Cell lines and cell culture
The human colorectal cancer cell line HCT116 was purchased from the China Center for Type Culture Collection (Wuhan, China); human colorectal cancer cell lines HT29 and SW480 were purchased from the American Type Culture Collection (Rockville, MD, USA); and the HEK293T cell line, human colorectal cancer cell line SW620 and human hepatic stellate cell line LX-2 were purchased from Cell Bank of Type Culture Collection of the Chinese Academy of Sciences (Shanghai Institute of Cell Biology). All cell lines were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Gibco) supplemented with 10% fetal bovine serum (FBS) (Gibco) and 1% penicillin and streptomycin (Beyotime Biotech Co. Ltd., China). The cells were cultured at 37 ℃ in a humidified incubator containing 5% CO2.
Exosome isolation and characterization
Exosomes in medium were isolated by using ExoQuick-TC Exosome Isolation kit (System Biosciences, Palo Alto, CA, USA) according to the manufacturer’s instructions. Briefly, HCT116, HT29, SW620 and SW480 cells (3 × 106 cells) were seeded in 100-mm dishes in culture medium with 10% FBS. After 24 h, the culture medium was changed to a culture medium without FBS. The culture supernatant was collected after 48h incubation and centrifuged at 4000 g for 30 minutes to remove cell debris. The supernatant was transferred to a sterile vessel, and one-fifth of ExoQuick-TC was added to supernatant. The samples were mixed before incubation overnight at 4 ℃. Exosomes were isolated by centrifugation at 1500 g for 30 minutes. Each pellet was resuspended with PBS and the protein concentration of exosome was determined by BCA assay (TAKARA, Japan) before further experiments.
For exosome transmission electron microscopy (TEM) observation, exosome samples were fixed with 2.5% glutaraldehyde at 4℃ overnight. Then, exosomes were added onto formvar/carbon-coated grids, stained with 2% uranyl acetate for 1 h. Exosome samples were analyzed by TEM (FEI, Tecnai F20) in 120kV.
The density and size of exosomes were tracked by using Nanosight NS300 system (NanoSight technology, Malver, UK). Exosomes were resuspended and diluted in PBS to achieve between 20 and 100 objects per frame. Exosome samples were injected into the sample chamber and measured in triplicate. Data was analyzed using the nanoparticle tracking analysis analytical software (version 2.3).
Exosome labeling and tracing
To assess transfer of exosomes into LX-2, CRCs-derived exosomes were labeled with DiO dye (Rengen Biosciences, China) according to the manufacturer’s instructions. Briefly, exosomes were incubated with DiO dye for 30 minutes at 37 ℃. Excessive DiO was removed with Spin Column. The LX-2 cells were seeded in 35 mm glass-bottom dishes (Cellvis, D35-14-1N) (1 × 104 cells/dishes) and cultured for 24 h. Dio-labeled exosomes, after filtering by 0.22 μm filters, were added to the culture media of LX-2 cells and incubated for 8 h at 37 ℃. The LX-2 cells were fixed with 4% paraformaldehyde for 15 minutes at room temperature and permeabilized with 0.1% Triton X-100 for 20 minutes at room temperature. The LX-2 cells were stained with 4',6-diamidino-2-phenylindole (DAPI) (Boster Biological Technology co., Ltd, China). The LX-2 cells were observed with confocal microscopy (Leica).
For exosome tracking experiments in vivo, purified exosomes were labeled by using DiR dye (KeyGEN BioTECH, China), followed by 120000 g ultracentrifugation for 60 minutes, and labeled exosomes were resuspended in PBS. Labeled exosomes (50 μg/mice) in 100 μL PBS were injected into BALB/c mice via the tail vein. As a control, mice were injected with the same volume of PBS. After 12 and 24 h, an IVIS imaging system (PerkinElmer) was used for tracking exosome distribution in vivo. To evaluate exosome uptake by liver cells, all mice were sacrificed after 24 h, and the liver tissues were harvested for immunofluorescence on frozen sections. Image capture and photography were performed using a confocal microscopy (SP8, Leica).
Animal model induction and treatment
In each animal experiment, mice were randomly allocated to each group. All animal experiments were performed in accordance with protocols approved by the institutional review board of Department of Laboratory Animal Science, Fudan University, and conformed to the National Guidelines for Animal Usage in Research. All animal experiments were performed on 5-7 weeks male nude mice (BALB/c nu/nu) (SLAC Laboratory Animal Co., Ltd, Shanghai, China).
To assess the changes of acetyl-CoA and citrate levels in CAFs in CRLM, the mice were injected with exosomes (10 μg in 100 μL PBS) derived from HCT116 cells via tail vein every other day for 3 weeks. Subsequently, HCT116 cells were injected into the spleens of the mice to induce liver metastasis model. Mice were then treated with exosomes via tail vein injection twice a week continuously. Four weeks after CRC cells injection, all mice were euthanized and liver tissues were collected for the isolation of primary fibroblasts.
To evaluate the roles of exosomes in fibroblasts, 10 μg of exosomes in 100 μL PBS were intravenously injected into mice through the tail vein every other day for 3 weeks. Mice in control groups were injected with equal volume of PBS. At the endpoints, mice were euthanized and liver tissues were collected for western blot assay.
For analyzing the roles of exosomal HSPC111 in CRLM, mice treated with exosomes (10 μg in 100 μL PBS) via tail vein injection every other day for 3 weeks. The same volume of PBS was injected into mice in control groups. Mice then were assigned to groups randomly. For the tracking assay in vivo, firefly luciferase (Hanbio, Shanghai, China) was transfected into HCT116 cells and SW480 cells, and tumor formation and metastasis were detected using in vivo bioluminescence imaging. Subsequently, 1 × 107 luciferase-labeled HCT116 and SW480 cells were injected into the spleens of the mice to induce the liver metastasis model. Briefly, mice were anesthetized with an intraperitoneal injection of 1% pentobarbital sodium (50 mg/kg). The spleen was exteriorized gently through a left lateral flank incision and injected with1 × 107 luciferase-labeled HCT116 and SW480 cells suspension in 100 μL slowly using five-gauge needle. The spleen was replaced to the abdominal cavity, and 3-0 absorbable sutures were used for abdomen closure. The whole operation process followed the principles of aseptic surgery. Exosomes were injected into the tail vein twice a week continuously. The distribution of CRC cells in various organs was analyzed with IVIS Lumina LT series III (PerkinElmer, MA, USA). Fluorescence intensity within specific regions of individual mice was quantified using the region of the interest (ROI) tools in the Living Image 4.5 software (PerkinElmer). The ROI value of fluorescent area to liver surface area was used to reflect liver metastasis, and the metastasic lesions were determined by hematoxylin and eosin (H&E) staining. For studies of CXCR2 inhibitor in vivo, mice were treated with navarixin (CXCR2 inhibitor) 50 mg/kg body mass or vehicle (1% methyl cellulose) orally once daily for 4 weeks. Four weeks after CRC cells injection, mice were euthanized and liver metastasis were evaluated by HE staining.
Isolation and culture of primary fibroblasts
For the isolation of human primary liver fibroblasts, paired fresh CRLM and distant non-tumor liver tissues were obtained from six CRLM patients (male : female = 3 : 3, mean age 57 ± 13 years) and normal liver tissues were obtained from six hepatic hemangioma patients (male : female = 2 : 4, mean age 54 ± 12 years) undergoing partial liver resection at Department of General Surgery, Huashan hospital, Fudan University (Shanghai, China). None of the patients had received radiotherapy or chemotherapy prior to surgery. The specimens were used after obtaining written, informed consent from the patients and with the approval of the Ethical Committee of Huashan hospital, Fudan University (Shanghai, China). Tissues were finely minced with scalpel and enzymatically dissociated in 1mg/mL collagenase IV (Solarbio® Life Sciences, China) at 37 ℃ for 2 hours with frequent shaking. The cell suspensions were filtered through a sterile 40 μm cell strainer (BD Biosciences, Milan, Italy). The cells were centrifuged at 300 g for 5 minutes and resuspended in DMEM with 10% FBS in 100 mm dish. Cells were maintained at 37 ℃ in a 5% CO2 humidified atmosphere. After incubating for 24 hours, the cells were attached to the culture plate, and the unattached cells were removed. We used established primary liver fibroblasts at the passage number of 4-8. The primary cells from the marginal region of encapsulated CRLM were identified as CAFs while cells from distant non-tumor liver tissues were identified as non-tumoral fibroblasts (NTFs) and cells from normal liver tissues were identified as normal fibroblasts (NFs). The mice primary liver fibroblasts were isolated in the same manner as described above. The primary cells from CRLM nodules in mice were identified as CAFs while cells from distant non-tumor mice liver tissues were identified as non-tumoral fibroblasts (NTFs) and cells from normal mice liver tissues were identified as normal fibroblasts (NFs).
Immunofluorescence
For cell immunofluorescence, cells were plated in 35 mm glass-bottom dishes. After treatment, the cells were washed with PBS twice, fixed with 4% paraformaldehyde for 15 minutes, and then permeabilized in 0.1% Triton X-100 for 20 minutes. After being blocked with 5% bovine serum albumin, the cells were incubated with appropriate primary antibodies overnight at 4 ℃. Then the cells were incubated with Alexa Fluor 594- and Alexa Fluor 488-conjugated secondary antibody for 1 hour at room temperature followed by stained with DAPI before imaging under a fluorescent microscope.
For tissue immunofluorescence, tissues were fixed in a mix of 2% paraformaldehyde and 20% sucrose solution for 24 hours at room temperature, followed by embedding and section. Then tissue sections were stained with appropriate primary antibodies overnight at 4 ℃ in a humidified chamber. Then the sections were washed with PBS and incubated with secondary antibody conjugated to Alexa Fluor 594 and 488 for 1 hour at room temperature. Nuclei were stained with DAPI. Fluorescent images were taken with a fluorescent microscope. Information of the antibodies are listed in Additional file 1: Table S1.
Metabolomic analysis
LX-2 cells were seeded in 60 mm dishes (1 × 105 cells) followed by treatment with 100 μg exosomes derived from HCT116 cells and incubated for 48 hours. After incubation, the LX-2 cells were washed twice with cold PBS and frozen in liquid nitrogen for quenching metabolism. To analyze metabolomics, samples were analyzed by Omicsolution Co., Ltd (Shanghai, China). Briefly, liquid nitrogen snap-frozen samples were thawed on ice, and metabolites were isolated in precooled 50% methanol (v/v). Samples were then vortexed for 5 minutes and incubated for 10 minutes at room temperature. The isolated mixture was placed at -20 ℃ overnight. After centrifugation at 4000 g for 30 minutes (4 ℃), the supernatant was used for metabolomic analysis by LC-MS. Chromatographic separations were performed by using of an ultra-performance liquid chromatography (UPLC) system (Shimadzu, Kyoto, Japan). An ACQUITY UPLC ZIC-HILIC column (150 mm × 2.1 mm, 3.5 μm, Merck, Germany) was used for reversed phase separation, and the injection volume of each sample was 5 μL. The flow rate was 0.25 mL/min, and the column oven was maintained at 30 ℃. The mobile phase consisted with solvent A (0.2% (v/v) ammonia in water) and solvent B (0.1% (v/v) formic acid in acetonitrile). Gradient curve conditions (concentration of solvent B) were designed as follows: 0-1.5 min, 90% B; 1.5-25 min, 90%-40% B; 25-28 min, 40%-90% B; 28-33 min 90% B. Metabolites eluted from the column were detected by using a TripleTOF5600plus mass spectrometer (Shimadzu, Kyoto, Japan). Principal component analysis was applied to reveal the relationship between samples and the expression of metabolites. Variable importance in projection (VIP) was used to measure the different expression of each metabolite in each group, and VIP values > 1 were considered as differential expression. All differentially expressed metabolites were selected for heat map and enrichment analysis.
Proteomic analysis
LX-2 cells were plated into 60 mm dishes and cultured with HCT116-derived exosomes for 48 hours. Then the cells were collected for proteomics analysis. Isobaric tags for relative and absolute quantification (iTRAQ)-based proteomic analysis was conducted by Omicsolution Co., Ltd (Shanghai, China). Briefly, the cell lysate was extracted from each sample, and a total of 100 μg of each sample were removed and labeled using an iTRAQ-8plex kit (AB SCIEX, USA) according to the manufacturer’s instructions. Each set of labeled peptides was pooled and resuspended in strong cation exchange (SCX) prefractionation using SCX chromatography on an Ultremex SCX column (4.6 × 250 mm) with a 3000 high-performance liquid chromatography system (Thermo Fisher DINOEX, Waltham, MA, USA). Each fraction was analyzed using nano liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS); nano LC-MS was performed using a TripleTOF® 5600 mass spectrometer for MS/MS (AB SCIEX) interfaced with Eksigent nano LC system. The functions of the multiple protein contents were obtained by searching the UniProt database. Proteins were filtered by using the following thresholds: p value < 0.05 and 1.2-fold change. All differentially expressed proteins were selected for heat map and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis.
Lentivirus-mediated knockdown or overexpression of gene expression
The short hairpin RNA (shRNA) targeting HSPC111 (shHSPC111 #1, shHSPC111 #2), shRNA targeting ACLY (shACLY) and negative control (shCtrl #1, shCtrl #2) were inserted into the lentiviral vector produced by Hanbio (Shanghai, China). HCT116 cells were transfected with lentivirus (pHBLV-U6-MCS-CMV-ZsGreen-PGK-PURO) targeting HSPC111 and LX-2 cells were transfected with lentivirus (pHBLV-U6-MCS-CMV-ZsGreen-PGK-PURO) targeting ACLY at a multiplicity of infection (MOI) of 20. The sequences of shRNAs are presented in Additional file 2: Table S2.
Lentiviral vectors that overexpress full-length human HSPC111 (pHBLV-CMV-MCS-6x-His-HSPC111-EF1-ZsGreen-T2A-puro) or ACLY (pHBLV-CMV-MCS-3x-Flag-ACLY-EF1-ZsGreen-T2A-puro), and corresponding negative control (pHBLV-CMV-MCS-EF1-ZsGreen-T2A-puro) were purchased from Hanbio (Shanghai, China). The overexpressed lentiviruses targeting HSPC111 were transfected with SW480 cells and those targeting ACLY were transfected with LX-2 at a MOI of 20. All stable cell lines were obtained by 6 μg/mL puromycin (Hanbio) selection for 1 weeks followed by subsequent assays.
Clinical specimens
A total of 66 human non-metastatic primary CRC tissues (defined as NMCT) and paired peritumoral normal colorectal tissues (defined as NCT), and 136 human primary CRC tissues with liver metastasis (defined as CTLM) were collected between 2014 and 2019 from Department of General Surgery, Huashan hospital, Fudan University (Shanghai, China). All procedures were performed with the approval of the Ethical Committee of Huashan hospital, Fudan University (Shanghai, China). Patient consent was obtained before the start of the study. The general clinical information of all patients was included in Additional file 3: Table S3. The follow-up data were obtained at two mouth intervals through telephone calls, outpatient visits and office visits. All patients were followed until February 2021. Overall survival (OS) was defined as the interval between surgery and either death or the last observation taken.
Tissue Microarray (TMA)
Representative tumor areas were microscopically selected on hematoxylin and eosin (H&E) stained sections and highlighted with a marker on the section and similarly on the respective donor block. Tissue cores were punched from the defined regions on the donor block by using 2-mm punch needles and inserted into the receiver block with the help of a needle of larger caliber. The receiver blocks were placed upside down, covered with melted paraffin in a 50 ℃ oven for 5 minutes, in order to ensure the inserted core had melted in to the receiver block. The receiver blocks, immediately, were frozen for 20 minutes followed by histological sectioning and mounting onto slides.
Immunohistochemistry (IHC)
Sections (3 μm) were taken from the TMA paraffin blocks and mounted on slide glasses and then air dried overnight. The sections were deparaffinized in xylene for 5 minutes and rehydrated in a descending series of alcohol (100%, 100%, 95%, 80%, 70%) for 5 minutes each. After heat-mediated antigen retrieval by using citrate buffer, the sections were washed in distilled water and PBS. Endogenous peroxidase activity was blocked by a 15 minutes incubation in 3% H2O2 in distilled water. The sections were again washed in distilled water and PBS, and then incubated with appropriate primary antibody for 1 hour at room temperature. After washing in PBS for 10 minutes, the sections were incubated with a horseradish peroxidase (HRP)-labeled polymer (Dako EnVision + System) for 1 hour and washed in PBS for 10 minutes, and then incubated in 3,3-diaminobenzidine chromogen solution (Dako EnVision). The sections were slightly counterstained with Harris hematoxylin (Solarbio® Life Sciences, China), dehydrated in an ascending series of alcohol (95%, 100%, 100%, 100%), and cleared in xylene. Cover slides were placed on the section using a resinous mountant. Information of the antibodies are listed in Additional file 1: Table S1. The immunohistochemical analysis of HSPC111 in the TMA sections was performed using a semiquantitative scoring system ranging from 0 to 3 points. The presence of cytoplasmic staining was considered positive, and according to the staining intensity, the immunopositivity was graded as none (0), weak (1), moderate (2), and strong (3).
Acetyl-CoA and citrate measurement
The concentration of acetyl-CoA was detected by using an Acetyl-CoA assay kit (Sigma MAK039) according to manufacturer’s protocols. Briefly, cells or tissues were washed with cold PBS, harvested in RIPA buffer (Beyotime Biotech Co. Ltd., China), and then sonicated. The cellular or tissue protein was precipitated with PCA (BioVision K808-200). After centrifugation at 13000 g for 2 minutes, supernatant was recovered and neutralized by the addition of potassium bicarbonate until the pH of the sample was in the range of 6-8 and precipitates were removed by centrifugation. The supernatant was used to measure acetyl-CoA concentration in triplicate by using fluorescence assay method. Fluorescence intensity was measured (λex = 535/ λem = 587 nm) in black 96-well flat-bottom plates with clear bottoms.
The concentration of citrate was measured by using a citrate colorimetric assay kit (BioVision K655-100) according to manufacturer’s protocols. Briefly, cells were rapidly homogenized with citrate assay buffer, then centrifuged at 15000 g for 10 minutes to remove cell debris. The cellular protein was precipitated with PCA (BioVision K808-200). The sample was added into duplicate wells of a 96-well plate to bring the volume to 50 μL with a citrate assay buffer. After incubation at room temperature for 30 minutes, optical density (OD) value was measured at 570 nm.
RNA sequencing
The total RNA of the LX-2 cells incubated with appropriate exosomes for 48 h in vitro was extracted using TRIzol reagent (Invitrogen) and subjected to Biomarker Technologies (Beijing, China) for RNA sequencing. RNA purity was assessed using Nanodrop 2000 (ThermoFisher) and Bioanalyzer 2100 (Aglient). The sequencing libraries were constructed using NEBNext UltraTM RNA Library Prep kit for Illumina (NEB, USA). The library products were sequenced on an Illumina Hiseq 2500 platform, and paired-end reads were generated. The depth of sequencing coverage was 10 × and at least 10 Gb clean data were obtained from each sample. All usable reads were uniquely mapped to a gene that was used to evaluate the expression level. The DEGseq R package was used to analyze differentially expressed genes in two samples based on the conditions of a fold change (FC) ≥ 1.5 and p-values < 0.05. All differentially expressed genes were used for heat map analysis.
Co-immunoprecipitation (Co-IP)
For Co-IP analysis, LX-2 cells were washed twice with PBS and lysed in NP-40 lysis buffer (Solarbio® Life Sciences, China) supplemented with protease inhibitor cocktail (PIC) (Solarbio® Life Sciences, China) for 30 minutes on ice. The lysates were clarified by centrifugation at 12000 rpm for 15 minutes at 4 ℃, and then incubated with antibodies (Additional file 1: Table S1) for overnight at 4 ℃ followed by mixing with protein A/G magnetic beads (MCE, China). Immunocomplexes were washed five times with NP-40 buffer and resuspended in 4 × protein SDS-PAGE loading buffer followed by boiling for 10 minutes. The supernatants were collected by centrifugation at 12000 rpm for 5 minutes, followed by western blot analysis.
HEK293T cells were co-transfected with His-HSPC111 and Flag-ACLY lentivirus were lysed with NP-40 lysis buffer. Then the cell lysate was mixed with a His antibody or Flag antibody (Additional file 1: Table S1) for overnight at 4 ℃ followed by incubation with Anti-His Magnetic Beads (Sango Biotech, China) and Anti-Flag Magnetic Beads (MCE, China), respectively. Immunocomplexes were washed and boiled for western blot analysis.
Chromatin immunoprecipitation (ChIP)
ChIP was performed using a SimpleChIP® Enzymatic Chromatin IP kit (Cell Signaling Technology, 9003) according to the manufacturer’s instructions. Briefly, crosslinking of proteins-DNA was carried out using 37% formaldehyde for 10 minutes at room temperature. The crosslinking was quenched by adding glycine to a final concentration of 0.125M for 5 minutes at room temperature and followed by washing with cold PBS twice. Cells were scraped into PBS containing 1mM PIC. The suspension was centrifuged and the pellet was resuspended in buffer A + DTT + PIC followed by incubation on ice for 10 minutes. The pellet was mixed in 1 mL buffer B + DTT containing 5 μL micrococcal nuclease and incubated at 37 ℃ for 20 minutes. Nuclei were pelleted by centrifugation at 16000 g for 1 minute and resuspended in ChIP buffer. The nuclei pellet was further lysed by brief sonication (10 sets of 60 s pulses), and lysates were clarified by centrifugation at 9400 g for 10 minutes. The supernatant was incubated with ChIP-grade H3K27ac antibody (ab4729, Abcam) overnight at 4 ℃ with rotation. After the addition of ChIP-grade protein G magnetic beads, the samples were incubated for 2 hours at 4 ℃ with rotation. The protein G magnetic beads were pelleted by placing the tubes in a magnetic stand (MCE, China) and washed with three times low-salt washes and one time high-salt washes. Then DNA was eluted from the protein G magnetic beads by ChIP Elution Buffer, and the crosslinking was reversed by proteinase K digestion. The eluted DNA was purified using spin columns and detected by real-time PCR. The primer sets are listed in Additional file 4: Table S4. Real-time PCR amplification was performed using SimpleChIP® Universal qPCR Master Mix (Cell Signaling Technology, 88989).
Western blot analysis
The harvested cells or tissues were lysed in RIPA lysis buffer, and the lysates were cleared by centrifugation at 12000 rpm for 20 minutes (4 ℃). Protein concentrations were detected by BCA assay. Protein was separated by 10% or 15% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and then transferred to nitrocellulose membranes (Millipore, USA). Then the membranes were blocked with 5% nonfat milk and incubated with appropriate primary antibody overnight at 4 ℃. Subsequently, the membranes were incubated with HRP-conjugated secondary antibody. The protein complex was detected using enhanced chemiluminescence reagents (Millipore, USA). Quantitative analysis of western blot was performed with ImageJ software (ImageJ v1.8.0). Information of the antibodies are listed in Additional file 1: Table S1.
Quantitative real-time PCR (qRT-PCR)
Total RNA was extracted from cells with TRIzol reagent (Life Technologies, USA) according to the manufacturer’s instructions. The extracted total RNA was eluted in nuclease-free water and the concentration of the RNA was determined using NanoDrop 2000 (Thermo Fisher Scientific). The cDNA was synthesized with the PrimeScript RT Master Mix (Takara, Japan) using 500 ng total RNA as template. qRT-PCR was performed using TB Green Premix Ex Taq (Takara, Japan) on an ABI PRISM 7900 Sequence Detection System (Applied Biosystems, Carlsbad, USA). The mRNA level of GAPDH was used as an internal control. The results of qPCR were defined from the threshold cycle (Ct), and relative expression levels of mRNA were calculated with 2-ΔΔCt method. The primer sets for quantitative RT-PCR are listed in Additional file 4: Table S4.
Conditioned medium (CM) preparation
LX-2 cells were cultured with appropriate exosomes (20 μg/mL) from CRC cells for 48 h. Medium was collected and centrifuged at 300 g for 10 minutes to remove the cells, at 2000 g for 10 minutes to remove the dead cells and at 10000 g for 30 minutes to remove the cell debris (4 ℃). Then, the supernatant was filtered through a 0.025 μm filter (Millipore, USA) to remove exosomes and stored at -80 ℃ for further studies.
Enzyme-linked immunosorbent assay (ELISA)
In order to evaluate the protein level of CXCL5 in the CM, cells were cultured in the presence of appropriate exosomes (20 μg/mL) for 48 hours before supernatants were collected, and the CXCL5 level was detected by CXCL5 ELISA kit (EK0728, BOSTER) according to the manufacturer’s protocols.
Cell migration assay
HCT116 cells or SW480 cells (2.5 × 105/well) were plated on the upper part of a 24-well transwell chamber (Corning, NY, USA) in 200 μL specific CM. The medium in the lower chamber was supplemented with DMEM containing 10% FBS. After 24 h, cells that migrated to the underside of the membrane were fixed using methanol, stained with 0.1% crystal violet, and imaged and counted with a microscope (Leica, Wetzlar, Germany). All experiments were performed in triplicate.
Wound healing assay
HCT116 cells or SW480 cells (1 × 106/well) were plated into 6-well plates in 2 mL 10% DMEM medium. After cells were grown to a confluent layer, the cell monolayers were scratched using a pipette tip. The scratched cells were removed by gently washing with non-FBS DMEM gently. Then the cells were cultured with specific CM for 48 hours. Images were obtained at time point 0 and 48 hours.
Statistical analysis
All the data were presented as mean ± standard deviation of the mean (SD). SPSS software version 22.0 was used for statistical analyses. The differences between groups were assessed using the Student t-test, ANOVA test and χ2 test. Kaplan-Meier analysis and log-rank tests were applied for survival analysis. A p value of < 0.05 was considered statistically significant.