Hyperactive pG-Tn5 Transposase adapter mix prepared in Dig-300 buffer. Cells were washed three times with Dig-300 buffer to remove unbound pG-Tn5 and resuspended in 300 µL Tagmentation Buffer to incubate for 1 h at 37°C.The reaction was terminated, and DNA was extracted using phenol-chloroform-isoamyl alcohol method. PCR was performed to amplify the libraries under the following cycling conditions: 72°C for 5 min, 98°C for 30 s, 20 cycles of 98°C for 10 s, and 60°C for 30 s, followed by a final extension at 72°C for 1 min and holding at 4°C. PCR products were then purified with DNA Clean Beads. After quantification and quality control, DNA libraries were sequenced on an Illumina NovaSeq platform. The following antibodies were used: anti-rabbit IgG (#2729, Cell Signaling Technology), YY1 (ab109237, Abcam). Peaks were called using SEACR (v1.1) and MACS2 (v2.2.6) 40, 44. The subsequent analyses were same as the ATAC-seq pipeline.
In vivo studies
To evaluate the function of YY1 in vivo, 1 × 106 PC9 cells with YY1 genetic editing were mixed with Matrigel (BD Biosciences) and subcutaneously implanted in the dorsal flank of BALB/c nude mice (5 weeks of age). When tumor sizes reached approximately 200–300 mm3, animals were randomized into vehicle and treatment groups (8 mice each). For treatment of tumor-bearing mice, erlotinib was given at a dose of 5 mg/kg/day by gavage. Tumor volumes were recorded by blind measurement with a caliper and calculated as length × width2 × 0.52. The institutional animal care and use committee of Ren Ji Hospital approved all animal protocols.
Statistical analysis
The sequencing data were deposited in the NCBI BioProject database under the accession number PRJNA973187. The TCGA data were downloaded from the UCSC Xena Explorer and processed as previously described 45. The Kaplan-Meier method with log-rank test was used for survival analysis. The pathway enrichment analysis of differentially expressed genes was performed using the clusterProfiler R package (v3.18.0) and Metascape (http://metascape.org) 46. Gene set enrichment analysis was performed using the GSEA software (v4.1.0). In all experiments, comparisons between two groups were based on two-sided Student’s t tests. Pearson’s correlation coefficient was used to measure the linear correlation between two variables. Graphics and statistics were generated using GraphPad Prism (v8.0) or R (v4.1.0). P values of < 0.05 were considered statistically significant.