Cell Culture
Human gastric cancer cell lines SGC7901, SNU719, AGS were provided by Nanjing Kegen Biotechnology Co., Ltd. The cells were cultured in RPMI-1640 medium containing 10% fetal bovine serum and 1% penicillin and streptomycin, placed in a humidified incubator containing 5% CO2 and 95% air, subculture and all experiments were performed at 37 °C. AIL stock solution was prepared with DMSO, stored at 4 °C, and diluted with RPMI-1640 medium to the required concentration immediately before use; the final concentration of DMSO in the medium was 0.1%. Cells in the control group were treated with DMSO (0.1%) without AIL.
Human tissues and organoids
Human gastric cancer tissues were taken from patients who underwent gastric cancer surgery in the First Affiliated Hospital of Sun Yat-sen University. They agreed and signed a donation and research consent form. It was approved by the clinical scientific research and animal experiment ethics committee of the First Affiliated Hospital of Sun Yat-sen University (Lunshen [2018] No. 087). This research complied with all the human participation ethics in research. Organoid related experiments were completed in the Digestive Diseases Center of the Seventh Affiliated Hospital of Sun Yat-sen University. Organoid strains from a total of 3 patients were used.
After the tumor was excised, it was placed in 50 U/ml penicillin-streptomycin (Thermo Fisher) frozen G solution. The tissue was minced on ice and incubated in DMEM containing 1 mg/ml collagenase V (Sigma-Aldrich) for 1 hour at 37 °C. Iced PBS was added to stop the digestion, and subsequently centrifuged at 4 °C (300 rcf, 5 min). The sample was further digested with TrypLE (Thermo Fisher) at 37 °C for 5 min, and then stopped with a large amount of PBS. The suspension was filtered through 40 nylon meshes, centrifuged, and the cells were fixed in the matrix. It was then passaged with TrypLE every 2 weeks. The medium for establishing and culturing human GC organoids was as described in the literature[20].
Colony Formation Assay and Cell Viability
AGS, SNU719, and SGC7901 cells were seeded in 6-well plates with 500 cells per well, and the experiment was repeated three times. AIL was added to the cells at different final concentrations (0, 0.05, 0.1 µM) and it was then incubated for 12 days. The cells were fixed for 30 minutes with 4% paraformaldehyde, stained with crystal violet for 30 minutes, and finally counted.
In a 96-well transparent bottom black board, 3,000 cells were seeded into each well (organoids were seeded in Matrigel). Medications were added in each well according to a 5-fold or 10-fold concentration gradient. After 72 hours, a luminometer (PerkinElmer Life and Analytical Sciences, Boston, MA) was used to determine the level of adenosine triphosphate (ATP) by CellTiter-Glo Luminescent Cell Viability Assay (Promega, Madison, WI).
Cell migration and invasion assay
Transwell migration assay and matrix invasion assay were used to determine cell migration and invasion ability. In the Transwell migration assay, a small chamber (8 µm pore size; Corning) was placed in a 24-well plate, and then 5 × 104 cells were suspended in 200 ul serum-free RPMI-1640 medium in the chamber; 20% pre-warmed fetal bovine serum was used in the well.The cells were incubated for a period of time (AGS/16 hours, SNU719/24 hours, SGC7901/42 hours) in 5% CO2 at 37 °C, and then fixed with 4% paraformaldehyde. In the matrix invasion assay, matrix gel and serum-free 1640 medium were injected into the transwell chamber at a ratio of 1:8 for pretreatment for 2 hours. 1 × 105 cells was suspended in 200 µL of serum-free RPMI-1640 medium, and pre-warmed medium containing 20% fetal bovine serum was then added to the well. The cells were incubated for a period of time (AGS/16 hours, SNU719/24 hours, SGC7901/42 hours) in 5% CO2 at 37 °C, and then fixed with 4% paraformaldehyde. A cotton swab was used to gently remove cells that have not migrated or invaded the membrane. Cells were stained with 0.1% crystal violet (Sigma, St Louis, MO), and cells in 8 randomly selected field of vision were counted under a light microscope (100x magnification) to determine cell migration or invasion.
The PDX mouse model
In vivo experiments had been performed according to the Institutional Animal Care and Use Committee (IACUC). It was approved by the clinical scientific research and animal experiment ethics committee of the First Affiliated Hospital of Sun Yat-sen University (Lunshen [2018] No. 087). The experiment was conducted in the Animal Center of the First Affiliated Hospital of Sun Yat-sen University.
Balb/c nude mice (female, 8 weeks old, 19–21 g in mass) were purchased from GemPharmatech Co., Ltd (Nanjing, Jiangsu) Experimental Animal Co., Ltd. and raised in an SPF environment. PDX method was as mentioned before[21].When the tumor volume reached about 150 mm3, they were randomly divided into three groups, and different reagents were given by intraperitoneal injection (IP). The tumor volume and body mass were measured every 3 days. The calculation formula of tumor volume is as follows: V = Πa b2/8 (where V is the tumor volume, a is the largest tumor diameter, and b is the smallest tumor diameter). Nude mice were sacrificed on day 30, and tumors were taken for measurement and weighing.
Biochemistry, histology and immunohistochemical staining
In order to evaluate the toxicity of Ailanthone, mice with subcutaneously transplanted tumor were sacrificed at the end of the experiment, and blood serum, liver and kidney were harvested. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured. Tumor or mouse tissue samples were immediately fixed in 10% neutral buffered formalin for 24 hours, gradually dehydrated in a solution with increasing ethanol content (75, 85, 95 and 100%, v/v), and finally embedded in paraffin blocks. Liver and kidney were fixed by paraffin-embedded formalin and cut into 3 mm sections. H&E staining was performed by the experimental histopathology laboratory in accordance with standard procedures. Observations were made under light microscope (100X magnification). Immunohistochemical (IHC) staining: Collect tissues after fixation, dehydration, embedding and sectioning. After heat-mediated antigen repair in 10 mM sodium citrate buffer (pH 6.2) or Tri-EDTA, 1.6% hydrogen peroxide was used to block endogenous peroxidase. Ki67(1: 1000, Abcam, ab15580), H2AX (1: 100, Proteitech, 10856-1-AP), Caspase3 (1:200, Proteitech, 19677-1-AP), XRCC1 (1: 100, Abcam, ab44830), P23 (1: 200, Proteitech, 10824-1-AP), HSP90 (1: 400, Proteitech, 60318-1-Ig, Mouse), and AKT (1: 500, Proteitech, 10176-2-AP) were used as a primary antibody for immunohistochemistry tests according to the protocol. The samples were stained with hematoxylin (HE) and eosin to indicate the nucleus and cytoplasm, respectively. DAB staining was performed and positive cells were counted in 5 random field of views per slide.
Quantitative real-time PCR
AGS, SNU719, and SGC7901 cells were sown in 6-well plates and incubated with different concentrations of AIL for 12 hours. AG RNAex Pro reagent (Accurate Biology, CAT#AG21102) was used to extract total RNA from tissue samples or cell lines. Evo M-MLV reverse transcriptase premix (Accurate Biology CAT#AG11706) was used to synthesize cDNA from 2 µg RNA of each sample. SYBR Green Premix Pro Taq HS qPCR kit (Accurate Biology, CAT# AG11701) was used for qRT-PCR. Each sample was tested 3 times. The data was analyzed using the 2-ΔΔCT calculation method. The primers were as follows:
GAPDH forward 5’-GACCCCTTCATTGACCTCAA-3’;
GAPDH reverse 5’-TGCTTCACCACCTTCTTGAT-3’;
HSP90 forward: 5’-CATAACGATGATGAGCAGTACGC-3’;
HSP90 reverse: 5’- GACCCATAGGTTCACCTGTGT-3’;
P23forward:5’- GAAAGCACAGTAATCACTGGTGT-3’;
P23 reverse:5’- ACGGTAGTCCAATAGAGCAACC-3’;
XRCC1 forward: 5’- TCAAGGCAGACACTTACCGAA-3’;
XRCC1 reverse:5’-TCCAACTGTAGGACCACAGAG-3’.
Western blotting
The cells were processed differently according to their corresponding requirements, and the protein concentration is determined by BCA Protein Assay Kit (KGI Biosciences), using SDS-PAGE Sample Loading Buffer, 5X (Beyotime) at 95° for 8 minutes. The lysate is separated on a polyacrylamide gel and transferred to nitrocellulose. The blot was detected with a specific antibody, and then the membrane was detected with the LI-COR Odyssey CLx Infrared Imaging System (LI-COR Biotechnology, Lincoln NE). Antibodies were H2AX (1: 1000, Proteintech, 10856-1-AP), XRCC1 (1: 1000, Abcam, ab44830), Caspase3 (1: 400, Proteintech, 19677-1-AP), P23 (1: 500, Proteintech, 10824-1-AP), AKT (1: 2000, Proteintech, 10176-2-AP), HSP90 (1: 1000, Proteintech, 60318-1-Ig), Tubulin (1: 1000, Proteintech, 10068-1- AP), GAPDH (1: 20000, Proteintech, 60004-1-Ig).
Immunoprecipitation
The protein expression level was determined by the western blotting results of cytoplasm or nuclear lysate. Nuclear and Cytoplasmic Protein Extraction Kit (Beyotime, CAT#P0028) was used to prepare cytoplasmic lysates and nuclear lysates, and protein concentration was measured. 2,000 µg of protein lysate (prepared 500 µL) was mixed with primary antibody and incubated overnight in a 4° shaker. 50 µL of protein A/G agarose (Thermo Fisher Scientific, CAT#20424) beads was washed 3 times with lysate, mixed in the above-mentioned protein mixture with the primary antibody and incubated overnight in a 4° shaker. The mixture was washed 6 times with lysis buffer (Thermo Fisher Scientific, CAT#87787) before SDS-PAGE Sample Loading Buffer was added, and then heated to 95° for 10 min at 5X (Beyotime), centrifuged at 3,000 rpm for 3 min, the supernatant was carefully aspirated and the protein level was finally detected via western blotting. Antibodies used were: XRCC1 (1: 100, Abcam, ab44830), P23 (1: 200, Proteintech, 10824-1-AP), HSP90 (1: 400, Proteintech, 60318-1-Ig), AKT (1: 400, Proteintech, 10176-2-AP).
Immunofluorescent(IF) staining
The collected organoids were fixed for 16–20 hours, then embedded, sliced, baked, and deparaffinized. After heat-mediated antigen retrieve in 10 mM sodium citrate buffer (pH 6.2) or Tri-EDTA, 1.6% hydrogen peroxide was used to quench endogenous peroxidase, then blocked with PBS containing 5% BSA and 0.2% Triton- X. The primary antibody was incubated in blocking buffer at 4 °C for 16–20 hours. The fluorescent secondary antibody was incubated for 1 hour at 20 °C, and then incubated in DAPI for 10 minutes. Fluorescence staining was imaged on a Zeiss LSM 780 confocal microscope. Primary antibodies used were: Ki67 (1: 1000, Abcam, ab15580), H2AX (1: 400, Proteintech, 10856-1-AP), Caspase3 (1: 400, Proteintech, 19677-1-AP ), XRCC1 (1: 100, Abcam, ab44830), P23 (1: 200, Proteintech, 10824-1-AP), HSP90 (1: 400, Proteintech, 60318-1-Ig), AKT (1: 400, Proteintech, 10176-2-AP). Secondary antibodies used were: Donkey anti-Rabbit /Alexa Fluor 488 (1:1000, Thermo Fisher Scientific, A-21206), Donkey anti-Rabbit /Alexa Fluor 546 (1:500, Thermo Fisher Scientific, A11040), Donkey anti-Mouse /Alexa Fluor 488 (1:000, Thermo Fisher Scientific, A-21202), Donkey anti-Mouse/Alexa Fluor 546 (1:500, Thermo Fisher Scientific, A10036).
RNA isolation and microarray
Total RNA was extracted from tissue samples, the concentration and purity of the extracted RNA were detected by Nanodrop 2000, RNA integrity was detected by agarose gel electrophoresis, and RIN value was determined by Agilent 2100. A single library construction requires that the total amount of RNA is not less than 5 µg, concentration ≥ 200 ng/µL, and the OD260/280 is between 1.8 and 2.2. mRNA capture and library preparation were completed by the advanced sequencing equipment of Shanghai Origingene Bio-pharm Technology Co., Ltd. using KAPA mRNA HyperPrep kit (Roche). Three biological library was sequenced on the Illumina Truseq TM RNA sample prep Kit platform of the facility, and each sample produced an average of 25 million single-ended reads of 75 bp. The high-quality DNA sequencing obtained after quality control was compared with the designated reference genome. For the PDX sample, it was first compared to the mouse reference genome. After eliminating the mouse data, the remainder was then compared to the human reference genome retrieved from Ensembl database (genome version GRCh38, gene annotation information Ensemble 92). Before alignment, cutadapt (version 1.9.1) was used for quality trimming and adaptor removal of the original reading. Using the annotation release 86 as a reference, read was sequenced on human genome GRCh38 using RSEM 1.3.0 and STAR 2.5.2, and the subsequent gene levels were counted. In R program (version 3.6.1), the DESeq2 package (version 1.24.0) was used for normalization and differential expression analysis of raw count data. Regularized logarithmic transformation was performed on the rlog function.
Gene Set Enrichment Analysis (GSEA)
GSEA was performed with the software (GSEA V4.0.3) developed by the Broad Institute of Massachusetts Institute of Technology and Harvard University (https://www.gsea msigdb.org/gsea/index.jsp). For the RNA-seq datasets of low and high expressionof P23, OXA group and AIL group, the normalized RNA read count was used for analysis, and the following settings were applied: number of permutations = 1000, type of permutation = gene set, enrichment statistics = weighting, measurement of gene ranking = signal noise. For the TCGA gastric cancer data set, the samples were grouped according to their expression above or below the median value. The normalized RSEM read count was used for analysis, and the following settings were applied: number of permutations = 1000, permutation type = phenotype, enrichment statistics = weighting, measurement of gene ranking = signal 2 noise. Recognized marker gene set 40, KEGG pathway or gene ontology (GO) terms and false discovery rate (FDR q) < 0.05 were set as significant enrichment.
Screening of differentially expressed genes (DEGs)
The expectation-maximization algorithm of RNA-Seq was used to normalize the 3-level transcriptome data of the data set, and the logarithmic transformation of all gene expression values was performed. Approximate data were normalized by quantiles and were normally distributed[22]. In this study, the R program package limma v3.28.14 was used to analyze the differential genes in the gene expression data, and its mRNA satisfied P < 0.01, false discovery rate (FDR) was < 0.01 and |log2 fold change (FC)| was > 1.5, where P < 0.05 indicated that the hypothesis test was statistically significant. FDR is a control indicator for the error rate of the hypothesis test. As an evaluation index of the selected differential genes, the number of false rejections was proportional to the number of null hypotheses rejected. FC is usually used to describe the degree of change from the initial value to the final value. In this study, the ratio of tumor tissue gene expression value to normal tissue gene expression value was used, also known as the fold change. The heatmap and volcano map of the differential genes were constructed in R language for visual comparison.
Flow cytometry and FACS
Flow cytometric analysis: Single cells prepared from organoids was suspended in PBS containing 10% FBS. The single cell suspension was stained with a fluorescently-labeled isotype control antibody or experimental antibody in the dark at 4 °C for 30 minutes, and washed three times in FACS buffer. The cells were filtered through a 40-HCLM nylon mesh and incubated in 3 HCLM DAPI in FACS buffer. Single-color stains and Fluorescence Minus One control were used when necessary. Annexin V-PI apoptosis assay and cell-cycle analysis were performed using Annexin V-FITC apoptosis detection kit (KeyGEN BioTECH, KGA105) and apoptosis detection kit (KeyGEN BioTECH, KGA512) according to the manufacturer's protocol. Cells were sorted and analyzed by CytoFlex LX (Beckman). FlowJo 10 software was used to analyze the data.Flow sorting: After the prepared organoid suspension single cell was fixed (Cytofix/Cytoperm Soln Kit, CAS: 554714), P23 was added (1:100, Thermo Fisher Scientific, MA3-414) and incubated at 4° for 30 min, then washed three times in FACS buffer, incubated with the fluorescent secondary antibody Donkey anti-Rabbit /Alexa Fluor 488 (1:200, Thermo Fisher Scientific, A-21206) for 15 minutes, and lastly dead cells were excluded after staining with DAPI. The cells were sorted on a SH800S (SONY) flow cytometer, and P23(+) and P23(-) cell populations of the GC organoid strains were separated by flow cytometry. The cells were collected in cold PBS supplemented with 10% fetal bovine serum, and the final sorted cell population was subjected to other experiments.
Clinical gastric cancer samples
The sample data were procured from the prospectively collected GC database of the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China. We reviewed the treatment records of patients undergoing gastric cancer resection from January 2010 to May 2012. The inclusion criteria were pathological TNM stages I–III gastric adenocarcinoma patients whom underwent radical surgery patients. Exclusion criteria: other history of malignant tumors, neoadjuvant therapy, loss to follow-up, no tissue samples. A total of 93 cases of tissue samples were collected from surgical specimens, and informed consent was obtained from the patients. This study was approved by the Ethics Committee of the First Affiliated Hospital of Sun Yat-sen University. The detailed clinical characteristics of the patients were shown in Table 1. The endpoint of the study was recurrence. The specimen tissues were tested by IHC, and the results were interpreted by two independent pathologists. They were blinded to the specific diagnosis and prognosis of each case, and used a semi-quantitative method for scoring. Among the samples, more than 10% of tumor cells staining were positive. The staining intensity was divided into negative, weak, moderate and strong. Low P23 expression was detected in the negative and low intensity samples, and medium and strong intensity returned with high P23 expression. Subsequently, the disease-free survival of patients in the high P23 expression group (n = 42) and low P23 expression group (n = 51) were evaluated. Progression free survival is defined as the duration from tumor resection to progressive disease. Follow up was completed in intervals of 3 months (0–2 years), 6 months (2–4 years), and once a year until recurrence or June 2020. The follow-up study included abdominal computed tomography and postoperative physical examination.
Table 1
Research status of 8 medicinal plants
Medicinal plants | Tumor type | References |
Chrysin | Prostate Cancer,Ovarian Cancer | 365 |
Hesperidin | Cholangiocyte,Oral Carcinogenesis | 282 |
Ailanthone | Prostate Cancer,Lung Cancer | 25 |
Indirubin | Prostate Cancer,Promyelocytic Leukemia༌Glioma | 157 |
Oridonin | Fibrosarcoma, Diffuse Large B Cell Lymphoma༌ Leukemia | 261 |
Celastrol | Melanoma,Breast Cancer༌Prostate Cancer | 321 |
Ursolic acid | Colorectal Cancer,Breast Cancer༌Leukemia | 563 |
Pseudolaric Acid B | Glioma,Lung Cancer | 59 |
Wedelolactone | Prostate Cancer,pancreatic cancer | 45 |
Table 2
Characteristics of 93 recurrence gastric cancer patients
Characteristics | Low(51) | High(42) | p |
Sex | Female | 26 | 16 | 10 | 0.419 |
| Male | 67 | 35 | 32 |
Diferentiation | Well | 62 | 41 | 21 | 0.003 |
| Moderate | 26 | 7 | 19 |
| Poor | 5 | 3 | 2 |
pT stage | T1 | 22 | 13 | 9 | 0.870 |
| T2a | 13 | 6 | 7 |
| T2b | 28 | 14 | 14 |
| T3 | 21 | 13 | 8 |
| T4 | 9 | 5 | 4 |
pN stage | N1 | 58 | 32 | 26 | 0.946 |
| N2 | 25 | 14 | 11 |
| N3 | 10 | 5 | 5 |
pM stage | M0 | 84 | 46 | 38 | 0.851 |
| M1 | 9 | 5 | 4 |
Statistical analysis
SPSS 22.0 software was used for statistical analysis. Data were expressed as mean ± standard deviation (s.d.), and P < 0.05 was considered statistically significant. Tumor volume was measured repeatedly using a general linear model. The difference between categorical variables was tested using χ2 test, and the difference between two groups was tested using student’s t test. Kaplan-Meier curve and log-rank test were used to analyze the clinical data of gastric cancer patients.