Clinical samples
Three pairs of matched prostate tumor, normal prostate tissue, and fresh bone marrow (BM) blood of AML patients were obtained from Huai’an First People’s Hospital with patients’ informed consent. Fresh tissues were divided into several parts for protein and RNA isolation. Mononuclear cells (MNCs) were isolated from BM blood samples using Ficoll Paque Plus (GE Heathcare, 17-1440-02-1) for further studies. All samples used in this study were approved by medical ethics committee of Huai’an First People’s Hospital, and China Pharmaceutic University. Clinicopathologic information of each patient was listed in Supplementary Table S1.
Cell Culture
All cell lines used in this study were cultured in recommended culture medium at 37°C in 5% CO2. Sources and culture mediums of these cell lines were detailedly listed in Supplementary Table S1. All cell lines were detected no mycoplasma contamination annually and had valid STR profile. MNCs isolated from BM blood of AML patients were cultured in IMDM medium supplemented with 20% FBS, 10 ng/mL human cytokines IL-3, IL-6, SCF, TPO, and FLT3 ligand (all from Sino Biological).
Bioinformatics data mining and analysis
Expression data and clinical data of 497 PRAD patients in the Cancer Genome Atlas (TCGA) database were downloaded from the cBioPortal platform (https://www.cbioportal.org). Additionally, we downloaded expression data of 51 normal prostate tissue samples from the Genomic Data Commons (GDC, https://portal.gdc.cancer.gov/). To explore the prognostic value of ADAR1, we assigned PRAD patients into three subgroup, ADAR1-high (top 10%), ADAR1-intermediate, and ADAR1-low (bottom 10%), based on the expression levels of ADAR1. All statistical analyses were performed using R software (version 3.6.3).
Synthesis of ZYS-1
To a solution of 2-fluoroadenosine (5.70 g, 20 mmol) and pyridine (3.22 mL, 40 mmol) in acetonitrile (80 mL) was added drop-wise SOCl2 (7.25 mL, 100 mmol) at 0 ºC. The reaction mixture was continuously stirred for 4 h, and then subsequently warmed to room temperature for overnight. The resulting suspension was concentrated in vacuo. To the reaction mixture was added methanol (120 mL), water (12 mL), and NH4OH (24 mL), followed by stirring for 0.5 h at r.t. The reaction mixture was concentrated and the solid was precipitated in the water phase, and the filter cake was obtained by filtration. The filter cake was re-dissolved in a small amount of methanol at 60 ºC, and dichloromethane was added dropwise. The solid was precipitated by cooling, filtered and washed with cold methanol to obtain the product ZYS-1 as a white solid (5.45 g, 90%). 1H NMR (400 MHz, DMSO-d6) δ 8.34 (s, 1H), 7.92 (s, 2H), 5.84 (d, J = 5.6 Hz, 1H), 5.58 (s, 2H), 4.67 (t, J = 5.4 Hz, 1H), 4.26–4.16 (m, 1H), 4.10 (q, J = 5.4, 5.0 Hz, 1H), 3.99–3.81 (m, 2H). 13C NMR (101 MHz, DMSO) δ 158.61 (d, J = 206.4 Hz,), 157.70 (d, J = 19.3 Hz), 150.75 (d, J = 19.7 Hz), 140.01, 117.58, 87.40, 83.74, 72.69, 71.17, 44.76.
Cell Proliferation Assays
Cell proliferations were evaluated using the Cell Counting Kit-8 (Share-bio, SB-CCK8). Briefly, 5000–20000 cells/well were seeded into 96-well plate and treated with several diluted concentrations of test compounds or DMSO as control in triplicates. After treatment for 72 h, 10 µL of CCK-8 solution was added into each well, and the absorbance at 450 nm was measured using a microplate reader (Bio-Tek SynergyH1) upon incubation for 4 h at 37°C. Data were fitted in nonlinear regression and IC50 values were calculated by GraphPad Prism 8.0. For cell growth assays, cell viabilities were determinated at 0, 24, 48, 72, and 96 h.
Western Blotting
Western blotting assay was performed similarly as described previously (63). Cells were lysed in RIPA lysis buffer (Thermo Fisher Scientific, 89901) at 4°C, and the lysate supernatant were added with protease inhibitor (Roche, 04693132001) and phosphatase inhibitor (Roche, 04906845001). Protein concentration of lysates was quantified using a BCA protein assay kit (Thermo Fisher Scientific, 23227). Equal amounts of protein were loaded onto 8–12% SDS-PAGE gel and transferred onto a polyvinylidene difluoride (PVDF) membrane, which was blocked with 0.5% skim milk in Tris-buffered saline supplemented with 0.5% Tween 20 (TBS-T) and then incubated with indicated primary antibodies. Flowing washing in TBS-T, membranes was incubated with horseradish peroxidase-conjugated (HRP) secondary antibody for 1–2 h. The blots were visualized by a chemiluminscent detection system (Tanon, Shanghai, China).
Cell migration and invasion assays
Each transwell (Falcon, 353097) was seeded with 5 × 104 cells which have been starved in FBS-free medium for 12 h. Then, cells were allowed to migrate or invade to the bottom chamber for at least 24 h. Cells in the bottom of the chamber were fixed in 4% paraformaldehyde (PFA) for 10 min and stained with crystal violet for 10 min. For invasion assay, upper chambers were precoated with matrigel (Corning, 356234) and incubated for 12 h at 37°C. Images were taken using an inverted microscope (Nikon) and cell counts were quantified using ImageJ software.
Colony formation assay
500–1000 cells/well were plated to 24-well plate and allowed for growth for 10–14 days, during which medium in wells were replaced every three days. 4% PFA was used to fix colony for 10 minutes, followed by staining with crystal violet for 10 minutes. Images were taken using an inverted microscope and colony numbers were quantified using ImageJ software.
Lentivirus transfection
Lentivirus particles of short-hairpin RNA against ADAR1 (pGV248-shADAR1) and its scrambled control (pGV112-shNC) were constructed and purchased from Genechem Co. Ltd. (Shanghai, China). For lentivirus transfection, 1–10 × 105 cells were seeded onto 6-well plate and allowed for adhesion for 24 h, followed by transfection with above lentivirus in the presence of HitransG P (Genechem, REVG005). After 16–24 h, medium containing virus was removed. Transfected cells were allowed for growth for 3–5 days, during which GFP fluorescence was observed every day, followed by treatment with 2 µg/mL puromycin for 24 h to select positive infected cells. All transfected cells were validated by qRT-PCR and western blotting, and maintained in medium containing 1 µg/mL puromycin. The lentiviral particles were used at 20 multiplicity of infection (MOI). The target sequences of the shADAR1 and shNC were as follows: shNC: 5′-TTCTCCGAACGTGTCACGT-3′; shADAR1 1#: 5′-GATACTACACCCATCCATT-3′; shADAR1 2#: 5′-AGGGTATGTTGACTTTGAA-3′.
Transient overexpression of MTDH, ADAR1 WT and ADAR1 Mut
All plasmids including pGV141-EV, pGV141-ADAR1 WT, pGV141-ADAR1 Mut, and pGV141-MTDH were constructed and purchased from Genechem Co. Ltd. (Shanghai, China). Briefly, 3 × 105 DU-145 cells were seeded onto 6-well plate and allowed for adhesion to 70% confluence. Each well was added 200 µL transfection complex in RPMI1640 medium containing 1 µg plasmid and 1 µL X-tremeGENE HP DNA Transfection Reagent (Roche, 06366236001). After 24–48 h transfection, expressions were validated by western blotting. The primers used for amplifying ADAR1 and MTDH cDNA by PCR were listed in Supplementary Table S2.
Cellular Thermal Shift Assay (CETSA)
CETSA was performed following the protocol by Huang et al (64). Cells were harvested and lysed in PBS containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, and 2 mM DTT after freeze-thawed three times using liquid nitrogen. Upon centrifugation, supernatant were added ZYS-1 or DMSO and incubated at 25°C. Subsequently, the supernatant were divided equally and denatured at various temperatures for 5 min. These samples were analyzed by SDS-PAGE and western blot. Relative protein band intensity was analyzed by Image J software and the apparent aggregation temperature (Tagg) was fitted and calculated by GraphPad Prism 8.0 software.
RNA-seq and Data Analysis
Cells were harvested and total RNA was extracted using Beyozol Total RNA Extraction Reagent (Beyotime, R0011) following the instruction of the manufacturer. Total mRNA was enriched by Obligo(dT) beads, then fragmented into short fragments and reversely transcribed into cDNA with random primers. After the second-strand cDNA was synthesized, cDNA fragments were purified, end repaired, added poly(A), and ligated to Illumina sequencing adapters. The ligation products were size selected by agarose gel electrophoresis, amplified by PCR, and sequenced using Illumina HiSeq2500 by Genedenovo Biotechnology Co., Ltd (Guangzhou, China). Raw reads were cleaned to remove adapters or low quality reads and rRNA mapped reads. Clean reads were mapped to Homo sapiens genome. Gene expressions were quantified by FPKM (fragment per kilobase of transcript per million mapped reads) value and differential expression analysis was performed by DESeq2 software between two different groups with false discovery rate (FDR) below 0.05 and absolute fold change ≥ 2 considered differentially expressed genes. Gene Ontology (GO), Gene Set Enrichment Analysis (GSEA), Kyoto Encyclopedia of Genes and Genomes (KEGG), and hallmark gene sets in Molecular Signatures Database (MSigDB) were used to analyze the enriched pathways. Bioinformatic analysis was performed using Omicsmart platform (https://www.omicsmart.com).
Cell Cycle and Apoptosis Assays
For cell cycle analysis, cells were collected and fixed with 70% ethanol overnight at 4°C. Cells were washed with PBS and stained with propidium iodide (PI) using Cell Cycle Analysis Kit (Beyotime, C1052) for 30 min at 37°C. After staining, samples were subjected immediately to flow cytometry analysis. For cell apoptosis analysis, YF®488-Annexin V and PI Apoptosis Kit (US Everbright, Y6002) was used following the manufacturer’s instructions. Cells were collected and stained with YF®488-Annexin V and PI for 20 min at room temperature away from light. To avoid disturbance from GFP fluorescence in shNC and shADAR1 cells, we used Annexin V-APC and 7AAD doubling staining method (Annexin V-APC and 7AAD Apoptosis Kit, US Everbright, A6030) to determine their apoptosis. Samples were subjected immediately to flow cytometry analysis. Flow cytometry was conducted using FACS Celesta (BD) and FACS Calibur (BD) and data was analyzed using FlowJo V10 software.
ADAR1 deaminase activity determination
The ability of compounds to inhibit purified ADAR1 protein deaminase activity was determined by Adenosine Deaminase (ADA) Activity Assay Kit (Fluorometric) (Abcam, ab204695) following the manufacturer’s instructions. Measure output on a fluorescent microplate reader at Ex/Em = 535/587 nm in a kinetic mode, every 2 minutes, for at least 60 minutes at 37°C protected from light. The fluorescent value in two time points in compounds treated group relative to control group was regarded as remaining ADAR1 deaminase activity.
Flow Cytometry
Tumors from xenograft mice were ground mechanically, digested, and filtered with 70-µm screen to provide single cell suspensions. Fresh spleens collected from xenograft mice were subjected to erythrocytes lysis (Biolegend, 420301). Mice cells were blocked with anti-mouse CD16/32 (TruStain FcX™, Biolegend, 101319), followed by cell surface staining with various labeled antibodies for 20 min at 4°C in the dark. MNCs isolated from BM blood of AML patients were blocked with Fc receptor blocking solution (Human TruStain FcX™ Biolegend, 422301). Subsequently, cells were centrifuged and resuspended in Cell Staining Buffer (Biolegend, 420201), and were subjected to flow cytometry directly. Anti-mouse antibodies used were as follows: FITC-CD3, APC-CD8a, FITC-CD11b, APC-Gr-l (ly6C/ly6G). Anti-human antibodies used were as follows: FITC-CD11b, APC-CD15. Flow cytometry was conducted using FACS Celesta (BD) and FACS Calibur (BD) and data was analyzed using FlowJo V10 software.
Immunohistochemistry (IHC)
Tumors and key organs from prostate cancer xenograft mice were fixed in 4% PFA followed by embedded in paraffin wax, sectioned in slides. Sections were incubated with 3% H2O2 for 10 min to quench endogenous peroxidase activity followed by antigen retrieval using unmasking solution. Nonspecific binding was blocked with 2% goat serum for 30 min. These sections were then subjected to ADAR1, Ki-67, Tunel and hematoxylin and eosin (H&E) staining. Organs from mice in acute toxicity study were carried out for H&E staining. A human prostate cancer tissue microarray was purchased from Servicebio technology Co., Ltd. (Wuhan, China). This slide was stained with anti-ADAR1 (1:200 dilution) antibody. Immunohistochemistry staining intensity was measured by Image-Pro Plus 6.0 software. The staining H-Score was calculated by the following formula: H-SCORE = (percentage of cells of weak intensity) × 1 + (percentage of cells of moderate intensity × 2) + percentage of cells of strong intensity × 3). Slide was visualized using CaseViewer2.2 software. Clinicopathologic information of each patient was listed in Supplementary Table S1.
Enzyme-Linked ImmunoSorbent Assay (ELISA)
Tumor was lysed in RIPA lysis buffer. IFN-γ level in lysate or plasma was detected using Mouse IFN-γ ELISA Kit (Beyotime, PI508) or Human IFN-γ ELISA Kit (Beyotime, PI511) according to manufacturer’s instructions. Briefly, samples were added into pre-immobilized IFN-γ antibody 96-well plate and incubated for 2 h in r.t. IFN-γ biotinylated antibodies were added into plate and incubated for 1 h in r.t. Subsequently, horseradish peroxidase labeled streptavidin was added into plate with incubation for 20 min followed by adding stop solution, and then immediately measured in 450 nm. The IFN-γ content in tumor or plasma was calculated based on standard curve.
Determination of editing level in cells
Cells were extracted using RNA-easy Isolation Reagent (Vazyme, R701-01) following the manufacturer’s instructions. RNA was reversely transcribed to cDNA using the HiScript® III 1st Strand cDNA Synthesis Kit (Vazyme, R312-02). To precisely measure the A-to-G levels in RNA target, NestPCR was performed to amplify cDNA product preventing mismatching. cDNA product was subjected to PCR amplification for 25 cycles followed by the second PCR for 35 cycles using Phanta® Max Super-Fidelity DNA Polymerase (Vazyme, P505-d1) (Primers used for NestPCR in Supplementary Table S2). cDNA products were purified by agarose gel electrophoresis, and then subjected to Sanger Sequencing. The editing level was quantified based on peak height ratio (%G/(A + G)) using BioEdit software.
Quantitative Real-Time PCR (qRT-PCR) and RNA editing site-specific qRT-PCR (RESSqPCR)
Total RNA was extracted using RNA-easy Isolation Reagent according to the manufacturer’s instructions. RNA was reversely transcribed to cDNA using HiScript® III RT SuperMix (Vazyme, R323-01) and then subjected to qRT-PCR using ChamQ SYBR qPCR Master Mix (Low ROX premix) (Vazyme, Q331-02). All qPCR assays were run in an ABI 7500 Fast Real-Time PCR system (Applied Biosystem, USA). In this study, each sample was run in triplicates. Relative gene expressions were calculated by 2−ΔΔCt method and GAPDH was used as endogenous control. To determine editing level with qPCR method, RESSqPCR was used to confirm the reduced editing level of known target of ADAR1 as previously reported. Relative RNA editing ratios (Relative edit/WT RNA) were calculated using the following formula: 2−(Ct Edit − Ct WT). The primers used for qPCR and RESSqPCR were listed in Supplementary Table S2.
Animal studies
Four- to five-week-old wild-type female BALB/c mice were purchased from Shanghai SLAC Laboratory Animals Co. Ltd. We subcutaneously implanted 1 × 106 tumor cells (mixed 1:1 v/v with matrigel) into the right flank of BALB/c mice age-matched to 6- to 8-week-old. Drug treatment was started when xenograft tumor size reached to around 70–100 mm3. ZYS-1 was administrated into mice via intraperitoneal injection twice every day before euthanatizing mice. Tumor volumes were measured every two days using the formula: L × W2/2. There were 8 mice in the vehicle group and 12 mice in both low dose group (20 mg/kg/day) and high dose group (40 mg/kg/day). At the end point of experiment, we euthanized the mice, and dissected, photographed, and weighed tumors and key organs. Tumors and organs were either preserved in 4% PFA or carried out directly to further analysis. All animal studies conformed to the guideline of experimental animal of China Pharmaceutic University.
Acute toxicity assay
Six- to eight-week-old wild-type female ICR mice were purchased from Shanghai SLAC Laboratory Animals Co. Ltd. Mice were intraperitoneally or orally administrated with various doses of ZYS-1 for single time, and body weights and conditions were recorded every two days for two consecutive weeks. Each dosing group contained 10 mice. At the end time, all mice were euthanized and key organs were dissected, weighed and kept in 4% PFA for H&E staining. LD50 was calculated using GraphPad Prism 8.0 software.
RNA immunoprecipitation (RIP) assay
Cells were lysed in lysis buffer (100 mM KCl, 5 mM MgCl2, 10 mM Hepes pH 7.0, 1 mM DTT, 50 units/mL RNase out, 1 × protease inhibitor cocktail, 1 × PBS) at 4°C for 2 h. Volume of 10% lysate was subjected to RNA isolation as input. 10 mg Protein A-Agrose beads (Sigma-Aldrich, P1406-250MG) was pre-treated with PBS three times and 2% BSA for 30 min and then incubated with 10 µL lgG control, ADAR1 or RPL22 antibody at 4°C for 2 h. Subsequently, beads-antibody complex was incubated with cell lysate on rotation at 4°C overnight. Beads were washed with PBS and then subjected to total RNA isolation. Purified RNA was reversely transcribed followed by qRT-PCR. For anti-ADAR1 RNA immunoprecipitation, extracted RNA was subjected to library construction and RNA-sequencing (Guangzhou Genedenovo Biotechnology Co., Ltd).
Microscale thermophoresis assay (MST)
Purified ADAR1 was first buffer-exchanged with spin-filter concentrator (amicon 30 kDa cutoff) to the dilution buffer of following composition: 25 mM HEPES, 200 mM NaCl pH7.5. Then the protein was labeled with the Monolith NT Protein Labeling Kit RED (NanoTemper, MO-L001) according to the manufacturer’s labeling protocol. Compounds were sequentially diluted covering the range of appropriate concentrations from 0.5 mM to 1 µM. The labeled protein was mixed with the same volume of diluted compound. Samples were loaded into Monolith standard-treated capillaries and the thermophoresis was measured at 22°C using 100% LED and 20% MST power on a Monolith NT.115 instrument (NanoTemper Technologies, München, Germany). Date analyses were performed with Mo.Affinity Analysis v2.2.4 and GraphPad Prism 8.0 software.
Structure-Based Virtual Screening
The three-dimensional structure of ADAR1 protein referred to the previously reported literature (48), and Discovery Studio 2021 (Accelrys, CA, USA) was employed to screen the combined ligand database, which was filtered by Lipinski’s rule of five, Veber’s rule and the ‘pan-assay interference compounds’. The structure-based virtual screening procedure contains two programs, LibDock and CDOCKER, LibDock is a molecular docking program based on a high-throughput algorithm, while the CDOCKER is a more accurate docking method that employs ligands flexibly to match the binding pocket. Based on the virtual screening procedure, we obtained 20 compounds from the ligand database for experimental study.
Statistical analysis
The unpaired, two-tailed Student’s t test was used to compare difference between two groups carried on SPSS 22.0 software, if not specifically indicated in figure legends. For bioinformatics analysis, Student’s t test and Mann-Whitney Wilcoxon test were used to compare continuous variables between two groups. Analysis of variance (ANOVA) was utilized to test the variance among multiple groups. Paired, two-tailed Student’s t test was used to compare ADAR1, ADAR2, and editing levels between 16 pairs matched tumor and normal tissues in TCGA, carried on GraphPad Prism 8.0 software. Kaplan-Meier plots and log-rank tests were used for relapse-free survival analysis. The values were expressed as mean ± SD with the numbers of replicates listed in figure legends. P < 0.05 was considered statistically significant.