1. Introduction
Colon cancer is the third most common malignant tumor worldwide. The global disease burden is projected to increase to 3.2 million new cases and 1.6 million deaths by 2040. The shift toward earlier age of onset and increased probability of being diagnosed in people under the age of 50 is contributed by a mixture of genetic, lifestyle, and environmental risk factors. Endoscopic therapy, surgical therapy, adjuvant chemotherapy, and targeted therapy are common treatments for colon cancer. Immunotherapy can be used for early-stage colon cancer, and both overall endoscopic mucosal resection and endoscopic submucosal resection can be performed via endoscopy to remove large and complex lesions. Despite available treatment methods, the five-year survival rate and prognosis of colon cancer remain suboptimal, thus highlighting the critical need for understanding the underlying molecular mechanisms for advancing early diagnosis and treatment of colon cancer.
Recently, an increasing number of reports have shown that the tumor microenvironment (TME) is closely associated with tumor development and treatment outcomes. The TME is composed of a complex junction of diverse cell types including tumor cells, immune cells, endothelial cells, peripheral blood vessels, extracellular matrix, and cancer-associated fibroblasts [
1,
2]. These cells constantly interact and modify the environment in which the tumor survives. Among the varied cell types, macrophages hold an irreplaceable role in the TME. On the one hand, macrophages are the most abundant stromal cells and the most critical immune cells in the TME to combat tumor growth. On the other hand, the polarization of macrophages can lead them to play entirely opposite roles in tumor. The plasticity and heterogeneity of macrophages is reflected by the fact that the macrophage phenotype and function are regulated by the surrounding environment, enabling them to switch from one phenotype to another. Notably, macrophages found in the TME are known as tumor-associated macrophages (TAMs). The phenotype of TAMs changes under the influence of the microenvironment. Specifically, macrophages are classified into two primary phenotypes: pro-inflammatory/anti-tumor type M1 and anti-inflammatory/pro-tumor type M2. While TAMs generally appear as M1-type in the early stage, they can transform into M2-type under the influence of the TME to facilitate tumorigenesis [
3,
4,
5,
6]. M2 macrophages are known to promote an immune-suppressive TME and enhance tumor-cell survival and metastasis. Thus, investigating the molecular switch behind the temporal differentiation of TAMs is imperative for developing effective therapeutic strategies for inhibiting tumor growth.
In this study, we focused on the crosstalk between colon cancer-derived exosomes and TAMs. Extracellular vesicles (EVs) are microscopic vesicles secreted by cells, previously thought to merely function as cargos for excreting waste products. Advanced understanding has revealed that EVs are important cell–cell communication vectors, and carry numerous cellular components including proteins, lipids, polysaccharides, metabolites, DNA, and RNA [
7,
8]. EVs include microvesicles, exosomes, ectosomes, oncosomes, and cytoplasts [
9]. Exosomes are the smallest class of EVs, and are about 40~180 nm [
10]. In recent years, tumor-derived exosomes (TDEs) are thought to play an important role in cancer. Specifically, TDEs have been demonstrated to contribute to macrophage heterogeneity by carrying and delivering a variety of RNAs, including microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA) [
11,
12,
13]. For instance, Chen et al. found that lung adenocarcinoma cells transferred miR-19b-3p to macrophages via exosomes, which downregulated PTPRD and activated STAT3, resulting in the M2 polarization of macrophages [
14]. Moreover, Xiang et al. found that ovarian cancer-derived exosomes enriched with miR-222-3p downregulated SOCS3 and induced STAT3 activation, which resulted in the acquisition of M2 phenotype in macrophages [
15]. In contrast to direct miRNA-induced mRNA degradation, lncRNAs and cirRNAs are known to indirectly influence gene expression by sponging target miRNAs. Unlike miRNAs with their high expression levels and short sequences, lncRNAs are highly heterogeneous primary sequences of more than 200 nucleic acids in length, once considered transcriptional noise due to their lack of protein-coding ability [
16]. LncRNAs have enhancer-like properties and can interact with miRNAs, mRNAs, RNA-binding proteins, and transcription factors [
17,
18]. LncRNAs can also stabilize mRNA translation and act as precursors to miRNAs [
18]. More important, lncRNAs can act as competing endogenous RNAs (ceRNAs) to target and absorb miRNAs like sponges. The role of lncRNAs in tumorigenesis and macrophage polarization has also been reported. A study led by Zhang et al. showed that renal cell carcinoma-derived exosomes can transfer lncARSR to sponge miR-34/miR-449 and increase STAT3 expression in macrophages, thereby promoting M2-like macrophage polarization [
19]. Moreover, Huang et al. found that exosomal circSAFB2 is able to sponge miR-620 and activate the JAK1/STAT3 pathway as means of mediating M2 macrophage polarization [
20]. Notably, among the various signaling pathways reportedly involved in macrophage polarization, those involving PI3K/AKT, JAK/STAT, and MAPK/ERK are especially prominent [
12,
21,
22]. In colon cancer, the upregulation of lncXIST has been demonstrated to sponge miR-34a and activate the Wnt/β-catenin pathway to promote colon cancer development [
23]. However, it is unclear whether lncXIST plays a role in macrophage polarization. In this study, we investigated the molecular mechanism of colon cancer-derived exosomes and showed that lncXIST is able to mediate M2 macrophage polarization by regulating miR-17-5p and PDGFRA.
3. Discussion
The tumor microenvironment is a tumor-permissive milieu orchestrated by malignant tumor cells to facilitate continued growth and metastasis. Macrophages are integral components of the innate immune system and constitute a significant fraction of the TME. In fact, macrophages account for up to 50% of the mass in solid tumors [
27]. Macrophages exhibit high plasticity and heterogeneity and can be modulated by tumor cells to polarize into tumor-promoting M2 phenotype. As efficient cargos for promoting cell–cell communication, it has become increasingly evident that tumor-derived exosomes are abundant in the TME and play an important role in maintaining an immunosuppressive microenvironment. A growing body of research suggests that tumor cell-derived exosomes can promote tumor development by inducing macrophage M2 polarization [
28,
29,
30,
31,
32]. While most studies have focused on the effects of exosome-encapsulated miRNAs on macrophages, research on the effect of tumor derived exosomal lncRNAs remains scarce, particularly in the context of colon cancer. In this study, we focused on identifying the role of colon cancer exosomal lncRNAs on promoting macrophage polarization.
First, we confirmed that colon cancer cells retain the capacity to induce M2 macrophage polarization. During our investigation, we observed that THP-1 (Mφ) cells co-cultured with either CT26 or HCT116 cells demonstrated increased M2 macrophage phenotypic markers including CCL22, IL10, and PDL1. However, treatment with GW4869, a commonly used exosome inhibitor, reversed these effects. This finding led us to postulate that exosomes may act as crucial mediators in remodeling the macrophage phenotype. To verify this hypothesis, we isolated exosomes from CT26 and HCT116 cells through ultracentrifugation, and co-cultured them with THP-1 (Mφ) cells. Our results indicated that THP-1 (Mφ) cells treated with tumor-derived exosomes showed significantly increased M2-related proteins including ARG1, TGF-β, PDL1, and CD206. In contrast to M2 markers, M1 markers such as INOS and CD86 were either unchanged in the CT26-exo group, or significantly reduced in the HCT116-exo group. These results unequivocally confirm that colon cancer cell-derived exosomes have the ability to induce macrophages to polarize toward the M2 phenotype. Similar phenomena have been reported in other tumors, including bladder, breast, lung, and pancreatic cancers [
22,
33,
34,
35]. To examine the effect of polarized macrophages on colon cancer progression, medium from CT26-exo- and HCT116-exo-treated THP-1 (Mφ) cells was collected and used to treat CT26 and HCT116 cells. As a result, the rates of proliferation, migration, and invasion were significantly enhanced, indicating that M2 macrophages induced by tumor cell-derived exosomes can, in turn, enhance tumor cell growth. Interestingly, Lin et al. has reported that exosomes derived from bladder cancer cells polarized macrophages to the M2 phenotype, which, in turn, promoted migration and invasion of bladder cancer cells [
22]. Moreover, Chen et al. found that exosomes from adriamycin-resistant breast cancer cells induced M2 macrophage polarization to mediate proliferation, migration and invasion of breast cancer cells [
14]. These studies further demonstrate the reliability of the present findings.
Having confirmed the ability of colon cancer-derived exosomes to promote M2 macrophage polarization, we set out to explore the underlying mechanism. RNA-seq followed by KEGG and GSEA enrichment analyses were performed on HCT116-exo-treated THP-1 (Mφ) cells. Through Western blotting analyses, we confirmed that HCT116-exo and CT26-exo simultaneously promoted the activation of three M2 macrophage-related signaling pathways including PI3K/AKT, MAPK/ERK/ERK, and JAK-STAT in THP-1 (Mφ) cells. Although all three pathways have been separately demonstrated to be implicated in M2 macrophage polarization, the activation of all three at the same time has not been previously reported. A study led by Lin et al. showed that bladder cancer cell-derived exosomes can activate the PI3K-AKT pathway to induce M2 polarization [
22]. Qiu et al. found that gastric cancer cell-derived exosomes are able to induce M2 macrophage polarization by activating the MAPK/ERK/ERK signaling pathway [
26]. In addition, Chen et al. showed that exosomes from lung adenocarcinoma cells could polarize macrophages toward the M2 phenotype by activating STAT3 [
14]. Moreover, Cai et al. demonstrated that STAT6 was an important factor in M2 polarization by oral squamous cell carcinoma-derived exosomes [
25]. In the present study, we hypothesize that colon cancer cell-derived exosomes may activate multiple pathways in the process of promoting M2 macrophage polarization. By screening for common DEGs between these three signaling pathways, we found that PDGFRA was most significantly upregulated in CT26-exo/HCT116-exo-treated THP-1 (Mφ) cells, both at the mRNA and protein levels. PDGFRA encodes the platelet-derived growth factor (PDGF) receptor α protein. When activated by PDGF, PDGFRA can, in turn, initiate key downstream PI3K-AKT, MAPK/ERK/ERK, and JAK-STAT pathways to regulate cell proliferation, growth, and differentiation [
36,
37]. Aberrant activation of PDGFRA has been reported in gastrointestinal mesenchymal tumors, glioma tumors, and gastric cancer [
38]. However, the role of PDGFRA in M2 macrophage polarization remain underexplored.
To explore potential exosome-encapsulated upstream regulators of PDGFRA, we first screened for potential target miRNAs using miRtarbase, TargetScan, miRDB, Tarbase, and Pic Tar databases. Subsequent qRT-PCR validation revealed that miR-17-5p expressions were significantly reduced in CT26-exo- and HCT116-exo treated THP-1 (Mφ) cells. Furthermore, through Starbase, miRcord, as well as colon cancer GEO databases (GSE186577, GSE84983, and GSE104364), we pinpointed to lncFGD5-AS1, lncSNHG14, and lncXIST as potential miR-17-5p-targeting lncRNAs. Verification via qRT-PCR indicated that lncXIST was highly expressed in colon cancer cells. Moreover, lncXIST was found encapsulated and highly enriched in colon cancer cell-derived exosomes, as well as colon cancer-derived exosome-treated THP-1 (Mφ) cells. In addition, lncXIST knockdown HCT116 cells were co-cultured with THP-1 (Mφ) cells. The results indicated that knockdown of lncXIST significantly reversed M2 macrophage polarization. Meanwhile, dual-luciferase reporter gene assay also confirmed the direct interaction between lncXIST and miR-17-5p, as well as between miR-17-5p and PDGFRA. Based on the above results, this study conjectured that lncXIST was transferred from colon cancer cells to THP-1 (Mφ) cells via exosomes. In addition, lncXIST could sponge miR-17-5p and, thereby, enhance PDGFRA expression and promote M2 macrophage polarization. To verify this, we knocked down and overexpressed lncXIST in THP-1(Mφ) cells and observed changes in miR-17-5p and PDGFRA levels by qRT-PCR. Results showed that the knockdown of lncXIST led to an increased expression of miR-17-5p and a reduced expression of PDGFRA. On the contrary, the overexpression of lncXIST inhibited miR-17-5p, but promoted PDGFRA expression. Furthermore, we overexpressed both lncXIST and miR-17-5p in THP-1 (Mφ) cells and found that elevated miR-17-5p expression inhibited PDGFRA expression as well as the degree of M2 macrophage polarization. Altogether, our data suggest that lncXIST from colon cancer-derived exosomes promotes PDGFRA expression by inhibiting miR-17-5p.
miR-17-5p is a member of the miR-17-92 cluster, a family whose organization and sequence are highly conserved among vertebrates, and whose evolutionary process has produced two mammalian paralogs due to gene duplication and deletion events: the miR-106b-25 cluster and the miR-106a-363 cluster. This means that miR-106 is a homolog of miR-17 [
39]. miR-17-5p [
40] is an important regulator of cell proliferation, autophagy and apoptosis, and plays an important regulatory role in cancer [
41]. Xu et al. found that lncNEAT1 promotes angiogenesis in gastric cancer by sponging miR-17-5p to increase the expression of TGF-βR2. Chen et al. found that lncPART1 promotes non-small cell carcinoma by sponging miR-17-5p to increase the expression of TGF-βR2 expression to promote the progression of non-small cell lung cancer cells [
42]. In addition, Xu et al. found that lncMIR17HG promotes colon cancer progression by sponging miR-375 to increase the expression of NF-κB/RELA [
43]. lncXIST is known for mediating transcriptional silencing of genes on the X chromosome and acts as a dosage compensation between male and female chromosomes, but it still has a developmental role in various diseases, including stroke, inflammation, cardiomyocyte hypertrophy, and cancer [
44,
45]. lncXIST was found to be involved in the development of various types of cancer and is dysregulated in tumor cells, promoting tumor proliferation, migration, angiogenesis, and chemotherapy resistance [
46,
47,
48]. Wei et al. found that lncXIST is upregulated in pancreatic cancer tissues and cell lines and participated in promoting EGFR expression by sponging miR-133a to promote pancreatic cancer proliferation [
49]. Liu et al. found that lncXIST was upregulated in thyroid cancer tissues and cell lines and promoted the activation of the MET-PI3K-AKT signaling pathway through sponging miR-34a to promote the development of thyroid cancer [
50]. LncXIST was also found to be upregulated in colon cancer tissues and cell lines. Specifically, Sun et al. found that lncXIST activated the Wnt/β-catenin pathway by sponging miR-34a to promote colon cancer development [
21]. Yang et al. found that METTL14 inhibited colorectal cancer development by suppressing lncXIST [
51]. In addition, lncXIST is also closely related to exosomes. Cheng et al. found that lncXIST-containing exosomes secreted by pancreatic cancer cells increased the expression of GDNF to promote perineural infiltration of pancreatic cancer cells by sponging miR-211-5p [
52]. Overall, these studies provide additional support for the reliability of the present finding, that lncXIST can be carried by exosomes and act as sponges for target miRNAs to promote colon cancer development.
Altogether, the results from this study demonstrated that exosomal lncXIST can induce macrophage M2 polarization by regulating the miR-17-5p/PDGFRA axis to promote colon cancer progression. However, there are some limitations of this study. First, this study mainly used colon cancer cell lines for in vitro experiments, while subsequent validation in more colon cancer cell lines and in vivo animal experiments are needed. Second, our data only demonstrated that the upregulation of PDGFRA promoted M2-like macrophage polarization, but not its activation. Further validation is required. Third, while PDGFRA was a common gene found for the AKT-, ERK-, and STAT3/6 signaling pathways, the effect of the lncXIST/miR-17-5p/PDGFRA axis on the activation of each pathway requires further examination. Fourth, the specific mechanisms by which M2-like macrophages promote colon cancer cell proliferation, migration, and invasion have not been explored in this study. To elaborate on this section, the ability of CT26 and HCT116 cells to proliferate, migrate, and invade needs to evaluated when co-cultured with culture medium from M2-like macrophages. Moreover, the levels of lncXIST, miR-17-ap, and PDGFRA should also be evaluated in the treated cancer cells. Furthermore, it is noteworthy that the repertoire of bioactives encapsulated in colon cancer-derived exosomes extends beyond lncXIST, potentially harboring additional factors that contribute to the induction of macrophage M2 polarization and, subsequently, facilitate colon cancer progression. These unexplored bioactives represent intriguing avenues for future research, as they may hold the key to unraveling novel mechanisms underlying the intricate interplay between the tumor microenvironment and cancer development. Therefore, further investigation into these exosomal contents is warranted to gain a more comprehensive understanding of their roles in modulating macrophage polarization and promoting colon cancer progression, ultimately informing the development of targeted therapeutic strategies.
4. Materials and Methods
4.1. Cell Lines and Cell Culture
CT26, HCT116, NCM460, and 293T cells were purchased from the Chinese Academy of Sciences CellBank (Shanghai, China). THP-1 (human monocytic-leukemia cells) cells were purchased from Wuhan Prolife Technology Co. (Wuhan, China). CT26 and HCT116 cells were cultured in RPMI-1640 medium (Hyclone) with 10% fetal bovine serum (GIBCO BRL, Grand Island, NY, USA) and 1% penicillin. NCM460 and 293T cells were cultured in DMEM high glucose medium (GIBCO BRL, Grand Island, NY, USA) with 10% Newzerum fetal bovine serum (NEWZERUM Ltd., Christchurch, New Zealand)and 1% penicillin. THP-1 cells were cultured using THP-1 complete medium (Wuhan Pricella Biotechnology Co., Ltd., Wu Han, China). All cells were maintained at 37 °C containing 5% CO2.
4.2. Macrophage Induction from Monocytes
At approximately 80–90% confluency, THP-1 cells were centrifuged and the supernatant was aspirated, and the cells were resuspended using 1 mL β-mercaptoethanol-free THP-1 complete medium. After counting, 106 cells were taken and inoculated in a single well of a 6-well plate, followed by the addition of 2 μL of 100 μg/mL phorbol-12-myristate-13-acetate (PMA; Sigma-Aldrich, St. Louis, MO, USA) (final concentration of 100 ng/mL) for induction. THP-1 cells were fully differentiated into macrophages (Mφ) after 48 h.
4.3. Co-Culturing System
To mimic exosome-mediated intercellular communication between colon cancer cells and TAMs, an in vitro indirect co-culture system was established in which macrophages and colon cancer cells were inoculated in the upper and lower chambers of transwell cell culture inserts with polycarbonate membranes, respectively. After 48 h, the cells were harvested.
4.4. Cell Transfection
Transfection was carried out using jetPRIME buffer (Polyplus-transfection SA, Illkirch, France) according to manufacturer’s protocol. Plasmids and siRNAs were designed and constructed by GenePharma (Shanghai, China). Sequence information is listed in
Supplementary Table S2.
4.5. Extraction of Exosomes
CT26 and HCT116 cells were cultured using complete medium prepared with exosome-free serum; cell culture medium was collected into 50 mL centrifuge tubes when the cell coverage reached 90%. The cells were then centrifuged at 4 °C for 5 min at 800× g, and again for 10 min at 2000× g. The supernatant was aspirated with a syringe and paired with a 0.22 μm filter for filtration, and the filtered supernatant of the cell culture medium was transferred to a sterile Beckman centrifuge tube and centrifuged in a Beckman ultracentrifuge (BeckmanCoulter, Miami, FL, USA) with the parameters of 36,900 speed (100,000× g) at 4 °C for 2 h. After centrifugation, the tubes were transferred to an ultraclean bench, where the supernatant was discarded, and exosomes were resuspended with PBS.
4.6. Transmission Electron Microscopy (TEM)
First, 10 μL of exosome suspension was added dropwise onto a copper mesh, and let stand for 1 min. The floating liquid was then absorbed using filter paper. Next, 10 μL of 1% phosphotungstic acid staining solution was added for staining. After 5 min, excess liquid was absorbed using filter paper. Then, 10 μL of purified water was added dropwise and absorbed with filter paper in order to remove the excess dye. After drying at room temperature, the morphology of exosomes was then observed and photographed using transmission electron microscopy (Thermo FisherScientific, Waltham, MA, USA).
4.7. Flow Cytometry of Exosomes
First, 10 μL of exosome suspension was withdrawn and subjected to serial dilutions using a flow-through sheath solution. Various concentrations of the diluted exosome suspension were then introduced into the detection system to ascertain the optimal particle count. The optimal dilution concentration was meticulously determined based on the real-time monitoring of particle numbers (less than 10,000 particles). Subsequently, the exosome suspension at the determined concentration was evenly apportioned into 3 distinct centrifuge tubes. One tube was labeled blank, the second and third tubes were labeled PE-CD81 and PE-CD63, respectively. These samples were then incubated under standardized conditions of 15 min at room temperature, ensuring adequate protection from light exposure. Following incubation, the prepared samples were analyzed using a CytoFLEX flow cytometer (Beckman Coulter, Miami, FL, USA) for further characterization and analysis.
4.8. Exosome Labeling
Exosomes were labeled using the PKH26 Infrared Fluorescent Cell Crosslinker Kit (Sigma-Aldrich, St. Louis, MO, USA). First, 50 μL of the exosome suspension and 250 μL of Diluent C solution were gently mixed by pipetting in a centrifuge tube. In a separate centrifuge tube, 5 μL of PKH26 dye and 250 μL of Diluent C solution were gently mixed by pipetting. The mixtures from the two centrifuge tubes were then combined and gently mixed by pipetting. The resulting mixture was incubated at room temperature for 5 min to allow for efficient labeling. To terminate the reaction, 2 mL of 5% BSA was added at the end of the incubation period. Finally, the labeled exosomes were subjected to ultracentrifugation and resuspended in 50 μL of PBS for further analysis.
4.9. Fluorescence Confocal Microscopy
THP-1 cells were seeded onto cell crawls positioned in 24-well plates and, subsequently, differentiated into macrophages by treating with 0.5 μL of 100 μg/mL PMA for 48 h. Following differentiation, the cells were further incubated for 24 h in the presence of PKH26-labeled exosomes (red). Subsequently, 5 μL of Hoechst33342 live cell staining solution (blue) was added to each well containing 500 μL of medium, followed by incubation at 37 °C for 10 min. At the end of the incubation, the dye-containing medium was discarded and cells were washed twice with PBS. Cytoskeleton staining was performed using β-tubilin (green). The cells were fixed with 500 μL of 4% paraformaldehyde for 15 min, followed by 3 washes with PBS. Each well was then incubated with 500 μL of PBS containing 0.1% Triton X-100 for 2 min to permeabilize the cells. The cells were then blocked at room temperature for 40 min using skim milk powder on a shaker. The cells were then washed and incubated with primary β-tubilin antibody diluted in blocking solution overnight at 4 °C. Following incubation, the cells were then washed and incubated with Alexa Fluor® 488 Donkey Anti-Mouse IgG (H + L) secondary antibody diluted in skimmed milk powder for 1 h at room temperature in the dark. The cell crawls were then removed and mounted on clean slides using fluorescent sealer (NaHCO3 3.7 g, anhydrous Na2CO3 0.6 g dissolved in 100 mL ddH2O and mixed 1:1 with glycerol). Finally, the slides were allowed to dry completely before being analyzed using the fluorescence confocal microscopy (Leica, Wetzlar, Germany).
4.10. Western Blotting Analysis
Total cellular and exosomal protein samples were obtained using Radioimmunoprecipitation Assay (RIPA) Lysis and Extraction Buffer supplemented with a mixture of protease and phosphatase inhibitors. The resulting lysates were quantified using Bradford’s reagent (Shanghai Beyotime Biotechnology Co., Ltd., Shanghai, China). Protein samples were separated by 10% SDS-PAGE and transferred onto a polyvinylidene difluoride (PVDF) membrane. The membrane was then blocked in 5% skim milk for 1h at room temperature followed by incubation with primary antibody at 4 °C overnight. Following incubation, the membrane was washed three times with TBST and subjected to secondary antibody incubation for 1 h at room temperature. Protein bands were detected and analyzed under chemiluminescence (Bio-Rad, Hercules, CA, USA).
4.11. Reagents and Antibodies
DMSO and GW4869 were purchased from Sigma (Sigma, USA) and MedChemExpress (MedChem Express, Monmouth Junction, NJ, USA), respectively. PE-CD63 and PE-CD81 antibodies were purchased from Abgent (San Diego, CA, USA). p-AKT(Thr308), p-AKT(Ser473), and p-ERK antibodies were purchased from Cell Signaling Technology (Danvers, MA, USA). p-STAT3, STAT3, p-STAT6, STAT6, CD63, CD81, and TSG101 primary antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Alexa Fluor®488 donkey anti-mouse IgG (H + L) secondary antibody was purchased from Jackson Immunoresearch Laboratories (West Grove, PA, USA). Calnexin, PDGFRA, and GAPDH antibodies were purchased from Shanghai PoWan Biotechnology Co. (Shanghai, China). β-Tubulin, AKT, and ERK antibodies were purchased from Wuhan Sanying Biotechnology Co. (Wuhan, China). ARG1, TGF-β, PDL1, and CD206 antibodies were purchased from Hangzhou Jingjie Bioscience and Technology Co., Ltd. (Hangzhou, China).
4.12. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR)
Total RNA was extracted using TRIGene reagent (Genestar, Beijing, China). miRNA was extracted using Exosomal RNA Isolation Kit (Norgen Biotek, Thorold, Ontario, Canada). DNase I, amplification grade (Invitrogen) was used to digest DNA contaminants. The concentration and quality of RNA samples were detected using microspectrophotometer Nanodrop 1000 (Thermo Fisher Scientific, Waltham, MA, USA). Complementary DNA (cDNA) synthesis of mRNA was performed using a reverse transcription kit Hifair
® II 1st Strand cDNA Synthesis SuperMix for Qpcr (Yeasen, Shanghai, China) and miRNA was performed using a reverse transcription kit StarScript III RT Kit (Genestar, Beijing, China). SYBR Green PCR premix (Epizyme, Shanghai, China) was used for qRT-PCR on a CFX96 Touch real-time fluorescent quantitative PCR detection system (Bio-Rad, USA). Relative level of gene expression was calculated using the 2
−ΔΔCt method. qRT-PCR primer (Sangon Biotech, Shanghai, China) sequences are shown in the
Supplementary Table S1.
4.13. MTS Proliferation Assay
Cell proliferation was analyzed through MTS analysis. First, 2.5 × 104 cells were cultured in 96-well plates under treatment for 24 h. Then, 10 μL of MTS was added to each well and incubated in the dark at room temperature for 2 h. The absorbance was measured at 490 nm using a colorimeter(Omega Biotech, Doraville, GA, USA).
4.14. Transwell Assay
Cells were digested, centrifuged, and resuspended with serum-free medium. CT26 and HCT116 cells were plated in transwell chambers containing serum-free medium. In the lower chamber of a 24-well plate, 400 μL of complete medium culture medium was added to each well, to which an additional 200 μL of CT26-exo/HCT116-exo-treated macrophage medium was added to the experimental group and 200 μL of PBS-treated macrophage medium was added to the control group. Using 500 μL of 4% paraformaldehyde, the cells were placed into the chambers and fixed for 15 min. Paraformaldehyde was aspirated and discarded, and 0.1% crystal violet staining solution was added for 30 min, followed by imaging under a microscope. The invasion assay needs to be pre-lined with organoGel (Guangzhou Ebix Biotechnology Co., Ltd., Guangzhou, China) in the invasion experiments.
4.15. Dual-Luciferase Reporter Assay
Fluorescein-labeled reporter gene detection was assessed using Dual-luciferase Assay (Shanghai Beyotime Biotechnology Co., Ltd., Shanghai, China). The sequences were constructed in pmirGLO plasmid by Beijing Prime Biotechnology Co. (Beijing, China). First, 293T cells were seeded in 96-well plates and cultured for 24 h. The cells were then co-transfected with either wild-type or mutant vectors with NC or miR-17-5p mimics. After a 48h incubation period, passive lysis buffer was added to each well, and the supernatant was extracted for further analysis. Lysis buffer sample (40 μL) and firefly fluorescein lyase assay solution (100 μL) were mixed and added to each well of the 96-well plate. Absorbance was measured at 580 nm using a microplate reader (Omega, Germany). Sea kidney fluorescinase assay working solution (100 μL, sea kidney fluoresceinase assay substrate: sea kidney fluoresceinase assay buffer = 1:100) was then added to each well and the absorbance was measured at 460 nm.
4.16. Bioinformatics Analysis
4.17. Statistical Analysis
The data were statistically analyzed and graphed using GraphPad Prism 8.0. The luminescence results of Western blotting bands were analyzed using ImageJ (Version 1.53m). T-test was used to analyze two independent samples with normal distribution, and one-way ANOVA test was used to comparing multiple samples. Multiple t-tests (and non-parametric tests) were used for comparing 2 independent samples with multiple data sets. Experiments were repeated independently 3 times. NS indicates no significance, * indicates p < 0.05, ** indicates p < 0.01, *** indicates p < 0.001, **** indicates p < 0.0001.