Data collection
The GSE98126 dataset was obtained from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/). Two glioma transcriptomic datasets, mRNAseq_325 (consisting of 325 cases) and mRNAseq_693 (693 cases), alongside their clinical data (survival time and status), were retrieved from the Chinese Glioma Genome Atlas (CGGA) at http://www.cgga.org.cn/index.jsp. In addition, a dataset of 698 glioma transcriptome sequences (combining TCGA-LGG and TCGA-GBM) and their respective clinical data were obtained from The Cancer Genome Atlas (TCGA) database. Notably, both CGGA and TCGA are public databases. Therefore, the information extracted is exempt from further ethical approval.
Data analysis
Differential gene expression within the transcriptome sequencing raw count data was analyzed using the DESeq2 package. To identify differentially expressed genes, we adopted the criteria of an adjusted P-value < 0.05 (Pval.adj < 0.05) and log2-fold change > 1 (|log2(FC)| > 1). Univariate Cox proportional hazards regression analysis was performed using the survival package. A P-value < 0.05 indicates a significant correlation between the gene and prognosis. The univariate Cox proportional hazards regression analysis results were further refined using least absolute shrinkage and selection operator (LASSO) regression analysis via the glmnet package. The LASSO regression outcomes were used to construct a multivariate Cox proportional hazards model. The formulated model was used to compute patient risk scores, using the equation: riskscore = Σ(βi * Xi), where Xi represents the expression value of a specific gene, and βi signifies the estimated value of the corresponding gene. Risk scores were used to categorize patients into low- and high-risk groups based on the median values of these scores. Additionally, the survival package was used for survival analysis, with a P-value < 0.05 indicating statistically significant differences between the survival curves of the high- and low-risk groups. The pheatmap package was used for heatmap visualization.
Specimens and cell culture of GBM
Brain tumor samples were obtained from consenting patients diagnosed with gliomas. From 2016 to 2021, the Department of Neurosurgery at Zhujiang Hospital collected 40 glioma samples preserved in paraffin, along with their corresponding clinicopathological information, from patients who underwent surgical procedures.
The GBM tissues underwent enzymatic digestion, and the GBM stem cells were cultured in Dulbecco’s modified Eagle medium (DMEM)/F12 (Gibco, USA) supplemented with epidermal growth factor (20 ng/ml, Peprotech, Cranbury, NJ, USA), basic fibroblast growth factor (20 ng/ml, Peprotech), and B27 (1:50, Gibco). GSC expansion was performed using both adherent and suspension culture methods. GSCs were placed in a laminin coated flask (10 μg/ml, Gibco) in an adherent culture system and grown in serum-free medium. In the suspension culture system, stem cell spheres were generated and propagated, as previously described [30]. GSCs from passages 2–10 were used. To obtain DGCs, GSCs were cultured in DMEM supplemented with 10% fetal bovine serum (Gibco). The cells were incubated at 37 °C with 5% CO2 and refreshed every 3 months using frozen reserves.
Immunohistochemistry and immunofluorescence staining
Tumor tissue samples from patients with glioma and xenograft samples of glioma were preserved using 4% formalin, embedded in paraffin, and then cut into 4 μm sections. After deparaffinization and dehydration, tissue sections were incubated in 3% hydrogen peroxide for 10 min, blocked with 5% bovine serum albumin (BSA) in phosphate buffered saline (PBS) for 1 h at room temperature, and the slides treated with primary antibodies against ABCB4 (Thermo Fisher Scientific, Waltham, MA, USA; PA5-78692), Ki67 (Abcam, Cambridge, UK; ab15580), and cleaved-caspase-3 (GeneTex, Irvine, CA USA; GTX86952) at 4 °C overnight. Secondary antibodies were added and incubated for 1 h at room temperature. DAB staining was used to detect specific molecules and the slides counterstained with hematoxylin.
Immunofluorescence staining was performed using primary antibodies against CD133 (CST, Shanghai, China; #64326), Nestin (CST; #33475), and ABCB4 (Thermo Fisher Scientific; PA5-78692). The immunostaining was carried out overnight at 4 °C, followed by incubation with fluorochrome-conjugated antibodies. Finally, DAPI was used to stain the cell nuclei. A fluorescence microscope or laser scanning confocal microscope (Nikon, Japan) was used to capture images.
Sphere formation assay
Individually separated GSCs were placed in 24-well dishes and incubated in serum-free medium at 37 °C for 7 days. A fluorescence microscope (Leica, Germany) was used to capture images of five randomly chosen regions from each group.
Extraction of RNA and quantitative reverse transcription polymerase chain reaction (qRT-PCR)
Total RNA was extracted from glioma cells using TRIzol reagent (Invitrogen, Waltham, MA, USA). An miRNeasy Mini Kit (Qiagen, Hilden, Germany) was used to extract RNA from the exosomes. The PrimeScriptTMRT reagent kit (TaKaRa, Shiga, Japan) was used to convert 1 μg total RNA into cDNA, with the assistance of the gDNA Eraserkit. For realtime-PCR, a SYBR®Premix Ex TaqTM Kit (Tli RNaseH Plus; TaKaRa) was used. A LightCycler 480 quantitative PCR instrument (Roche, Indianapolis, IN, USA) was used. Relative RNA expression was determined using the ΔΔCt method. Sangon Biotech Ltd. (Shanghai, China) provided the primers (Supplementary Table S1).
Western blot (WB) analysis
A Whole Cell Lysis Assay (KeyGEN BioTECH, Nanjing, China) was used to extract both total and exosome proteins. Protein extracts were isolated using 8–12% sodium dodecyl-sulfate polyacrylamide gel electrophoresis and subsequently transferred onto polyvinylidene fluoride membranes (Millipore, USA). Following the addition of 5% BSA (Sigma-Aldrich, St Louis, MI, USA), the membranes were incubated with primary antibodies (Supplementary Table S2) for 12 h at 4 °C. The membranes were then incubated with secondary antibodies conjugated to horseradish peroxidase for 1 h at ambient temperature. Enhanced chemiluminescence (ECL; Millipore) was used to visualize the protein bands. ImageJ software was used to analyze the strength of the protein bands, which were normalized to those of GAPDH. The full, uncropped western blots can be found in the Supplemental Material (Fig. S5).
Infection with lentiviruses and transfection with siRNA
A lentivirus containing ABCB4 short hairpin RNA was constructed by Hanbio (Shanghai, China). The cells were cultured with the lentivirus and polybrene for 24 h. Stable control and specific knockdown cells were selected and maintained in the presence of puromycin (2 μg/mL; Solarbio, Beijing, China). Hanbio provided the siRNA for ATF3 and negative control sequences, which were then transfected using the lipofectamine® 3000 reagent (Cat# L3000015; Thermo Fisher Scientific).
Drugs
TMZ powder (Sigma-Aldrich) was dissolved in dimethyl sulfoxide (Sigma-Aldrich) to a final concentration of 100 mM.
Assessment of cellular viability
Cell viability was determined using a CCK-8 assay. Cells were seeded in a 96-well dish at 3 × 103 cells/well and incubated for 24 h. Following exposure to various concentrations of TMZ for 48 h, or incubation in 200 μM TMZ for several days, CCK-8 solution (Dojindo, Kumamoto, Japan) was added. Cell viability was measured at 450 nm using a microplate reader (BioTek, Seattle, WA, USA).
Flow cytometry
Apoptosis was detected using the Annexin V-FITC kit (KeyGEN BioTECH). Cells were exposed to TMZ (200 μM) for 48 h and subsequently suspended in a binding buffer that included annexin V and propidium iodide. Following a 15-min incubation in the absence of light, the apoptosis rate was assessed using a FACSCalibur device (BD Bioscience, USA).
In vivo xenograft assay
Male nude BALB/c mice, aged 5–8 weeks, were acquired from the Central Animal Facility at Southern Medical University. The study protocols were approved by the Animal Care and Use Committee of the Southern Medical University. BALB/c nude mice were stereotactically injected with GSC-Luc stably transfected with Lentivirus-shABCB4 (ABCB4 KD) or lentivirus-vector control (Ctrl KD) into the right hemicerebrum. Each treatment group consisted of 15 mice. Starting on the seventh day post-transplantation, mice carrying the tumor were administered TMZ at 20 mg/kg/day, 5 days/week, for three cycles. Tumor growth was observed using an in vivo imaging device (IVIS Lumina II, USA) following administration of the luciferase substrate D-luciferin (Yeasen, Shanghai, China) via intraperitoneal injection. After 4 weeks, seven nude mice were euthanized in each group. Brains were collected and immunohistochemically stained. The survival curves were plotted for the remaining mice.
Co-culture assay
GSCs and DGCs were co-cultured in a Transwell plate (Corning, Corning, NY, USA) with a 0.4-μm polycarbonate filter for 48 h at a 1:1 ratio. The upper chamber contained GSCs, whereas the lower chamber contained DGCs.
Exosome isolation
The culture medium of GSCs and DGCs was centrifuged at 4 °C for 10 min at 300 ×g, and the pellets were aspirated. After recentrifugation at 4 °C at 2000 ×g and 5000 ×g for 15 min each, the pellets were removed. The liquid was then centrifuged at 12,000 ×g for 30 min, and the liquid collected. Next, the suspension underwent ultracentrifugation at 4 °C at 100,000 ×g for 70 min (Beckman Coulter, Brea, CA, USA), to collect the exosome pellets. The exosomes were washed with sterile PBS and purified by centrifugation at 100,000 ×g for 1 h, then resuspended in 100 μL PBS and stored at −80 °C.
Exosome characterization
Exosome morphology was analyzed using transmission electron microscopy (TEM, Hitachi HT7650, Japan). To determine the size distribution of the isolated exosomes, a Zetasizer Nano-Zs (Malvern Panalytical, Malvern, UK) was used in accordance with the manufacturer’s guidelines. A bicinchoninic acid (BCA) protein assay kit (KeyGEN BioTECH) was used to determine the concentration of the isolated exosomes according to the manufacturer’s instructions. To examine the protein indicators of exosomes, WB was performed using antibodies against CD9, ALIX, CD63, and calnexin.
Exosome uptake assay
GSC exosomes were labeled with fluorescent PKH26 using a cell linker kit (Sigma-Aldrich) to observe exosome trafficking. The exosomes labeled with PKH26 were rinsed with PBS, centrifuged at 100,000 ×g for 20 min at 4 °C, and the exosomes suspended in PBS. After labeling with PKH26, the exosomes were co-cultured with DGCs for 24 h, and a confocal fluorescence microscope used to observe the absorption of exosomes by DGCs.
Luciferase reporter assay
Before transfection, 1 × 105 GSCs were seeded in a 24-well dish. The following day, cells were co-transfected with the control pcDNA3.1 empty vector (pcDNA3.1, 225 ng) or ATF3 plasmid (225 ng)-based firefly luciferase reporter and the Renilla luciferase control vector pGL4.73 (50 ng; Promega, Madison, WI, USA) using the K-2 transfection reagent (Biontex, München, Germany). Following 48-h incubation, the Renilla levels and firefly luciferase activities were assessed in accordance with the manufacturer’s guidelines (Promega). Relative values were obtained by normalizing firefly luciferase values to Renilla luciferase values.
Chromatin immunoprecipitation (ChIP)
Following the cross-linking and lysis of cells, DNA fragments were sonicated on ice to achieve lengths ranging from 200–1000 base pairs. Protein-DNA complexes were precipitated using an Anti-ATF3 antibody (CST; #33593) and control IgG. Procedures were performed in accordance with the Pierce Agarose ChIP Kit guidelines (Thermo Scientific). Immunoprecipitated DNA and the corresponding input DNA were subjected to reverse cross-linking at 65 °C overnight. This DNA was then used in a qPCR assay with the SYBR Green qPCR mix to investigate the potential ATF3-binding locations in the ABCB4 promoter, employing specific primers (Fig. S4).
Statistical analyses
GraphPad Prism 6 (GraphPad Software Inc., Boston, MA, USA) was used for all statistical analyses. The data is displayed as the average ± standard error from three separate trials. Statistical significance was evaluated using either Student's t-test or one-way analysis of variance with Bonferroni correction to account for multiple comparisons. Survival rates were calculated utilizing the Kaplan–Meier technique, and variations in mortality were assessed through the log-rank test. Significance was determined for all tests using a two-sided approach, with P-values < 0.05 considered significant.