Sample collection
This study was approved by the Ethics Committee of Nanfang Hospital (NFEC-2019-057) and the First Affiliated Hospital (2021-Ani-58), Zhejiang University School of Medicine. All participants provided written informed consent to take part in the study. The detailed patient information is available in Table S1. Sample tissues were obtained from the biopsies of 110 patients diagnosed with ICC. All samples were assessed by pathological examinations.
Tandem mass tag (TMT) -based proteomic analysis
A total of Formalin-fixed paraffin-embedded (FFPE) samples (0.5-1 mg) were dewaxed and rehydrated and then subjected to acidic hydrolysis with formic acid (FA). Proteins were denatured with 6 M urea (Sigma-Aldrich, Germany) and 2 M thiourea (Sigma-Aldrich, Germany), and then digested into peptides with trypsin (1:20, Hualishi, Beijing, China) and Lys-C (1:80, Hualishi, Beijing, China) with the assistance of pressure-cycling technology (PCT) 23, 24. Peptides were labelled with TMTproTM 16 plex (Thermo Fisher Scientific™, San Jose, USA) 25. Each batch contained 15 experimental samples and one pooled sample in the TMT126 channel for normalization. The fractions (60 per batch) were separated using offline high-pH reversed-phase with a Thermo Dionex Ultimate 3000 RSLCnano System and then combined to a total of 30 fractions per batch. Subsequently, the fractionated samples were separated with a Thermo Dionex Ultimate 3000 RSLCnano System and then analysed with a Q Exactive HF mass spectrometer using the data-dependent acquisition (DDA) mode (Thermo Fisher Scientific™, San Jose, USA). The database searching included all reviewed human entries from UniProt (downloaded on 14 April 2020, containing 20,365 proteins) using Proteome Discoverer (version 2.4, Thermo Fisher Scientific™, Waltham, MA). The detailed parameters were described previously 26, 27.
Molecular subtype identification
The “ConsensusClusterPlus” package was applied to the proteomic data to identify potential molecular subtypes of ICC 28. Differentially expressed protein (DEP) analysis was performed with the “Limma” package to assess the proteomic data 29. An empirical Bayesian method was applied to estimate the fold change between every two molecular subtypes using moderated t-tests. The adjusted p-values for multiple tests were calculated using the Benjamin-Hochberg correction.
Single-sample GSEA (ssGSEA) and gene ontology (GO) analysis
ssGSEA was applied to evaluate the enrichment scores of each sample. The “GSVA” package was used for this analysis 30. The hallmark gene sets were downloaded from The Broad Institute. Gene sets of immune cells populations were came from Bindea G et al. 31. The correlation between the risk and the enrichment scores was performed using Spearman’s coefficient. GO analysis was performed with the “clusterProfiler” package 32.
DNA extraction, library preparation, and targeted enrichment
Next-generation sequencing (NGS) analyses were performed according to protocols reviewed and approved by the Ethics Committee of the First Affiliated Hospital of Zhejiang University and Nanfang Hospital. DNA extraction, library preparation, and target capture enrichment were performed using previously described protocols with minor modifications 33. Briefly, genomic DNA from white blood cells was extracted using the DNeasy Blood & Tissue Kit (Qiagen) and was applied as the normal control to remove germline variations. After deparaffinizing the FFPE samples with xylene, genomic DNA was extracted using the QIAamp DNA FFPE Tissue Kit (Qiagen). After quantification of DNA with Qubit 3.0 using the dsDNA HS Assay Kit (Life Technologies), a Nanodrop 2000 (Thermo Fisher) was used to evaluate the DNA quality.
The KAPA Hyper Prep kit (KAPA Biosystems) was used to prepare libraries according to previously described protocols 34. Briefly, 1-2 μg of genomic DNA was sheared by a Covaris M220 instrument into ~350-bp fragments. End repair, A-tailing, and adaptor ligation of fragmented DNA were performed with the KAPA Hyper DNA Library Prep kit (Roche Diagnostics). Agencourt AMPure XP beads (Beckman Coulter) were used for size selection. DNA libraries were generated by polymerase chain reaction (PCR) followed by purification using Agencourt AMPure XP beads.
WES was performed with a customized xGen lockdown probe panel (Integrated DNA Technologies). The blocking reagents were human cot-1 DNA (Life Technologies) and xGen Universal Blocking Oligos (Integrated DNA Technologies). Dynabeads M-270 (Life Technologies) and the xGen Lockdown Hybridization and Wash kit (Integrated DNA Technologies) were used to perform the capture reaction. PCR amplification was performed on captured libraries with KAPA HiFi HotStart ReadyMix (KAPA Biosystems). The KAPA Library Quantification kit (KAPA Biosystems) was used to quantify the purified library. A Bioanalyzer 2100 was used to calculate the fragment size distribution.
Sequencing and bioinformatics analysis
The HiSeq4000 platform (Illumina) was used to sequence the target enriched libraries with 2×150-bp paired-end reads. bcl2fastq (v2.19) was employed to demultiplex the sequencing data. Trimmomatic 35 was used to remove low-quality (quality<15) or N bases. The alignment of the data to the hg19 reference human genome was then performed by the Burrows-Wheeler Aligner (bwa-mem) 36, followed by further processing using the Picard suite (available at: https://broadinstitute.github.io/picard/) and the Genome Analysis Toolkit (GATK) 37. Single nucleotide polymorphism (SNP) and insertion/deletion (indel) calling for GATK was performed with VarScan2 38 and HaplotypeCaller/UnifiedGenotyper, and the mutant allele frequency (MAF) cutoff was set as 0.5%. Common variants were removed according to dbSNP and the 1000 Genome project. The patients’ whole blood controls were used to filter out the germline mutations.
Gene fusions and copy number variations (CNVs) were identified by FACTERA 39 and ADTEx 40, respectively. For the tissue samples, the log2 ratio cutoffs for the copy number gain and the copy number loss were defined as 2.0 and 0.6, respectively. TMB was defined as the number of somatic, coding, base substitution, and indel mutations per megabase of the examined genome, and was calculated as previously described 41. Briefly, all base substitutions were considered, including nonsynonymous and synonymous alterations, and indels in the coding region of targeted genes, except for known hotspot mutations in oncogenic driver genes and truncations in tumour suppressors.
Random forest algorithm for mutation importance ranking
A random forest algorithm was applied to the WES data to identify the most important mutations associated with the inflammatory response in ICC. Briefly, the gene mutation dataset and inflammatory response were applied as input. The “ranger” package was used to find the best hyperparameter in the regression process for the random forest model 42. The “e1071” package was then used to generate the random forest model 43.
Tissue dissociation
RPMI 1640 (Gibco, Cat. no. 11875–093, US), with 1 mM protease inhibitor (Solarbio, Cat. no. P6730, China), was used to transport ICC tissues. Tissues were digested with a dissociation enzyme cocktail prepared by dissolving 2 mg/mL Dispase II (Sigma-Aldrich, Cat.42613-33-2 US), 1 mg/mL Type VIII Collagenase (Sigma-Aldrich, Cat. no. C2139, US), and 1 unit/mL DNase I (NEB, Cat. no. M0303S, US) in PBS with 5% foetal bovine serum (FBS; Gibco, Cat. no. 16000–044, US) for 40 min at 37°C. The cells were dissociated and collected every 20 min, and then filtered using a 40 μm nylon cell strainer (Falcon, Cat. no. 352340, US). Red blood cell lysis buffer (Invitrogen, Cat. US) with 1 unit/mL DNase I was used for removing red blood cells. Finally, cells were washed in PBS with 0.04% Bovine Serum Albumin (BSA; Sigma-Aldrich, Cat. no. B2064, US).
Library preparation and sequencing
The concentration of the single-cell suspension was computed with countess (Thermo) and adjusted to 1000 cells/μL. Cells were loaded according to the Chromium single-cell 3′ kit standard protocol to capture 5,000-10,000 cells/chip position (V2 chemistry). The library construction and all the other processes were performed according to the standard manufacturer’s protocol.
scRNA-seq data processing
Illumina HiSeq X Ten was used to obtain single-cell libraries using 150 nt paired-end sequencing. Reads were processed with the Cell Ranger 2.1.0 pipeline using default parameters. The STAR software was used to align FASTQs to the human reference genome (hg19). For each sample, gene-barcode matrices were generated by counting the unique molecular identifiers (UMIs) and filtering non-cell-associated barcodes. Finally, a gene-barcode matrix containing the barcoded cells and the gene expression counts was generated.
scRNA-seq data analysis
Single-cell transcriptome analysis was performed with “Seurat” with the default parameters 44. A nonlinear dimensional reduction was performed with the t-distributed stochastic neighbour embedding (t-SNE) method. The gene features of each cluster were found by “Seurat”. The “GSVA” package was used to compute the score of each gene set as previously described. The “nichenetr” package was adopted to analyze the cell-cell interaction (CCI) by exploring the ligand and target gene pairs 10. The regulatory potential score was calculated using network propagation methods.
Cell lines and culture
The following cell lines were purchased from American Type Culture Collection (ATCC) and maintained in a humidified atmosphere in the CO2 incubator (Thermo, 3111) at 37 °C: ICC HCCC9810, CCLP-1, HuCCT1, RBE, KMCH1, LICCF, intrahepatic bile duct epithelial HIBEC, promonocytic THP1, and Jurkat. HCCC9810, THP1, HuCCT1, and Jurkat were maintained with RPMI-1640 media, while HIBEC, CCLP-1, LICCF, and KMCH1 were cultured with DMEM (Dulbecco’s Modified Eagle’s Medium) media, supplemented with both fetal bovine serum (10%, Gibco, USA) and penicillin/streptomycin (1%, Gibco, USA).
IHC staining for tissue sections
CCA tumor tissues were fixed with 4% paraformaldehyde before embedding them in paraffin. Tissue sections (4 μm) were later deparaffinized with xylene and rehydrated with graded alcohol. Antigen epitope retrieval was induced by microwave heating. Tissue sections were then blocked for 1h, at room temperature, in goat serum blocking solution (Proteintech, B900780, China) and phosphate buffered saline (PBS). Before primary antibody overnight incubation at 4°C, the next day ICCA tissue samples were stained using an anti-mouse/rabbit universal immunohistochemical detection kit (pk10006, proteintech) following the manufacturer’s instructions. Mounted sections were examined by light microscopy (Leica), and images were analyzed with Image-Pro Plus (version 6.0).
Immunofluorescence (IF) staining for tissue sections
Tissue samples were deparaffinized with xylene and rehydrated with graded alcohol. Antigen epitope retrieval was induced by microwave heating. Tissue sections were permeabilized with 0.2% Triton X-100 and then blocked with 5% BSA, for 1h, at room temperature, and then incubated with the primary antibody, overnight, at 4°C. The following day, tissue sections were washed with PBS three times. Samples were then stained with fluorescent secondary antibodies (Invitrogen, USA) in the dark, for 1h, at room temperature, and then washed with PBS three times before mounting them onto slides using Mowoil supplemented with DPAI to stain the nucleus. Finally, a confocal microscope (Nikon, Japan) was used to take images of ICCA tissue samples. Acquired images were analyzed using ImageJ (version 4.0).
IF staining for cell culture
To induce THP1 cells to differentiate into M0 macrophages,1 X 105 THP-1 cells were treated with 80 nM Phorbol 12-myristate 13-acetate (PMA) once plated into 12 well plates, after 48h, 2 x 105 ICCA tumor cells were plated for co-cultured with M0 macrophages on glass coverslips in 12 well plates. Immunofluorescence assays were performed as follows. First, cells were washed with PBS and fixed with 4% paraformaldehyde, before permeabilizing them with 0.2% Triton X-100 at room temperature. Samples were next blocked with 2% BSA for 1h, at room temperature, and then incubated with the primary antibody for 1h at room temperature. To examine the co-localization of proteins of interest (CD68, APOE, and C1QB), cells were stained with Alexa Fluor ® 488 and 594-labled fluorescent secondary antibody (Invitrogen, USA) for 1h in the dark at room temperature. Coverslips were washed three times with PBS before mounting them onto slides using Mowoil supplemented with DPAI to stain the nucleus. Finally, cells were examined with a confocal microscope (Nikon, Japan). The acquired images were analyzed using ImageJ (version 4.0).
In vivo tumour model
C57BL/6J mice were purchased from the Model Animal Research Center of Nanjing University (China). All mice were housed in the specific pathogen free (SPF) facility of the First Affiliated Hospital, Zhejiang University School of Medicine, with approval from the Institutional Animal Care & Use Committee (IACUC). To establish a spontaneous ICC mouse model, a plasmid mixture carrying T3-EF1a-NICD, PT3-myr-AKT-HA, and SB100 was injected at high pressure into the tail vein of the mice. One week later, the mice were treated with 10 μg/g CSF1R antibody via intraperitoneal injection, and the same injections were performed every 3 days thereafter. On day 28, tumors were collected and further analyzed.
Hematoxylin and eosin (HE) staining
Mouse tumor tissue sections (4μm) were deparaffinized with xylene, and then rehydrated with graded alcohol. Tissue sections were washed for three times using PBS before stained nucleus with hematoxylin for 30 min at room temperature before washed with PBS for three times. Then the sections were exposed to the ammonia water to change the hematoxylin stained nuclei from a reddish to blue-purple appearance. Subsequently, 75% alcohol was used to rinse the tissue sections for 2 min at room temperature, then stained cytoplasm with eosin for 1h at room temperature. And then used graded alcohol directly rinsed after the eosin staining. Finally, the xylene was used to displace the anhydrous alcohol before mounted with slides. sections were examined by light microscopy (Leica), images were analyzed with Image-Pro Plus software (version 6.0).
Ki67 IHC staining
Prepared tumor tissue sections(4μm) deparaffinized with xylene and rehydrated with graded alcohol. Antigen epitope retrieval were induced by microwave heating before blocked in blocking solution (Proteintech, B900780, China) in phosphate buffered saline (PBS) for 1h at room temperature. Then Ki67 primary antibody (1:200, 550609, BD Biosciences) were incubated for overnight at 4°C, the following day, tissue sections were washed three times with PBS before stained using anti-mouse/rabbit universal immunohistochemical detection kit (pk10006, proteintech) according to the manufacturer’s instructions. Mounted sections were examined by light microscopy (Leica), images were analyzed with Image-Pro Plus software (version 6.0).
Fluorescence-activated cell scanning (FACS) analysis
THP1 cells were treated with 80 nM PMA for 48 h to induce THP1 to M0 macrophages and then co-cultured with ICC tumour cells. Before the cells were incubated with fluorochrome-labelled antibody, they were stimulated with Cell Stimulation Cocktail (plus protein transport inhibitors) (eBioscience, 00-4975-03, USA) at 37 °C for 6 h. For surface marker analysis, live cells were re-suspended in 1×PBS supplemented with 2% FBS and stained with anti-human CD3 (UCHT1, BioLegend, 300406) at 4 °C for 30 min. Subsequently, the cells were fixed and permeabilized with Fixation and Permeabilization Solution (BD Bioscience, America) for 30 min and then incubated with anti-human TNF-α (MAb11, Biolegend, 502909) at 4 °C for 30 min. Data were acquired with an ACEA NovoCyte flow cytometer and analysed with FlowJo software (version 10).
Mouse ICCA tissue dissociation assays were performed as previously described. Cells were resuspended with DMEM (Dulbecco’s Modified Eagle’s Medium) media supplemented with 2% FBS, stimulated with Cell Stimulation Cocktail (plus protein transport inhibitors) (eBioscience, 00-4975-03, USA) for 6 h at 37 °C, centrifugated at 500 xg for 5 min, and then incubated with 2.5 μg/ml Fc blocker (Biolegend, 15660) on ice for 15 min. Next, suspensions were incubated with fluorochrome-labeled antibodies (CD45, CD3, CD4, and CD8) at 4 °C for 30 min, then washed with 1×PBS supplemented with 2% FBS and centrifuged at 500 xg for 5 min. Cells were fixed and permeabilized with Fixation and Permeabilization Solution (BD Bioscience, America) at room temperature for 20 min in the dark, before incubating them with anti-mouse TNFα (MP6-XT22, Biolegend, 506305). For analysis assay, the samples were washed and resuspended in 1×PBS supplemented with 2% FBS. The data were acquired with a five-laser flow cytometer (BD Bioscience, Fortessa) and analysed with FlowJo software (version 10).
Statistical analysis
Survival analysis was performed with the “survival” package 45. The hazard ratio (HR) was determined via univariate Cox regression analysis. Statistical tests were selected based on the specific assumptions relative to the data distribution and its variability. Sample data were analyzed by a two-tailed Student's t-test to identify statistically significant differences between two groups; the Kruskal-Walis test was used to identify differences between three groups. The data were represented as means ± standard error of mean (SEM). A p-value < 0.05 indicated statistical significance.
Data availability
The data that support the findings of this study are available from the corresponding author upon reasonable request.