Mice and cell lines
Female WT C57BL/6, BALB/c, and NOD-SCID IL2Rgnull (NSG) mice (6–8 weeks old) were purchased from Shanghai Jie Si Jie Laboratory or Beijing Biocytogen and allowed to acclimatize for 1–2 weeks before experimentation. All animal experimental procedures were approved by the Institutional Animal Care and Use Committee of Shanghai General hospital affiliated with Shanghai Jiao Tong University School of Medicine (2019-A012-01).
The cell lines B16-F0, B16-F10, and 4T1 were obtained from the American Type Culture Collection (ATCC). B16-GMCSF cells were generated by retroviral-mediated gene transfer, following the previously described 44. B16-GMCSF-OVA was ovalbumin (OVA)-transfected clone derived from B16-GMCSF cells which were transfected with the plasmid pCI-neo-mOVA (Cat. 25099, Addgene). Cells were cultured using RPMI-1640 (Corning) with 10% fetal bovine serum (Corning) and 1% Pen/Strep (GIBCO). Cells were incubated in an incubator maintained at 37° C and 5% CO2.
Tumor challenge and treatment
For the B16 tumor challenges, 2 × 105 B16-F0 or B16-GMCSF tumor cells were resuspended in Hanks balanced salt solution (Gibco) and intradermally (i.d.) injected into the right flank of C57BL/6J mice on day 0. For the 4T1 model, 2 × 105 4T1 cells were orthotopically injected into the mammary fat pad of BALB/c mice on day 0. For studies in immune-compromised mice, the Uba6-null or control 4T1 cells were done in the NSG mice. Treatments were given as single agents or in combinations with the indicated regimen for each drug. Inosine (Cat. 4060, Sigma-Aldrich) was administered by oral gavage once a day at 400mg/kg. Control groups received vehicles (sterilized water). Treatment was initiated on day 4 and ended on day 21 post tumor implant. The combination of Rat monoclonal anti-CTLA4 antibody (100µg per mouse, clone 9H10, Bio X Cell) and anti-PD1 antibody (200µg per mouse, clone RPM1-14, Bio X Cell) (ICB) treatment were injected intraperitoneally (i.p.) on days 7, 10, 13 and 16 for the indicated tumor models. Rat IgG2a isotype control was used in control mice corresponding to the ICB treatment group. Each tumor was measured every 3 days with a caliper beginning on day 7 after the challenge until either the survival endpoint was reached, or no palpable tumor remained. Tumor volume was calculated using the formula: (L × W2)/2 and expressed as mm3. Mice that had no palpable tumors that could be measured on consecutive measurement days were considered complete regressions.
T cell-mediated cytotoxicity assays
CD8+ T cells were isolated from the spleen of Balb/c or C57BL/6 mice using a CD8a+ T cell isolation kit (Miltenyi Biotec, Germany) according to the manufacturer’s protocol. and then cultured in complete RPMI 1640 media (10% FBS, 20 mM HEPES, 1 mM sodium pyruvate, 0.05 mM 2-mercaptoethanol, 2 mM l-glutamine, and 50 U /ml of streptomycin and penicillin). Freshly isolated CD8+ T cells were stimulated with anti-CD3/CD28 antibody (BioLegend, USA) to induce differentiation into an effector state. On day 3, recombinant mouse IL-2 (BioLegend, USA) was added to the culture at 20 ng/ml. For the generation of activated OT-1 T cells, splenocytes were harvested from the spleen of OT-1 transgenic mice and stimulated with 100 ng/mL of OVA peptide (SIINFEKL) for 24 hr to expand CD8+ OT-1 T cells. After washing to remove the peptide, cells were cultured in media for an additional 2 days before use in co-culture assays. 4T1 and B16-GMCSF cells were maintained in complete RPMI-1640 media. For the effect of inosine on T cells, isolated CD8+ T cells were pretreated with a serial dilution of inosine for 48 hours during T cell activation. After washed, in vitro-activated CD8+ T cells co-cultured with indicated tumor cells at a different effector to target ratios. For the effect of inosine on tumor cells, 4T1 cells or B16-GMCSF-OVA were seeded and pretreated with a serial dilution of inosine for 48 hours, after washed and then co-cultured with activated CD8+ T cells or OT-1 T cells respectively at a different effector to target ratios. Tumor cells were plated at equal density in all wells and activated CD8+ T cells were added at target-to-effector 1:0, 1:2, 1:5 ratio (Target: Tumor cells; Effector: activated CD8+ T cells). Cell viability is calculated as the quantification of the number of live cells and is also expressed as relative cell viability by calculating the fold change (FC) of remaining alive target tumor cells following the incubation with T cells at the indicated inosine treatment compared to that in the untreated control. After a two or three-day co-culture with T cells, the number of viable tumor cells was counted using the automated cell counting system.
RNA-seq transcriptome analysis of tumor cells
Total RNA of Uba6-null or sgCtrl 4T1 cells was extracted from cell pellets and libraries prepared with the NEB Next Ultra Directional RNA Library Prep Kit for Illumina (New England Biolabs, USA) were sequenced on an Illumina NextSeq 500 instrument. Clean reads obtained by filtering the raw reads with Cutadapt (v 1.9.1) were aligned to the mouse reference genome (assembly GRCm38) using the HISAT2 v2.1.0. and subsequently assembled using Stringtie (v 1.3.3). Cuffdiff (v1.3.0) was applied to calculate Fragments Per Kilobase of exon per Million fragments mapped (FPKMs) for coding genes in each sample, and differentially expressed genes calling was applied using DESeq2 (v 1.30.1), in which significance was assessed by Benjamini-Hochberg False Discovery Rate (FDR) to account for multiple hypothesis testing. ClusterProfiler (v 3.18.1) was used to annotate genes with gene ontology (GO) terms and perform GSEA using the Hallmark gene signature collection from mSigDB 45. Ingenuity Pathway Analysis (QIAGEN) was used for ingenuity upstream regulator analysis 46.
Proteomics analysis of tumor cells
About 107 of Uba6-null and sgCtrl 4T1 cells were suspended in a solution of 9.5 mol/L urea, 1% DTT, 40 ml/ml protease inhibitor cocktail, 0.2 mmol/L Na2VO3, and 1 mmol/L NaF. The mixture was centrifuged at 40000 x g at 15℃ for 1 h and the supernatant was collected. The sequencing-grade trypsin was added to the supernatant containing about 1.5 mg of protein at an enzyme-to-protein ratio of 1:50 and incubated at 37℃ for 14 h. The peptides were desalted using a 1.3 ml C18 solid-phase extraction column (Sep-Pak® Cartridge) (Waters Corporation, Milford, USA) and analyzed by two-dimensional (2D) strong cation-exchange, (SCX)/reversed-phase (RP) nano-scale liquid chromatography/mass spectrometry (2D-nanoLC/MS). Proteins and peptides were identified using a target-decoy approach with a reversed database and queried against the Mouse UniProt FASTA database. The quantification of peptides and proteins with “label-free quantification” (LFQ) was performed by MaxQuant.
Identification of proteins interacting with inosine
Chemical proteomics by LiP-SMap approach was performed as in previous studies 22. At first, 4T1 cells were lysed by bead-beating in PBS at 4℃. After centrifugation at 16,000 g for 10 min at 4℃, the supernatant was collected and aliquoted in equivalent volumes containing 100 µg proteins each. To identify the proteins that interacted with inosine, 0.33 nmol/µg (total protein) of inosine was added to each aliquot and incubated at 25℃ for 10 min. Limited proteolysis was conducted by adding protein kinase K (Sangon Biotech, China) at a 1:100 enzyme/substrate ratio. The generated protein fragments were digested by trypsin with a 1:50 trypsin/substrate ratio to generate peptides for mass spectrometry analysis. Peptide fragments were analyzed by Nano Acuity Ultra High-Pressure liquid chromatography coupled with Thermo Q Exactive mass spectrometer (Thermo Fisher, USA). Proteins and peptides were identified using a target-decoy approach with a reversed database and queried against the Mouse UniProt FASTA database. The quantification of peptides and proteins with “label-free quantification” (LFQ) was performed by MaxQuant.
Thioester Assay
Recombinant human ubiquitin and FAT10 were purchased from Boston Biochem. Plasmids pEnter-UBA6 and pEnter-USE1 were used for the expression of Flag-UBA6 and Flag-USE1. The two plasmids were transfected into HEK293, respectively, using Lipofectamine 2000 reagent. Purification of Flag-UBA6 and Flag-USE1 were carried out using Anti-FLAGM2 Affinity Gel (Sigma) as described by the manufacturer’s instructions. Flag-UBA6 (0.5µM) and Flag-USE1 (0.5µM) were incubated with vehicle, inosine, or TAK243 at room temperature for 15min. Then, ubiquitin (5µM) or FAT10 (2µM) with ATP (250µM) were added. The reaction mixtures were incubated at 37℃ for 30 min before 2×Lammli sample loading buffer was added to quench the reaction. The thioester detection was fractionated by SDS-PAGE under nonreducing conditions and immunoblotted with anti-UBA6 antibody (Proteintech, 1:1000) and anti-USE1 antibody (ABclonal, 1:1000).
Generation of CRISPR-edited tumor cell lines
Uba6 was deleted in Cas9-expressing 4T1 and B16-GMCSF mouse tumor cell line for validation experiments using a lentiviral delivery system (lentiCRISPR v2, Addgene) to express sgRNAs, and puromycin selection. For determining the knockout efficiency of the Uba6 gene, Western Blotting was used to measure the protein expression of UBA6 in sgCtrl control and sgUba6 4T1 or B16-GMCSF cells. The Uba6-null 4T1 or B16-GMCSF cells were selected for experiments.
CRISPR sgRNA sequences:
Control sgRNA 1: GCGAGGTATTCGGCTCCGCG
Control sgRNA 2: ATGTTGCAGTTCGGCTCGAT
Uba6 sgRNA 1: AAGTCCTGTGTCTTCTTAAG
Uba6 sgRNA 2: AAATCGATGATGGATTATAC
Metabolomic analysis in tumor-bearing mice
Plasma metabolites in B16-F0 tumor-bearing mice or Abx-treated mice were measured. A total of 244 metabolites in plasma were detected by ultra-high-performance liquid chromatography (HPLC) coupled with a tripleTOF 5600 plus mass spectrometer (Applied Biosystems, USA). The metabolomic data were analyzed by pattern recognition analyses (principal component analysis and Heat-map).
Antibiotic treatments
Six-week-old C57BL/6J mice were treated with a cocktail of broad-spectrum antibiotics (1 g/L ampicillin, 1 g/L neomycin, 1 g/L metronidazole, and 0.5 g/L vancomycin) in drinking water for 3 weeks. The mice were allowed 3–4 days to recover before tumor implants. For measuring the levels of purine metabolites, the fresh fecal pellets, and plasma were collected at day 0 after two hours in collection cages with a paper liner. For evaluating the effect of ICB on tumor growth, IgG2a or a combination of anti-CTLA4 and anti-PD1 Abs (ICB) treated B16-F0tumor-bearing mice as the indicated time points.
The correlation analysis between survival and metabolites in human cancer patients
We reanalyzed the public dataset 17 regarding the metabolic profiles of 743 renal cell carcinoma (RCC) patients (Phase III trial: CheckMate 025, NCT01668784), among which 394 patients received nivolumab and 349 patients received everolimus. The overall survival in nivolumab or everolimus-treated RCC patients grouped by metabolite level (the upper half was as a high-level group; the lower half was as a low-level group defined by the median value of individual metabolites) were measured using Kaplan–Meier plot.
Quantitative real-time PCR (qPCR)
Total RNA of indicated tumor cells was extracted by using RNAprep pure Tissue Kit (Tiangen Biotech, China), according to the manufacturer’s protocol. RNA (2µg) was reverse transcribed using the PrimeScript™ RT reagent Kit with gDNA Eraser (Takara, China). Quantitative RT-PCR was performed using PowerUp™ SYBR™ Green Master Mix with QuantStudio 6 Flex System (Thermo Fisher, USA). Relative mRNA expression was determined by the ΔΔCt method and normalized to Gapdh. All qPCR primers used are listed in Supplementary Table 6.
Western blot
ORF of human UBA6 and USE1 in pEnter, with C terminal Flag, were purchased from Vigene Biosciences (JiNan, China). Moreover, UBA6 with the UFD domain (residues 949–1052) deletion (UBA6ΔUFD) was generated by PCR. The amplified DNA fragment was cloned into pEnter. The human embryonic kidney cell line HEK293 was purchased from ATCC and was cultured in DMEM supplemented with 10% of FBS and 50 U/ml of penicillin/streptomycin. Cells were transfected using Lipofectamine 2000 reagent as described by the manufacturer’s instructions. 500 µM of inosine or vehicle was added 24 h after transfection. At 48h, cells were harvested and lysed in lysis buffer (50 mM Tris HCl, pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% TRITON X-100). Cleared lysates were subjected to anti-FLAG immunoprecipitation using Anti-FLAGM2 Affinity Gel (Sigma, USA) overnight at 4°C. Samples were washed three times with TBS. Proteins were separated on 8% or 12% Laemmli SDS gels and subjected to western blot analysis using an anti-UBA6 antibody (Proteintech, 1:1000), anti-USE1 antibody (ABclonal, 1:1000), and anti-β-Actin antibody (Sangon Biotech, 1:1000).
Prepare the single-cell suspension from 4T1 tumor tissues
4T1 cells (2×105) were implanted in BALB/c female mice. Mice were treated with IgG2a (Ctrl) or a combination of inosine and ICB (anti-CTLA4 and anti-PD1 Abs) (Combo) as described above. On day 13, tumors were harvested and minced with scissors before incubation with collagenase A (2 mg/ml, Roche) and DNase I (50 µg/ml, Roche) in RPMI-1640 completed medium (10%FBS, 1% P/S) for 30 min at 37°C. Tumor samples were homogenized by repeated pipetting and filtered through a 70µm nylon filter (BD Biosciences) in FACS staining buffer (PBS/0.5% albumin) to generate single-cell suspensions. After red blood cell (RBC) lysis (RBC Lysing Buffer, Biolegend), all samples were washed and re-suspended in FACS staining buffer for further single-cell RNA sequencing (scRNA-seq) or flow cytometry.
Analysis of tumor-infiltrating immune cells by scRNA-seq
Tumor-infiltrating immune cells from 4T1 tumor-bearing mice with IgG2a (Ctrl, n = 2) or a combination of inosine and ICB (anti-CTLA4 and anti-PD1 Abs, n = 2) (Combo) treatment were enriched using CD45+ MicroBeads kit (Miltenyi Biotec, Germany). 2 biological replicates in the vehicle and inosine + ICB groups were performed. The single-cell RNA-seq was performed as described 47. Briefly, cells were counted and loaded into the 10x Genomics device. After reverse transcription, barcoded cDNAs were purified, amplified, end-repaired, and ligated with Illumina adapters to generate a single multiplexed library according to the manufacturer’s protocol. All resulting libraries were sequenced on the Illumina Novaseq 6000 platform (Illumina, USA).
Preliminary sequencing results were de-multiplexed the cellular barcodes and aligned reads to the transcriptome GRCm38 (mouse) using the Cell Ranger v2.1.1 pipeline. Mean and dispersion values were calculated for each gene across the remaining 16, 199 cells (Ctrl group) and 9, 842 cells (Combo group), and variably expressed genes were selected for principal component analysis (PCA). Then, t-SNE was performed using default parameters for visualization in two dimensions. All CD45+ immune cells were clustered as described 48. Unsupervised clustering using a shared nearest neighbor modularity optimization-based algorithm identified 32 distinct clusters. 14 major clusters were identified by mapping canonical marker genes in the two-dimensional tSNE map. Detailed descriptions of the immune cell subsets and their marker genes are included in the figures and main text of the relevant sections.
Flow cytometry assay
For flow cytometry analysis of in vivo experiments, tumor single cells were isolated from mouse 4T1 tumors as described above and pre-incubated (15 min, 4°C) with an anti-CD16/32 monoclonal antibody (clone 93, Biolegend) to block nonspecific binding and then stained (30 min, 4°C) with appropriate dilutions of various combinations of the following fluorochrome-conjugated antibodies: anti-CD45-AF 700 (clone 30-F11), anti-CD11b-PE (M1/70.15), anti-F4/80-APC (clone BM8), anti-MHC Class II-FITC (clone M5/114.15.2), anti-CD206-PE (clone 19.2), anti-CD8-Percp-Cy5.5 (clone 53 − 6.7), anti-CD4-PE (clone RM4-5) antibodies, all purchased from Biolegend or ThermoFisher. For tetramer staining in the 4T1 model, tumor antigen-specific CD8+ T cells were detected with PE-conjugated H-2Ld tetramer to peptide SPSYVYHQF (MuLV env gp70, 423 to 431) was purchased from MBL International. Antibodies were used at 5 ug/ml, and tetramer staining was performed in FACS buffer for 20 min at room temperature and followed by surface staining on ice for 20 min. Dead cells and doublets were excluded based on forward and side scatters and Fixable Viability Dye eFluor 506 (Thermo Fisher, USA).
For in vitro analysis of the effect of inosine on MHC Class I antigen expression, B16-GMCSF or 4T1 cells were seeded and treated with a serial dilution of inosine for 48 hours. Cells were non-enzymatically detached from the wells, washed with FACS staining buffer, and then incubated with FITC-conjugated anti-mouse H-2Kd antibody (clone SF1-1.1, BioLegend) for 30 min on ice. After washing, cells were resuspended in FACS staining buffer, and then > 2000 cells were analyzed by flow cytometry. Acquisition and analysis were performed on Canto II Flow Cytometer using BD FACSDiva software (BD Biosciences, USA) and all analyses were performed with FlowJo software v10 (BD).
Tumor cells viability and apoptosis assays
For the effect of inosine on tumor cell growth in vitro assay, 4T1 or B16-GMCSF cells were seeded in 96-well plates (1,000 cells per well) and allowed to seed for 24 h, after which they were treated with inosine. For in vitro cytokine stimulations and growth inhibition assays, sgCtrl or UBA6-null 4T1 or B16-GMCSF tumor cells were plated in media containing the indicated combinations of cytokines: 10 ng/ml IFNγ (PeproTech, USA), 10 ng/ml TNFα (PreproTech, USA), or 10 ng/ml IFNγ + 10 ng/ml TNFα. Treatment was given only once at the beginning, after the seeding of cells. Subsequently, every 24 h, MTT reagent (Sigma, USA) was added to the cell culture media for 3 h at 37°C. The supernatant was then discarded and lysed with DMSO to dissolve the formazan product. Absorbance was measured by a spectrophotometric plate reader.
For flow cytometry analysis of apoptosis, 4T1 or B16-GMCSF cells were treated inosine for 48h, and following trypsinization and washes in FACS staining buffer, tumor cells were stained for 20 min on ice using the manufacturer’s recommended concentrations of Annexin-V PE and 7-AAD from the PE Annexin V Apoptosis Detection Kit 1(BD Pharmingen, USA) according to the manufacturer’s instructions. The staining of cell surface markers was then analyzed using the Canto II flow cytometry system (BD Biosciences, USA). The analysis was carried out using FlowJo software.
Integrative gene knockout screening platform and survival analysis based on TIDE
We collected cancer data sets with both patient survival durations and tumor gene expression profiles from The Tumor Immune Dysfunction and Exclusion (TIDE) website and tools 23,31. Candidate genes were plotted based on mean log2 fold change (logFC) of gRNA counts compared to control selection and normalized z-score generated using the pheatmap R package and presented as the expression level of the individual gene was standardized to zero mean and one standard deviation. The normalized logFC and Z-score in CRISPR screens help identify regulators/genes whose knockout can mediate the efficacy of lymphocyte-mediated tumor killing in cancer models. Higher logFC and Z-score mean that knockout of gene resistant to lymphocyte-mediated tumor cell killing, contrast, lower logFC, and Z-score mean that knockout of gene mediates the enhancement to lymphocyte-mediated tumor cell killing. Kaplan–Meier plots of overall survival (OS) or disease-free survival (DFS) of cancer patients treated with ICT who had high UBA6 vs. low UBA6 in the tumors as respectively defined by the median expression levels were accessed by TIDE program. To test the association between UBA6 gene expression level and patient survival, Kaplan-Meier survival analysis was performed using the program described in the Gene Expression Profiling Interactive Analysis (GEPIA2) 49.
Human Tissue Samples collection and histological evaluation of UBA6 expression
Human tumor tissues were collected from individual participants following the guidelines approved by the Institutional Review Board at Beijing Friendship Hospital affiliated to Capital Medical University (2017-P2-141-02). A total of 22 human tissue samples were obtained from patients with cancers of esophageal (n = 7), stomach (n = 4), lung (n = 3), colon (n = 3), and others (n = 5) (gender: 12 males and 10 females; mean ± SD age, 62.4 ± 8.6 years; median age, 63 years; range, 39–77 years). Pearson correlation test was utilized to confirm an insignificant association of patient age and sex with UBA6 expression (p = 0.62). The disease control rate and the objective response rate (n = 22 of this cohort with immunotherapy response rate available) were comparable to previously reported in unselected patients 50. Standard immunohistochemical (IHC) assays were performed for UBA6 evaluation as described previously 51. In brief, tumors were harvested before immunotherapy and fixed in 10% neutral-buffered formalin. After deparaffinization and rehydration, 4µm tissue sections were subjected to heat-induced epitope retrieval. Slides were processed with the VECTASTAIN Elite ABC HRP Kit and DAB Substrate Kit (Vector Laboratories). Slides were then incubated with anti-UBA6 antibody (Proteintech, 1:1500). Five visual fields from different areas of each slide were independently evaluated by 2 pathologists who were blinded to the group allocation during the staining and when assessing the outcomes. Necrotic areas in the tumors were excluded from the evaluation. IHC intensity scores of UBA6 were ranked into 4 groups: negative (−), positive-low (+), positive-medium (++), and positive-high (+++). In the IHC scoring of patient samples, the score “low” corresponded to negative (-) to positive-low (+), while the score “high” corresponded to the range from + + to +++.
ICB treatment and assessment of ICB response in cancer patients
This clinical study was approved by the institutional ethics committee of Beijing Friendship Hospital affiliated with Capital Medical University and was conducted following clinical practice guidelines. The study was designed by the authors in collaboration with the sponsors, and all 22 cancer patients are recruited for evaluation of Anti-PD1 Ab combined with paclitaxel treatment. Anti-PD1 Ab (Sintilimab, Innovent Biologics) was provided by the sponsor or procured as commercial products, and paclitaxel was procured as commercial products. Anti-PD1 Ab was administered at a dose of 200mg/per time as a 60-minute intravenous infusion every 3 weeks. Paclitaxel was administered at 175 mg/m2 intravenously daily for 3 weeks. Disease assessments were performed with the use of computed tomography (CT) or magnetic resonance imaging at baseline, every 8 weeks until disease progression or discontinuation of treatment. Imaging data were evaluated by the investigators to assess tumor response. The clinical objective response was determined as the investigator-assessed best response based on immune-related response evaluation criteria in solid tumors (irRECIST) 52 using unidimensional measurements (CR: complete response, PR: partial response, SD: stable disease, PD: progressive disease). The assessment of responses for patients was conducted independently in a double-blind fashion from the time of randomization to objectively documented disease progression or subsequent therapy.
Bioinformatics and statistical analysis
Statistical tests employed with the number of replicates and independent experiments are provided in the figure legends or text. Unless mentioned otherwise, all graphs with error bars are presented as mean ± s.e.m. GraphPad Prism (v.8) is used for basic statistical analysis and plotting. Statistical significance is determined by one-way ANOVA with Tukey and Dunnett’s posttests and two-way ANOVA with a Bonferroni test for multiple comparisons, or an unpaired Student’s t-test for pair-wised comparison. Multiple hypothesis testing corrections were applied where multiple hypotheses were tested and are indicated using FDR. Kaplan-Meier survival curves are graphed and analyzed using the log-rank test for multiple comparisons. P-value < 0.05 was indicated as statistically significant.
Material availability
Materials that are not available commercially can be requested from the corresponding author.
Data availability
Data are available within the Article, Supplementary Information, or available from the authors upon request.