Highlights
-
•
Exosomes play essential role in the metastasis of colorectal cancer from TME aspect.
-
•
Finding out the prominent regulating exoDEPs by label-free proteomics in this research provided a lot of key information of CRC metastases.
-
•
Metabolism, cytoskeleton-related pathways and immunosuppression are two key mechanisms by which exosomes regulate CRC malignant behavior.
-
•
The discovery of the “all or none” exoDEPs was of great significance. The exoDEPs expressed only in SW620 cells can more clearly show their ability to promote the invasion and metastasis of CRC cells.
Keywords: Exosomes, Label-free proteomics, Tumor microenvironment (TME), Metastasis, Colorectal cancer
Abstract
Exosomes are secreted nanovesicles consisting of biochemical molecules, including proteins, RNAs, lipids, and metabolites that play a prominent role in tumor progression. In this study, we performed a label-free proteomic analysis of exosomes from a pair of homologous human colorectal cancer cell line with different metastatic abilities. A total of 115 exoDEPs were identified, with 31 proteins upregulated and 84 proteins downregulated in SW620 exosome. We also detected 30 proteins expressed only in SW620 exosomes and 60 proteins expressed only in SW480 exosomes. Bioinformatics analysis enriched the components and pathways associated with the extracellular matrix, cytoskeleton-related pathways, and immune system changes of colorectal cancer (CRC). Cellular function experiments confirmed the role of SW620 exosomes in promoting the proliferation, migration, and invasion of SW480 cells. Further verifications were performed on six upregulated exoDEPs (FGFBP1, SIPA1, THBS1, TGFBI, COL6A1, and RPL10), three downregulated exoDEPs (SLC2A3, MYO1D, and RBP1), and three exoDEPs (SMOC2, GLG1, and CEMIP) expressed only in SW620 by WB and IHC. This study provides a complete and novel basis for exploring new drug targets to inhibit the invasion and metastasis of CRC.
Introduction
Colorectal cancer (CRC) is the third most common tumor and the second leading cause of cancer-related mortality worldwide [1]. Although modern research has shed light on the pathogenesis of CRC and provides enhanced screening strategies, the prevalence of CRC is still increasing. Despite an improved outcome with the current treatment regimen, a huge number of patients relapse, have distant metastasis, and develop drug-resistant disease within 5 years after operation [2,3]. More than 50% of CRC patients have distant metastasis, particularly in the liver, lungs, and bone. Patients with distant metastasis, notably liver metastasis, have a 5-year survival rate below 50%; however, patients without metastasis can largely be cured by resection of the primary tumor and have a 5-year survival rate exceeding 80%. Therefore, it is very important to clarify the mechanism of CRC invasion and metastasis and identify the key regulating proteins as new biomarkers or therapeutic targets for improving the prognosis of CRC patients.
The tumor microenvironment (TME) has been identified as a resident of a variety of cells, including tumor cells, stromal cells, extracellular matrix (ECM), immune cells, blood vessels, and differentiated cells [4]. It establishes a communication network for crosstalk and signaling between tumor cells, stroma, and other interstitial cells. It is vital for the initiation of tumor proliferation and angiogenesis, evasion of apoptosis and immune surveillance, suppression of the immune system, and metastasis. The intercellular communication of the TME creates a niche for reprogramming resident cells, extracellular matrix, and angiogenesis, which prompts the metastasis of cancer cells.
Exosomes are vital cell-cell communicators of information within the TME by horizontal transfer of proteins, lipids, DNA, and RNA [5]. It is a small vesicle with a diameter of 30–150 nm released from the majority of cell types and has a typical lipid bilayer structure [6]. Exosomes exist in cell culture supernatants, serum, plasma, saliva, urine, amniotic fluid, and other biological fluids. Tumor-derived exosomes modulate stromal cells, reprogram ECM, induce transformation of normal cells adjacent to the tumor, stimulate endothelial cells to form vascular, and alter the invasive behavior of tumor cells. Exosomes play an important role in regulating tumor metastasis, especially liver metastasis, through various functions. For example, exosomes from the TME of CRC can reprogram the cancer cell metabolic machinery to increase glycolysis by inhibiting mitochondrial oxidative phosphorylation and contribute toward immune-induced tumor dormancy and antitumor immunotherapy. Identifying the key regulatory exosomal proteins related to metastasis and elucidating their functions are very important for exploring new therapeutic targets for inhibiting CRC metastasis.
Proteomics is considered valuable for early disease diagnosis, prognosis, and monitoring of disease development. Label-free, or unlabeled quantitative technology, is the mass spectrometry analysis of protein enzymatic peptides by liquid chromatography-mass spectrometry (LC-MS) [7]. It overcomes the limitations of traditional isotope labeling, which necessitates a high sample concentration. It is not limited by the labeling kit and can be used for mass sample detection. Moreover, it has less pre-processing, more original information of samples, and a higher coverage for low-abundance peptides to determine the most precise information.
SW620 (highly metastatic) and SW480 (weakly metastatic) are a pair of homologous human CRC cell line with different metastatic abilities. In this study, we first performed the cellular function experiments and confirmed the promoting role of SW620 exosomes on the proliferation, migration, and invasion of SW480 cells. We then used a label-free differential proteomics on this pair of cell lines to identify the exosomal differentially expressed proteins (exoDEPs) regulating CRC metastasis in the TME. After analysis, 115 exoDEPs (31 upregulated and 84 downregulated in SW620 exosomes), and 90 “all or no” exoDEPs (30 only in SW620 and 60 only in SW480) were identified. Bioinformatics analysis enriched the components and pathways associated with the ECM, cytoskeleton-related pathways, and immune system changes of CRC. Further validation results of the 12 selected exoDEPs were consistent with the proteomics results. The exoDEPs identified in this study provide new potential therapeutic targets and research directions in the TME for CRC metastasis.
Materials & methods
Ethics
This study was approved by the Institutional Review Board of the First Affiliated Hospital of Dalian Medical University. Written informed consent was obtained from all patients.
Cell culture
CRC cell lines SW620 (highly metastatic) and SW480 (weakly metastatic) were purchased from the Chinese Academy of Sciences cell bank. All cell lines were incubated in DMEM (Gibco, Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco, Thermo Fisher Scientific), 100 U/mL penicillin G, and 100 μg/mL streptomycin (Gibco, Thermo Fisher Scientific) at 37 °C and 5% CO2/95% air.
Exosome extraction
SW480 and SW620 cells were cultured in complete growth medium. After the cells were cultured to nearly 70% confluence, the medium was replaced with exosome-free medium, and the cells were incubated for an additional 48 h. The primary supernatants were collected and centrifuged at 4 °C for 30 min at 2 × 103 g to remove dead cells for purification (three biological replicates). Next, we isolated the extracellular vesicles (EVs) by ultracentrifugation. In brief, the purified supernatants were placed in ultra-high-speed centrifuge tubes (balanced with 1×PBS) and centrifuged at 110 × 103 g for 75 min at 4 °C. We then obtained the exosome pellets and resuspended it in 1 × PBS. The EVs suspension was added to the purification column and centrifuged at 4 °C for 5 min at 4 × 103 g, and the effluent was the purified exosomes. An electron micrograph and nanoparticle tracking analysis (NTA) of purified exosomes were subsequently performed.
Protein extraction and determination of protein concentration
A 200 µL SDT lysate (4% SDS, 100 mM Dithiothreitol (DTT), and 100 mM Tris HCl) was added to each purified exosome sample. The exosomes were broken by ultrasound for 2 min, soaked in boiling water for 5 min, and centrifuged at 20 × 103 g at 4 °C for 20 min. The supernatant was collected, and the protein was quantified using the BCA method. The FASP method was used for the enzymolysis. The steps were as follows: an appropriate amount of DTT was added to each sample until its final concentration was 100 mm, boiling water bath for 5 min, and cooling to room temperature, 200 µL UA buffer (8 m urea, 150 mm Tris HCl, pH 8.0) was added and mixed well. The sample was transferred to a 10 kDa ultrafiltration centrifuge tube and centrifuged at 12 × 103 g for 15 min, 200 µL UA buffer was added and centrifuged 12 × 103 g for 15 min and the filtrate was discarded. A solution of 100 uL IAA (50 mM IAA in UA) was added, oscillated at 600 rpm for 1 min, kept away from light for 30 min, and centrifuged at 12 × 103 g for 10 min. A solution of 100 uL UA buffers added, centrifuged 12 × 103 g for 10-min and repeated twice. A solution of 100 uL NH4HCO3 buffer was added and centrifuged for 14 × 103 g, and this step was repeated twice for 10 min. A solution of 40 uL trypsin buffer (6 μg trypsin in 40 uL NH4HCO3 buffer) was added, centrifuged at 600 rpm for 1 min, and stored in 37 °C for 16–18 h. After centrifugation at 12 × 103 g for 10 min, the filtrate was collected, and 0.1% Trifluoroacetyl (TFA) solution was added. The peptides were desalted using a C18 cartridge and freeze-dried under vacuum. The peptides were dried and dissolved in 0.1% FA. The peptide concentration was determined using LC-MS analysis.
LC-MS/MS analysis
An appropriate amount of the peptide was taken from each sample and separated using an easy NLC 1200 chromatography system (Thermo Fisher Scientific, Waltham, MA, USA). Buffer solution: 0.1% formic acid solution; B solution: 0.1% formic acid acetonitrile solution (acetonitrile: 85%). The column was equilibrated to 95%. The sample was injected into a trap column (100 µm × 20 mm, 5 um, C18, Dr. Maisch GmbH) and then separated on a chromatographic column (75 µm × 150 mm, 3 um, C18, Dr. Maisch GmbH) at the flow rate of 300 NL/min. The liquid gradient was set as follows: 0–2 min, with a linear gradient of liquid B as 5%–8%; 2–90 min, with a linear gradient of liquid B as 8%–23%; 100 min, with a linear gradient of liquid B as 23%–40%; 100–108 min, with a linear gradient of liquid B as 40%–100%; and 120 min, with a linear gradient of liquid B as 100%. The peptides were separated and analyzed by data-dependent acquisition mass spectrometry with a Q-active plus mass spectrometer (Thermo Fisher Scientific, Waltham, MA, USA). The analysis time was 120 min; detection mode, positive ion; scanning range of parent ion, 300–1800 m/Z; resolution of primary mass spectrometry, 70 × 103 m/Z200; AGC target, 1E6; and primary maximum it, 50 ms. Peptide secondary mass spectrometry analysis was performed according to the following methods: MS2 scan was triggered after each full scan; resolution, 17,500 at m/z 200; AGC target, 1E5; maximum, 50 ms; MS2 activation type, HCD; isolation window, 1.6th; and normalized collision energy, 27.
Protein identification, quantification, and bioinformatic analyses
The mass spectrometry database retrieval software used in this project is maxquant1.6.0.16. The protein database is from the UniProt protein database (download: 08/05/2019; Homo sapiens: 173,282 entries). Maxquant library search software analysis parameter settings were as follows: Enzyme: Trypsin; Max Missed Cleavages: 2; Main search Peptide Tolerance: 4.5 ppm; First search Peptide Tolerance: 20 ppm; First search Peptide Tolerance: 20 ppm; Fixed modifications: Carbamidomethyl (C); Variable modifications: Oxidation(M), Acetyl (Protein N-term); PSM (Peptide-Spectral Matching) FDR: 0.01; Protein FDR: 0.01; Protein quantification: Razor and unique peptides were used; LFQ: True; LFQ min. ratio count: 1; Match between runs: True.
After analyzing, all exosomal proteins were reliably identified and quantified based on P-value,0.05. Protein ratios with 2.0-fold change was considered to be differentially expressed. The proteins identified obviously more than 2.0-fold change between this pair of cell lines were identified as “all or no” proteins. For cell components, biological processes and molecular functions were identified using R software (version 3.6.3) for gene ontology (GO) annotation. Pathway analysis and enrichment of exoDEPs were annotated using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (http://www.genome.jp/kegg/pathway) using R software (version 3.6.3). For GO and KEGG analysis, seven R software packages, namely, “colorspace,” “stringi,” “ggplot2,” “DOSE,” “clusterProfiler”, “org.Hs.eg.db”and “enrichplot,” were used to analyze and to generate the figures. Interaction between exoDEPs was analyzed using Cytoscape (version 3.8.2) [8]. The enrichment analysis was determined by a two-tailed Fisher's exact test to identify the enrichment of the differentially expressed proteins against the background of all identified proteins with a corrected P-value,0.05.
Western blot
Western blotting was performed as described previously [9]. Primary antibodies against CD9, CD63, CD81, HSP90, FGFBP1, SIPA1, THBS1, TGFBI, COL6A1, RPL10, SLC2A3 (GLUT3), MYO1D, RBP1, SMOC2, GLG1, and CEMIP (Protein-Tech Group, Rosemont, IL, USA) were used.
Western blotting was performed to verify the expression of exosome marker proteins and selected exoDEPs in SW620 and SW480 exosomes. Equivalent amounts of total protein (20 μg) were resolved by 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis, and the antibodies were diluted as follows: anti-CD9 (1:1000), anti-CD63 (1:1000), anti-CD81 (1:2000), anti-HSP90 (1:200), anti-FGFBP1 (1:500), anti-SIPA1 (1:2000), anti-THBS1 (1:500), anti-TGFBI (1:200), anti-COL6A1 (1:200), anti-RPL10 (1:1000), anti-GLUT3 (1:500), anti-MYO1D (1:200), anti-RBP1 (1:500), anti-SMOC2 (1:500), anti-GLG1 (1:1000), and anti-CEMIP (1:1000) (Protein-Tech Group, Rosemont, IL, USA). Proteins were detected using an enhanced chemiluminescence reagent (Santa Cruz Biotechnology, Dallas, TX, USA). Western blot signals were quantified using FluorChem E (Protein Simple, San Jose, CA, USA). All analyses were performed using western blots in at least two biological replicates.
Clinical sample collection and IHC
To identify the exoDEPs in clinical human CRC pathologic tissues, 33 CRC samples from 11 patients were retrospectively selected from the pathology database of the Affiliated First Hospital Dalian Medical University, China, between 2020 and 2021, for IHC analysis. All patients received routine preoperative preparation and surgical therapy. None of the patients had a family history of cancer nor received preoperative neoadjuvant therapy. There were five cases (45.45%) combined with clinical and pathological diagnosed regional lymph node metastasis (stage III/IV), and six cases (54.54%) without (Stage I/II). This retrospective study was approved by the First Affiliated Hospital of Dalian Medical University Medical Research and Ethics Committee. Written informed consent was obtained from all patients prior to tissue acquisitions. The clinical stage of the patients was defined according to the American Joint Committee on Cancer TNM staging system. Detailed patient clinical information is shown in Table 5.
Table 5.
Detailed clinical information from 11 patients.
Patient number | Gender | Age | CEA (ng/ml) | CA199 (ng/ml) | Tumor site | Tumor size (cm) | Tumor pattern | Pathological type | IHC | Metastasis (liver/lung/ ascites) | Lymph node metastasis | TNM stage | Stage |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Patient 1 | Female | 45 | 5.38 | 5.38 | Carcinoma of the straight B junction | 4.5 × 5 | Ulcerative | Moderately and poorly differentiated tubular adenocarcinoma | EGFR(Membranous plasma+); Ki-67(+30%);p53(Missense mutation);MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | 241(4/12) | pT4bN2M0 | IIIC |
Patient 2 | Female | 77 | 6.1 | 5.76 | Upper rectum | 6.5 × 4.3 | Ulcerative | Moderately and well differentiated tubular adenocarcinoma with papillary adenocarcinoma | EGFR(Membrane weak+); Ki-67(+70%); p53(Mutation); MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | no | pT4aN0M0 | IIB |
Patient 3 | Male | 63 | 3.07 | 2.5 | Sigmoid colon | 2.3 × 2.1 | Uplift | Moderately differentiated tubular adenocarcinoma | EGFR(Membrane +); Ki-67(+20%);p53(Nonsense mutation);MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | no | pT2N0M0 | Ⅰ |
Patient 4 | Female | 63 | 3.55 | 12.17 | Carcinoma of the straight B junction | 5.5 × 5 | Ulcerative | Moderately differentiated tubular adenocarcinoma | EGFR(Membrane weak+); Ki-67(+70%); p53(Mutation); MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | no | pT3N0M0 | IIA |
Patient 5 | Female | 65 | 1.41 | 9.06 | Ascending colon | 7.3 × 5 | Ulcerative | Moderately differentiated tubular adenocarcinoma with mucinous adenocarcinoma | EGFR (Membranous plasma+); Ki-67(+60–70%); p53(+30%); MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | 201(1c);202(1c) | pT3N1cM0 | IIIB |
Patient 6 | Male | 75 | 46.88 | 16.35 | Low rectum | 6 × 3.5 | Ulcerative | Moderately and poorly differentiated tubular adenocarcinoma | EGFR (most membranous plasma+); Ki-67(+80%); p53(+,mutation) MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | 251(13/16); 252(3/3) | pT4aN2M0 | IIIC |
Patient 7 | Female | 65 | 2.14 | 46.5 | Low rectum | 4.5 × 3.5 | Uplift | 粘液腺癌 (侵透肌层, 侵及肌层外纤维组织, 未见明确神经侵犯) | EGFR (+); Ki-67(+60%); p53(90%+, mutation); MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | 251(1/9 + 1c);252(1/6) | pT4aN2M0 | IIIC |
Patient 8 | Female | 69 | 9.74 | 0.74 | Hepatic flexure of colon | 5 × 3.5 | Ulcerative | Mucinous adenocarcinoma | EGFR (+); Ki-67(+70%); p53(95%+, mutation); MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | 221(1/11);223(1/2) | pT4aN0M0 | IIB |
Patient 9 | Male | 79 | 7.29 | 6.09 | Ascending colon | 4.5 × 3 | Ulcerative | Papillary adenocarcinoma with moderately differentiated tubular adenocarcinoma | EGFR (plasma+); Ki-67(+80%); p53(+,mutation)MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | 211(1/5) | pT4aN1M0 | IIIB |
Patient 10 | Female | 67 | 0.76 | 8.1 | Low rectum | 1.5 × 1.5 | Uplift | Papillary adenocarcinoma with well and moderately differentiated tubular adenocarcinoma | EGFR (Membrane weak+); Ki-67(+70%); p53(Missense mutation); MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | no | pT1N0M0 | Ⅰ |
Patient 11 | Female | 60 | 3.21 | 8.89 | Low rectum | 4 × 3 | Ulcerative | Poorly differentiated tubular adenocarcinoma | EGFR(Membranous plasma+); Ki-67(+80%); p53(weak or strong+70%); MLH-1(+); MSH-2(+); MSH-6(+); PMS-2(+) |
no | no | pT2N0M0 | Ⅰ |
The paraffin-embedded tissue sections were deparaffinized, dehydrated through graded alcohols, and subjected to antigen retrieval using citrate buffer (pH 6.0) for 7 min at high heat. The sections were washed with PBS and blocked with 0.3% hydrogen peroxide. The slides were incubated at 4 °C overnight with the following primary antibodies: anti-FGFBP1 (1:200), anti-SIPA1 (1:200), anti-THBS1 (1:200), anti-TGFBI (1:200), anti-COL6A1 (1:200), anti-RPL10 (1:200), anti-GLUT3 (1:200), anti-MYO1D (1:200), anti-RBP1 (1:200), anti-SMOC2 (1:200), anti-GLG1 (1:200), and anti-CEMIP (1:200) (Protein-Tech Group, Rosemont, IL, USA). The slides were visualized using a diaminobenzidine chromogen solution (Dako, Glostrup, Denmark) and counterstained with routine hematoxylin, followed by dehydration with graded ethanol series and mounting of the slides. The immunostaining densities of proteins were quantitatively assessed using NIS-Elements BR 3.0 (Nikon, Tokyo, Japan). In brief, after placing the sections on a microscope (Nikon E800), the images were transferred to a computer using a digital camera (Nikon 80i). Three visual fields were randomly inspected on all slides under high-power magnification. The mean optical densities of the positive areas were measured. The results are expressed as the exact values of the relative optical density units.
Cell proliferation assay
Cell proliferation assays were performed using a Cell Counting Kit‐8 (CCK‐8, KeyGEN BioTECH, Jiangsu, China) according to the manufacturer's protocol. Exosomes equaling 500 ng of exosomal proteins were added per well, if needed.
In vitro migration and invasion assay
Migration and invasion assays using the Transwell system (24 wells, 8 μm pore size filters, Costar, New York, USA) were performed as described in our previous studies [9]. Exosomes equaling 5 μg of exosomal proteins were added to the upper chamber together with the cells, if needed.
Statistical analysis
Statistical analyses and graphics were conducted using GraphPad Prism 9.0 (GraphPad Software, San Diego, CA, USA). Comparisons of quantitative data were made using Student's t-test (two-tailed; P < 0.05, considered statistically significant). Results are presented as mean ± standard error of the mean.
Results
Isolation and identification of exoDEPs secreted by SW620 and SW480 cell lines
To identify the exosomal proteins closely associated with CRC metastasis, we used a pair of homologous human CRC cell lines, SW620 (highly metastatic) and SW480 (weakly metastatic), as the model system. Under the light microscope, SW620 showed a spherical shape and discrete growth pattern, while SW480 showed a shuttle shape and aggregate growth pattern (Fig. 1A). After isolation, transmission electron microscopy (TEM) was used to visualize the sizes and structures of the prepared exosomes. As indicated in Fig. 1C, exosomes were detected as a heterogeneous population of small (< 150 nm) particles showing round and cup-shaped morphology electron micrographs. NTA confirmed the observed size distributions (Fig. 1D). Median sizes of 116.8 nm (mean 120.3 ± 6.64 nm) and 111.8 nm (mean 112.0 ± 5.60 nm) were observed for SW620-exo and SW480-exo, respectively. Particle concentrations were calculated resulting in 5.4 × 107/mL (mean 5.3 ± 1.1 × 107/mL) (SW620-exo) and 8.6 × 107/mL (mean 8.6 ± 1.7 × 107/mL) (SW480-exo). Expressions of exosomal markers CD9, CD63, CD81, and HSP90 were verified by WB (Fig. 1B). These vesicles were used for downstream proteomic analyses.
Fig. 1.
Identification of exosomes. (A) Cell morphology and distribution characteristics under Light microscope. (B) Western blot analysis confirmed exosome-specific and cell-specific protein marker expression. Protein sizes are indicated. (C) Transmission electron microscopy (TEM) pictures visualized the morphology of isolated exosomes. Scale bar: 100 nm. (D) Nanoparticle tracking analysis (NTA) revealed the size distribution and particle concentration of isolated exosomes.
Label-free proteomics identify the metastasis-associated exoDEPs in the CRC
After purifying the exosomes of SW620 and SW480, we first lysed the exosomes to obtain the proteins and further hydrolyzed the protein peptides, which were directly semi-quantitatively detected by liquid chromatography-mass spectrometry (LC-MS). According to the principle of label-free proteomics, the frequency (counts) of peptide capture and detection in mass spectrometry is positively correlated with its abundance in the mixture. Therefore, the protein count detected by mass spectrometry reflects the abundance of protein. Using an appropriate mathematical formula, the mass spectrometry detection count can be linked with the amount of protein to quantify the secreted proteins of the two cells to determine the upregulated or downregulated proteins with the largest expression difference and the “all or none” expressed proteins. The technical schematic diagram and the heat map and volcano map of the differential proteomics are shown in Fig. 2.
Fig. 2.
. Label-free proteomics was used to identify the metastasis-associated exoDEPs in SW620 and SW480 cells. (A) The technical schematic diagram of label-free proteomics. (B) The heat map shows the clustering results of significant differentially expressed proteins and whether there are differentially expressed in SW620 vs. SW480 comparison group. (C) Volcano plot shows non-regulated proteins (blue) and significantly up (red) and downregulated (green) proteins in SW620 exosomes. The X axis represents log2-transformed fold change values. Y axis shows the −log10 p-value adjusted for multiple comparisons.
In total, 942 exoDEPs were quantified in all three mechanical and biological repetitions and searched against the database. By setting the quantification ratio 2.0-fold change as the threshold, 115 exoDEPs (2.0-fold change, P < 0.05) were identified, including 31 upregulated and 84 downregulated proteins in SW620 exosomes. The top 10 upregulated and downregulated exosomal proteins in SW620 are listed in Tables 1 and 2, respectively. (Table S1 lists all 31 upregulated proteins and Table S2 lists all 84 downregulated proteins in SW620 exosomes).
Table 1.
Top 10 upregulated exoDEPs in SW620.
Protein IDs | Protein names | Gene names | Sequence coverage [%] | Mol. weight [kDa] | SW620/SW480 | P. value |
---|---|---|---|---|---|---|
Q14512 | Fibroblast growth factor-binding protein 1 | FGFBP1 | 19.2 | 26.264 | 35.5622503 | 0.03006523 |
P24821 | Tenascin | TNC | 31.2 | 240.85 | 31.9171121 | 0.02235115 |
Q96FS4 | Signal-induced proliferation-associated protein 1 | SIPA1 | 0.7 | 101.82 | 13.2653467 | 0.00628316 |
Q8N6F7 | Galanin peptides | GAL | 25.2 | 13.302 | 6.59087605 | 0.00043934 |
P07996 | Thrombospondin-1 | THBS1 | 25.7 | 129.38 | 5.15722985 | 0.00596742 |
Q15582 | Transforming growth factor-beta-induced protein ig-h3 | TGFBI | 26.4 | 74.68 | 4.29955673 | 0.00079523 |
Q08380 | Galectin-3-binding protein | LGALS3BP | 30.4 | 65.33 | 4.23649295 | 0.02398548 |
P18827 | Syndecan-1 | SDC1 | 10.6 | 16.936 | 3.95082411 | 0.00969672 |
P98160 | Basement membrane-specific heparan sulfate proteoglycan core protein | HSPG2 | 31.7 | 464.01 | 3.65451637 | 0.00490496 |
P50995 | Annexin A11 | ANXA11 | 39.7 | 45.597 | 3.63266542 | 0.00082676 |
Table 2.
Top 10 downregulated exoDEPs in SW620.
Protein IDs | Protein names | Gene names | Sequence coverage [%] | Mol. weight [kDa] | SW620/SW480 | P. value |
---|---|---|---|---|---|---|
P11169 | Solute carrier family 2, facilitated glucose transporter member 3 | SLC2A3 | 20.8 | 31.874 | 0.011749168 | 0.0296785 |
O94832 | Unconventional myosin-Id | MYO1D | 28.2 | 116.2 | 0.038787995 | 0.00027188 |
P29374 | Retinol-binding protein 1 | RBP1 | 56 | 10.644 | 0.072787169 | 0.00388128 |
P98172 | Ephrin-B1 | EFNB1 | 16.2 | 38.006 | 0.074930021 | 0.00119442 |
P08133 | Annexin A6 | ANXA6 | 37.1 | 75.872 | 0.078012029 | 0.00031708 |
Q96TA1 | Niban-like protein 1 | C9orf88 | 10.9 | 82.682 | 0.126490412 | 0.01065253 |
Q9Y696 | Chloride intracellular channel protein 4 | CLIC4 | 26.6 | 26.694 | 0.137963444 | 0.03000887 |
P05556 | Integrin beta-1 | ITGB1 | 20.9 | 88.414 | 0.14864844 | 0.00146909 |
P16422 | Epithelial cell adhesion molecule | EPCAM | 34.8 | 37.893 | 0.163589346 | 0.00048008 |
P61081 | NEDD8-conjugating enzyme Ubc12 | UBE2M | 8.7 | 20.9 | 0.165887295 | 0.0115204 |
To find more accurate and in-depth information associated with metastasis of CRC, we also identified 90 “all or none” exoDEPs in this analysis, wherein 30 proteins were found only in SW620 exosomes, and 60 proteins were found only in SW480 exosomes. The top five proteins expressed only in SW620 and SW480 exosomes are listed in Tables 3 and 4, respectively. (Table S3 lists all 30 proteins expressed only in SW620 exosomes, and Table S4 lists all 60 proteins expressed only in SW480 exosomes).
Table 3.
Top 10 exoDEPs only expressed in SW620.
protein IDs | Protein names | Gene names | Sequence coverage [%] | Mol. weight [kDa] | Sequence coverage 480-mean [%] | Sequence coverage 620-mean [%] | Intensity | LFQ intensity 480-mean | LFQ intensity 620-mean |
---|---|---|---|---|---|---|---|---|---|
Q8NCR9 | Clarin-3 | CLRN3 | 12.4 | 25.321 | 3.866667 | 6.6 | 79,843,000 | 0 | 34,011,666.7 |
Q8NDC0 | MAPK-interacting and spindle-stabilizing protein-like | MAPK1IP1L | 6.9 | 24.269 | 0 | 4.6 | 58,364,000 | 0 | 30,015,333.3 |
Q8WUJ3 | Cell migration-inducing and hyaluronan-binding protein | CEMIP | 11.2 | 153 | 0 | 7.2 | 5.88E+08 | 0 | 283,110,000 |
Q92896 | Golgi apparatus protein 1 | GLG1 | 3.4 | 134.55 | 0 | 2.766667 | 1.37E+08 | 0 | 68,899,333.3 |
Q9H3U7 | SPARC-related modular calcium-binding protein 2 | SMOC2 | 17.7 | 49.674 | 0 | 16.86667 | 8.83E+08 | 0 | 448,096,667 |
Table 4.
Top 10 exoDEPs only expressed in SW480.
Protein IDs | Protein names | Gene names | Sequence coverage [%] | Mol. weight [kDa] | Sequence coverage 480-mean [%] | Sequence coverage 620-mean [%] | Intensity | LFQ intensity 480-mean | LFQ intensity 620-mean |
---|---|---|---|---|---|---|---|---|---|
P05121 | Plasminogen activator inhibitor 1 | SERPINE1 | 2.5 | 45.059 | 1.666666667 | 0 | 36,502,000 | 11,615,333.33 | 0 |
P50151 | Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-10 | GNG10 | 42.6 | 7.2053 | 42.6 | 0 | 98,311,000 | 29,713,666.67 | 0 |
P41221 | Protein Wnt-5a | WNT5A | 9 | 40.886 | 7 | 0 | 213,720,000 | 62,549,666.67 | 0 |
P31146 | Coronin-1A | CORO1A | 3.9 | 51.026 | 1.866666667 | 0 | 25,254,000 | 8,137,000 | 0 |
Q92817 | Envoplakin | EVPL | 0.4 | 231.63 | 0.4 | 0.133333333 | 22,870,000 | 6,611,900 | 0 |
Exosomes from SW620 promoted the proliferation, migration, and invasion of SW480
To investigate the role of exosomes in the proliferation of CRC cell lines, we performed CCK-8 assays. The results showed that after treatment with SW620 exosomes, the proliferation of SW480 cells increased. After treatment with SW480 exosomes, the proliferation of SW620 cells decreased more obviously (Fig. 3A). This result indicated that SW620 exosomes could promote the proliferation of SW480 cells, whereas SW480 exosomes could inhibit the proliferation of SW620 cells, thus showing that the inhibitory role of SW480 exosomes was stronger. To investigate the role of exosomes in the migration and invasion of CRC cell lines, we performed transwell assays. Results showed that after treatment with SW620 exosomes, migration and invasion of SW480 were significantly enhanced by 60% (Fig. 3B-C). These results indicated that exosomes play a crucial role in CRC invasive behaviors through extracellular effects, and the promoting role of SW620 exosomes on SW480 cells was very prominent. The changes in cell shape and pseudopodia of this pair of cell lines were not obvious after treatment.
Fig. 3.
. The changes in the migration and invasion ability of CRC cells after treated with SW620-exo and SW480-exo. (A) SW620-exo enhanced proliferation of SW480 cells and SW480-exo inhibited proliferation of SW620 cells by CCK-8 analysis. (B) SW620-exo enhanced migration of SW480 cells by Transwell invasion assay. (C) SW620-exo enhanced invasion of SW480 cells by Transwell invasion assay. (P < 0.05, Student's t-test) (*p, 0.05).
In summary, the promoting role of the exosomal proteins of SW620 and the inhibiting role of the exosomal proteins of SW480 on tumor invasion and metastasis have been elucidated in the cellular functional experiments above. However, the key proteins and pathways involved remained unknown. Therefore, we performed bioinformatics analysis for all exoDEPs to explore in-depth information in further research.
Functional enrichment analysis and protein-protein interaction network analysis of upregulated and downregulated exoDEPs
To characterize the functions and subcellular locations of the upregulated and downregulated exoDEPs between this pair of cell lines, GO annotation, KEGG pathway, and PPI network analysis were carried out. The GO annotation analysis included biological processes, cellular components, and molecular functions. For biological process classification, exoDEPs are mainly associated with cell junction assembly, cell junction organization, extracellular structure organization, regulation of cellular component size, and viral life cycle. For cellular components, exoDEPs are mainly associated with cell-substrate adherens junctions, cell-substrate junctions, focal adhesions, melanosomes, and pigment granules. For molecular function, exoDEPs are mainly associated with actin binding, actin filament binding, cadherin binding, calcium-dependent protein binding, and cell adhesion molecule binding (Fig. 4A-C).
Fig. 4.
Bioinformatics analysis of upregulated and downregulated exoDEPs. (A) GO annotation includes Biological Processes (BP), Cellular component (CC) and Molecular Function (MF) analysis. Coloring of proteins is based on functional enrichment analysis; (B) The enriched pathways by KEGG analysis; (C) Visible clusters of the protein-protein interaction map was assigned to enriched ontologies among the upregulated and downregulated exoDEPs with calculated P-value adjusted for multiple comparisons.
In the KEGG pathway analysis, the enriched pathways included the PI3K-Akt signaling pathway and ECM-receptor interaction in metabolism, endocytosis, regulation of actin cytoskeleton, focal adhesion, and cell adhesion molecules in cellular processes (Fig. 4D). The enriched pathways suggest that the exosomal proteins were mostly active in the process of tumor metabolism, cellular movement, and skeleton connection in the microenvironment.
To identify the chief nodes and important connectors among the exoDEPs, we performed the PPI network analysis using Cytoscape 3.8.2. There were 114 exoDEPs that had at least one known interaction with other proteins in the PPI network (P < 1.0e-16). The top 20 active proteins with three or more interactions were PS1TP5BP1, GAPDH, ACTG1, FN1, ITGB1, RAC1, ACTN4, TSG101, ACTN1, TFRC, ITGA2, ANXA2, THBS1, SDC1, LDHA, APOE, P4HB, HSPG2, MYO1C, and ARPC2. These proteins were mostly associated with pathways including endocytosis, regulation of actin cytoskeleton, Rap1 signaling pathway, cAMP signaling pathway, and PI3K-Akt signaling pathway. The direct interactions between these highly active proteins and enriched pathways are also shown in Fig. 4E.
Bioinformatics analysis of “all or none” exoDEPs
For the “all or none” exoDEPs, GO annotation, KEGG pathway, and PPI network analysis were carried out separately. For biological process classification, exoDEPs expressed only in SW620 are mainly associated with the regulation of synapse structure or activity, synapse organization, response to amyloid-beta, positive regulation of intracellular protein transport, and positive regulation of cellular protein localization. ExoDEPs expressed only in SW480 cells are mainly associated with cell-substrate adhesion, cell-matrix adhesion, positive regulation of interleukin-8 production, negative regulation of the canonical Wnt-signaling pathway, and regulation of hair follicle development. For cellular components, exoDEPs expressed only in SW620 were mainly associated with glutamatergic synapse, endoplasmic reticulum lumen, clathrin-coated pit, interstitial matrix, and glial cell projection. ExoDEPs expressed only in SW480 are mainly associated with focal adhesion, cell-substrate adherens junction, cell-substrate junction, collagen-containing extracellular matrix, and proteasome regulatory particles. For molecular function, exoDEPs expressed only in SW620 were mainly associated with amide binding, clathrin binding, glycosaminoglycan binding, peptide binding, and signal sequence binding, while exoDEPs expressed only in SW480 were mainly associated with actin monomer binding, cadherin binding, cell adhesion mediator activity, cell adhesion molecule binding, and proteasome binding (Fig. 5A).
Fig. 5.
Bioinformatics analysis of “all or none” exoDEPs. (A) GO annotation includes Biological Processes (BP), Cellular component (CC) and Molecular Function (MF) analysis. Coloring of proteins is based on functional enrichment analysis; (B) The enriched pathways by KEGG analysis; (C) Visible clusters of the protein-protein interaction map was assigned to enriched ontologies among the “all or none” exoDEPs with calculated P-value adjusted for multiple comparisons.
For the KEGG pathway analysis, exoDEPs expressed only in SW620 were mostly active in pathways including the adherens junction, endocrine and other factor-regulated calcium reabsorption, endocytosis, focal adhesion, and phospholipase D signaling pathway. ExoDEPs expressed only in SW480 were mostly active in pathways including Epstein-Barr virus infection, citrate cycle (TCA cycle), Huntington disease, proteasome, and Alzheimer disease (Fig. 5B).
In the PPI network analysis, 88 exoDEPs had at least one known interaction with other proteins in the PPI network (P = 5.08e-07) (Fig. 5C). The top 20 active proteins with three or more interactions were CDH1, APP, CYCS, WNT5A, PSMA2, FYN, ICAM1, IDH1, PPP2CB, PSMD14, ITGA1, PSMD2, DNM1, ENO2, MCAM, CS, AP2B1, SERPINE1, QSOX1, and TJP1. These proteins were mostly associated with pathways including 2-oxocarboxylic acid metabolism, citrate cycle (TCA cycle), proteasome, synaptic vesicle cycle, endocrine and other factor-regulated calcium reabsorption, pathogenic Escherichia coli infection, viral myocarditis, cell adhesion molecules (CAMs), phospholipase D signaling pathway, carbon metabolism, apelin signaling pathway, and Epstein-Barr virus infection.
In summary, metabolism and cell movement regulation are the main mechanisms involved in “all or none” exoDEPs. Compared to the exosomes of SW620, the proteins that disappeared in the exosomes of SW480 may play a role and may degrade or they may be transmitted as messengers. These proteins play an important role in the invasion and metastasis of CRC.
Validation of exoDEPs by WB on CRC cell line exosomes
To obtain independent evidence for exoDEPs identified by label-free proteomics, we chose six upregulated proteins, three downregulated proteins, and three proteins only expressed in SW620 for verification by WB. The results showed that six upregulated proteins, FGFBP1, SIPA1, THBS1, TGFBI, COL6A1, and RPL10 were overexpressed in the SW620 exosomes (Fig. 6A) and three downregulated proteins, GLUT3, MYO1D, and RBP1, were overexpressed in SW480 exosomes (Fig. 6B). Three proteins expressed in SW620 exosomes, namely SMOC2, GLG1, and CEMIP, were verified as overexpressed in the SW620 exosomes (Fig. 6C). However, the differences in expression identified by WB were not coordinated with mass spectrometry. In the verification of the upregulated proteins, the expression differences of SIPA1 and TGFBI were the most obvious and not FGFBP1. The differences in the expression of THBS1 and COL6A1 were not obvious as in mass spectrometry. In the verification of SMOC2, GLG1, and CEMIP that were expressed only in SW620, both SMOC2 and GLG1 showed little expression in SW480 exosomes, while the expression of CEMIP in SW480 exosomes could still be seen.
Fig. 6.
Validation of exoDEPs by WB on CRC cell line exosomes. (A) Six upregulated proteins FGFBP1, SIPA1, THBS1, TGFBI, COL6A1, and RPL10 were verified overexpressed in SW620 exosomes. (B) Three downregulated proteins GLUT3, MYO1D, and RBP1 were verified to be overexpressed in SW480 exosomes. (C) Three proteins SMOC2, GLG1 and CEMIP were verified to be obviously overexpressed in SW620 exosomes. (P, 0.05, Student's t-test) (*p < 0.05, **p < 0.01).
Validation of exoDEPs by IHC on clinical human CRC pathologic tissues
In this study, we chose six upregulated exoDEPs, FGFBP1, SIPA1, THBS1, TGFBI, COL6A1, and RPL10; three downregulated exoDEPs, GLUT3, MYO1D, and RBP1 in SW620; and three exoDEPs, SMOC2, GLG1, and CEMIP, that were expressed only in SW620 for verification by IHC staining of compared non-lymph node metastatic (n = 6) and lymph node metastatic CRC tissues (n = 5) (Fig. 7).
Fig. 7.
Validation of exoDEPs by IHC on clinical human CRC pathologic tissues. (A) Six upregulated proteins FGFBP1, SIPA1, THBS1, TGFBI, COL6A1, and RPL10 were found to be overexpressed in CRC tissues with lymph node metastasis. (B) Three downregulated proteins GLUT3, MYO1D, and RBP1 were found to be overexpressed in CRC tissues without lymph node metastasis. (C) Three proteins SMOC2, GLG1, and CEMIP were verified obviously overexpressed in CRC tissues with lymph node metastasis. (P,0.05, Student's t-test) (*p < 0.05, **p < 0.01).
The results indicated that six upregulated exoDEPs, namely FGFBP1, SIPA1, THBS1, TGFBI, COL6A1, and RPL10, were overexpressed in CRC tissues with lymph node metastasis, and three downregulated exoDEPs, namely GLUT3, MYO1D, and RBP1, were overexpressed in CRC tissues without lymph node metastasis. Three exoDEPs expressed only in SW620, namely SMOC2, GLG1 and CEMIP, were found to be overexpressed in CRC tissues with lymph node metastasis. Most proteins are expressed in tissues between tumors rather than in cells. Compared with the expression in cancer tissues, the expression levels of all identified exoDEPs in paracancerous tissues were lower. The immunostaining densities of proteins were quantitatively assessed using NIS-Elements BR 3.0 (Nikon, Tokyo, Japan). In brief, after placing the sections on a microscope (Nikon E800), the images were transferred to a computer using a digital camera (Nikon 80i). Three visual fields were randomly examined on all slides under high-power magnification. The mean optical densities of the positive areas were measured. The results were expressed as the exact values of the relative optical density units. This analysis confirmed that all three proteins were significantly upregulated in both non-metastatic and metastatic CRC tissues.
Discussion
Exosomes are nanovesicles that play a major role in the TME, especially in cell-cell communication by horizontal transfer of signaling molecules. TME-derived exosomes also support metastatic niche formation, suppress the antitumor immune response, and impart drug resistance. Our proteomic analysis aimed to reveal the exosomal proteins that affect CRC invasion and metastasis. In this study, we used a label-free proteomics method to perform comparative proteomic analysis of exosomes from a pair of homologous human CRC cell lines SW620 (highly metastatic) and SW480 (weakly metastatic), which have different metastatic abilities. A total of 942 exoDEPs were quantified, among which 115 exoDEPs (31 upregulated and 84 downregulated in SW620 exosomes) showed obvious differential expression (2.0-fold change, P < 0.05). The total 90 “all or none” exoDEPs were identified in this analysis, wherein 30 proteins were found expressed only in SW620 exosomes, and 60 proteins were found expressed only in SW480 exosomes.
To verify the data obtained in this research, we chose six upregulated exoDEPs in SW620, including FGFBP1, SIPA1, THBS1, TGFBI, COL6A1, and RPL10 and subjected them to WB and IHC. All six proteins were upregulated in SW620 exosomes and CRC tissues with metastasis. The expression of the six proteins correlated with poor pathological type and late stage.
Fibroblast growth factor-binding protein 1 (FGFBP1) was the first upregulated exoDEP in SW620 cells. It is a secreted chaperone that mobilizes paracrine-acting FGFs, stored in the extracellular matrix, and presents them to their cognate receptors. Several studies have demonstrated FGFBP1 overexpression in various tumors, including head and neck squamous cell carcinoma (HNSCC), mammary, cervix, prostate, melanoma, colon, and pancreatic cancer [10], [11], [12], [13]. In previous studies, the role of FGFBP1 was mostly related to cancer cell proliferation and angiogenesis. Recent studies have indicated the promoting metastatic role of FGFBP1. HSD11B2 was reported to enhance CRC cell migration and invasion capacity by upregulating the expression by FGFBP1, increasing the phosphorylation of AKT in the PI3K-Akt pathway [14]. FGFBP1 also acts as a downstream target of the FBW7/c-Myc axis and promotes cell proliferation and migration in pancreatic cancer [15]. There are no reports on the exosomal role of FGFBP1 in tumor progression. In this study, we verified the upregulation of exo-FGFBP1 in a highly metastatic CRC cell line. The results confirmed the positive exosomal role of FGFBP1 in the invasion and metastasis of CRC cells from the TME. However, the understanding of the specific mechanism requires further study.
Signal-induced proliferation associated protein 1 (SIPA1) is the third most upregulated exoDEP in SW620 cells. It is a GTPase-activating protein expressed in proliferative active lymphoid cells as a mitogen-induced nuclear protein. SIPA1 has a tight interaction in controlling cellular adhesion in cancer metastasis [16]. Overexpression of SIPA1 was found in colon, prostate, breast, and other several types of cancers [17,18]. SIPA1 promotes cancer metastasis by inhibiting adhesion and downregulating the expression of ECM-related proteins [19]. The findings of this study verified the metastasis-promoting role of SIPA1 as an exosomal protein in the CRC microenvironment. IHC results indicated that SIPA1 was mostly expressed in the tissue around or between cancer cells. Combined with other research results, exo-SIPA1 may be secreted to decrease intercellular adhesion in the CRC.
Thrombospondin-1 (THBS1) is the fifth most upregulated exoDEP in SW620 cells. As a glycoprotein, it is an endogenous inhibitor of angiogenesis and tumor progression. Downregulation of THBS1 is reported to be correlated with poor prognosis in lung, breast, and cervical cancer [20], [21], [22]. For THBS1 and CRC, a recent study indicated that low THBS1 expression correlated with late stage of liver and lymph node metastasis and significantly worse overall survival rate than high THBS1 expression [23]. Although most results indicated the negative role of THBS1 in tumor progression, we verified the upregulation of THBS1 in highly metastatic CRC cell line exosomes. This result appears to be more complicated. The role of THBS1 as an exosome in the extracellular microenvironment is contrary to its role in the cell. In previous studies, the positive role of exo-THBS1 in CRC invasion and metastasis has rarely been confirmed. This special finding may provide new research directions for THBS1 in the CRC microenvironment.
Transforming growth factor β‐induced protein (TGFBI) is the sixth most upregulated exoDEP in SW620 cells. It is a secreted extracellular matrix protein consisting of 683 amino acids and includes four evolutionarily conserved fasciclin‐1 domains and a C-terminal Arg‐Gly‐Asp motif [24]. The role of TGFBI in cancer is unclear. It appears to be tissue-specific, acting as a tumor suppressor or promoter. Downregulation of TGFBI was observed in leukemia [25], whereas hypermethylation of the TGFBI promoter, which suppresses TGFBI expression, was observed in ovarian, prostate, and lung carcinomas [26,27]. These findings indicated the tumor-suppressor functions of TGFBI. Conversely, TGFBI is also known to perform tumor promoter functions in various cancers. TGFBI upregulation has been reported as a promising prognostic marker in oral squamous cell carcinoma, renal cell carcinoma, and pancreatic cancer [28]. In this study, we verified the promoting role of TGFBI in the invasion and metastasis of CRC as an exosome, which has not been previously reported. The exosomal role of TGFBI is very promising and requires further study.
Collagen type VI α1 chain (COL6A1) is another upregulated exoDEP and has an anchoring function that plays roles in cell migration, differentiation, and embryonic development [29]. Compared to the corresponding normal tissues, COL6A1 is more active in tumor tissues such as cervical cancer, prostate cancer, and lung cancer [30,31]. Overexpression of COL6A1 enhances the motility and metastasis of cancer cells [32]. Although the role of COL6A1 has been elucidated, its role as an exosome has seldom been determined.
Ribosomal protein L10 (RPL10), also known as UL16, is a 25 kDa protein, which was first found in non-tumorigenic Wilms tumors, and participates in the late steps of 60S ribosomal assembly [33]. RPL10 mainly plays a role in protein synthesis. However, an increasing number of studies have shown that RPL10 also has ribose functions in vitro, such as participating in signaling pathways in different cell biological processes, regulating cell proliferation, migration, and differentiation. In terms of tumor, some studies have confirmed that the expression level of RPL10 in human epithelial ovarian cancer is higher than that in normal ovarian tissue. When its expression level decreases, the viability, migration, and invasion ability of epithelial ovarian cancer cells decrease, but the apoptosis level increases [34]. Similarly, in a study of prostate cancer, high levels of RPL10 expression can promote the progression of advanced tumors [35,36]. Tumor immunity has been a promising research direction in recent years. Proteomic studies have shown that RPL10 can participate in the JAK/STAT signaling pathway and promote the progression of T-lymphocytic leukemia [37]. Our research also confirmed the promoting role of exo-RPL10 in CRC metastasis. Based on the above results, we speculate that RPL10 may have a function beyond the ribosome, such as immunosuppression in the process of tumor development.
The top three downregulated exoDEPs in SW620, including SLC2A3, MYO1D, and RBP1, were also verified. They were upregulated in SW620 exosomes and CRC tissues without metastasis. The overexpression of the three proteins demonstrated a correlation with the pathological type and early stage.
Solute carrier family 2 member 3 (SLC2A3), also known as GLUT3, facilitates glucose transport. In previous research, tumors with high expression of SLC2A3 were shown to enrich immune cell infiltration in the TME. An association between the overexpression of SLC2A3 and poor clinical outcomes has been reported in CRC [38,39]. In Yao's research, SLC2A3 accelerated aerobic glycolysis in gastric cancer cells by activating the SLC2A3-STAT3-SLC2A3 feedback loop and then promoting phosphorylation of the STAT3 signaling pathway and downstream glycolytic targeting gene. SLC2A3 also potentially contributes to the M2 subtype transition of macrophage infiltration in the gastric cancer microenvironment [40]. However, in our study, exo-SLC2A3 was found to inhibit the malignant behavior of CRC metastasis. The opposite role of intracellular and extracellular is a valuable part of this study and warrants further research.
Unconventional myosin-1d (MYO1D) belongs to the EGFR family (except ErbB3) at the plasma membrane. It binds only with unphosphorylated EGFRs and anchors them to the underlying actin cytoskeleton at the plasma membrane. Overexpression of MYO1D enhances colorectal and breast cancer cell motility and viability by upregulating EGFR levels, thereby promoting colorectal tumor progression in vivo in mice. MYO1D is upregulated in human CRC tissues from the advanced stages. Overexpressed MYO1D contributes to CRC, possibly as a novel oncogene, thus serving as an additional target for the suppression of RTK signaling. However, in this study, exo-MYO1D in the microenvironment might play an inhibitory role in the malignant behavior of CRC. The detailed mechanism requires further verification and study.
Retinol-binding protein 1 (RBP1) is a cytosolic carrier that regulates retinol homeostasis in various human and rodent tissues [41,42]. It possesses high-affinity binding of retinoic acid and possibly functions as a chaperone-like protein to regulate the prenuclear phase of retinoic acid signaling [43,44]. Studies have reported that RBP1 is abnormally expressed in several human cancers, including bladder cancer, laryngeal cancer, and tongue squamous cell carcinoma [45], [46], [47]. RBP1 overexpression promotes cell autophagy in OSCC cells by interacting with CKAP4 [48]. However, in this study, exo-RBP1 in the microenvironment might play an inhibitory role in the malignant behavior of CRC. The detailed mechanism requires further verification and study.
In our research, the discovery of the “all or none” exoDEPs was of great significance. The exoDEPs expressed only in SW620 cells can more clearly show their ability to promote the invasion and metastasis of CRC cells. We screened out ninety “all or none” exoDEPs, including 30 ones only expressed in SW620 and 60 ones only expressed in SW480. Among these exoDEPs, we performed WB and IHC experiments using SMOC2, GLG1, and CEMIP to verify the results of proteomics. Overexpression of the three proteins demonstrated a correlation with the late pathological stage.
Secreted modular calcium-binding protein2 (SMOC2) belongs to the secreted protein acidic and rich in cysteine (SPARC) family of matricellular proteins. It regulates the expression of ECM and Matrix metalloproteinases (MMPs) and modulates cell-matrix interactions, focal adhesion, and actin stress fiber organization by activating cellular integrins [49]. The expression and functional significance of SMOC2 have been explored in many types of cancers, such as breast, colon, lung, and liver cancers [49], [50], [51], [52]. As an intestinal stem cell marker, SMOC2 elevation is necessary to increase cell motility, proliferation, and liver metastasis in colon cancer. However, in a recent study by Jang, SMOC2 was suggested as an independent prognostic marker for better clinical outcomes in a large cohort of CRC patients and acts as a tumor suppressor in CRC progression [53]. In this study, we confirmed that SMOC2 promotes the invasion and metastasis of CRC as an exosome and from the extracellular perspective. Therefore, combined with these controversial results, the role of exo-SMOC2 is largely unknown and needs to be further explored.
Golgi glycoprotein 1 (GLG1, also called E-selectin Ligand 1, Esl1) is a cysteine-rich fibroblast growth factor receptor (FGFR). GLG1 has been reported to be associated with the progression of various carcinomas. FGFR signaling possesses broad mitogenic and cell survival mechanisms and is involved in a variety of biological processes, including embryonic development, cell growth, and tumor invasion [54]. In a study by Tamami Morisaki, GLG1 was found to be significantly associated with the T stage with respect to invasion depth [55]. It should be noted that GLG1 may be associated with the invasion potential of CSCs via FGFR signaling.
CEMIP, also known as KIAA1199 (cell migration-inducing protein), is a secreted protein that plays a role in extracellular ligand binding and processing. The biological role of CEMIP has been studied in cancer biology, and a number of studies have demonstrated its high expression levels in cancer cell lines and its association with cancer invasion and metastasis [56,57]. It plays a crucial role in the apoptosis, proliferation, invasion, and migration of various cancer cells. In addition, CEMIP is also involved in the regulation of various signaling pathways such as epithelial-mesenchymal transition (EMT), PI3K/Akt, MEK/ERK, and Wnt/β-catenin. In CRC research, CEMIP was found to be significantly overexpressed in the adenoma-adenocarcinoma pathway of colorectal carcinogenesis [58]. CEMIP mRNA was also detected in the plasma of patients with colorectal neoplasia than in the plasma from healthy controls [59]. These results suggest that CEMIP is both an endogenous and a secreted protein in the CRC. However, its role in promoting cancer cell metastasis as an exosome has not been extensively studied.
Reviewing the whole study, we think that there are some limitations that need to be improved. First, the clinical sample volume is limited, and a larger clinical sample volume collected is needed in the future study to establish the reference of clinical features. Second, the validated exoDEPs may play an important regulatory role in the progress of CRC, therefore, elucidation of deeper mechanism research is needed in future study.
In conclusion, in this study, we first found that SW620‐derived exosomes could significantly enhance the proliferation, migration and invasion abilities of SW480 cells. These results indicated that highly invasive colorectal cancer cells may transport their oncogenic characteristics to less invasive cancer cells through the exosomes to accelerate disease progression. We then used a label-free proteomics as a large-scale purification method to compare this pair of homologous CRC cell lines with different metastatic abilities to reveal the most associated exosomal proteins regulating CRC metastasis. Finally, six upregulated exoDEPs (FGFBP1, SIPA1, THBS1, TGFBI, COL6A1, and RPL10), three downregulated exoDEPs (SLC2A3 [GLUT3], MYO1D, and RBP1), and three exoDEPs (SMOC2, GLG1, and CEMIP) expressed only in SW620 were validated by western blot (WB) and immunohistochemistry (IHC). Verification of these exosomal proteins revealed their potential biochemical characteristics and regulatory functions in CRC invasion and metastasis.
Author contributions
Xinlu Liu and Ge Liu; Methodology, Xinlu Liu and Na Li; Software, Xinlu Liu; Validation, Xinlu Liu, Na Li, Chi Zhang and Xiaoyu Wu; Formal Analysis, Xinlu Liu and Na Li; Data Curation, Xinlu Liu, Na Li, Chi Zhang, Xiaoyu Wu, Gang Dong and Shoujia Zhang; Writing – Original Draft Preparation, Xinlu Liu; Writing – Review & Editing, Xinlu Liu, Na Li and Chi Zhang; Visualization, Xinlu Liu; Supervision, Xinlu Liu and Ge Liu.
Funding
There is no funding for this research.
Supplementary Tables
Table S1
Protein IDs | Protein names | Gene names | Sequence coverage [%] | Mol. weight [kDa] | SW620/SW480 | P. value |
---|---|---|---|---|---|---|
Q14512 | Fibroblast growth factor-binding protein 1 | FGFBP1 | 19.2 | 26.264 | 35.5622503 | 0.03006523 |
P24821 | Tenascin | TNC | 31.2 | 240.85 | 31.9171121 | 0.02235115 |
Q96FS4 | Signal-induced proliferation-associated protein 1 | SIPA1 | 0.7 | 101.82 | 13.2653467 | 0.00628316 |
Q8N6F7 | Galanin peptides | GAL | 25.2 | 13.302 | 6.59087605 | 0.00043934 |
P07996 | Thrombospondin-1 | THBS1 | 25.7 | 129.38 | 5.15722985 | 0.00596742 |
Q15582 | Transforming growth factor-beta-induced protein ig-h3 | TGFBI | 26.4 | 74.68 | 4.29955673 | 0.00079523 |
Q08380 | Galectin-3-binding protein | LGALS3BP | 30.4 | 65.33 | 4.23649295 | 0.02398548 |
P18827 | Syndecan-1 | SDC1 | 10.6 | 16.936 | 3.95082411 | 0.00969672 |
P98160 | Basement membrane-specific heparan sulfate proteoglycan core protein | HSPG2 | 31.7 | 464.01 | 3.65451637 | 0.00490496 |
P50995 | Annexin A11 | ANXA11 | 39.7 | 45.597 | 3.63266542 | 0.00082676 |
P46782 | 40S ribosomal protein S5 | RPS5 | 16 | 22.391 | 3.43144522 | 0.0340731 |
P67809 | Y-box-binding protein | YBX1 | 16.5 | 29.374 | 3.11206098 | 0.03244381 |
P02751 | Fibronectin | FN1 | 27.8 | 259.21 | 3.06819854 | 0.03725025 |
P12109 | Collagen alpha-1(VI) chain | COL6A1 | 3.8 | 108.34 | 3.06807273 | 0.00024771 |
Q9UN37 | Vacuolar protein sorting-associated protein 4A | VPS4A | 16.7 | 26.829 | 2.70042167 | 0.00500222 |
Q9P265 | Disco-interacting protein 2 homolog B | DIP2B | 7.1 | 171.49 | 2.68856637 | 0.0007878 |
P46779 | 60S ribosomal protein L28 | RPL28 | 21.3 | 9.657 | 2.64353933 | 0.03593148 |
Q5VW32 | BRO1 domain-containing protein BROX | BROX | 11.1 | 42.872 | 2.43025282 | 0.00954632 |
Q99816 | Tumor susceptibility gene 101 protein | TSG101 | 15.9 | 40.917 | 2.40154634 | 0.00648237 |
O43657 | Tetraspanin-6 | TSPAN6 | 25.6 | 14.958 | 2.33242004 | 0.00169953 |
P05452 | Tetranectin | CLEC3B | 27.5 | 17.794 | 2.26469372 | 0.011433 |
P61313 | Ribosomal protein L15 | RPL15 | 15.2 | 19.338 | 2.25663371 | 0.00551006 |
P53990 | IST1 homolog | IST1 | 57.3 | 23.375 | 2.25109795 | 0.00531066 |
P13010 | X-ray repair cross-complementing protein 5 | XRCC5 | 16.4 | 64.243 | 2.23381025 | 0.00371267 |
Q8J015 | 60S ribosomal protein L13a | RPL13A | 14.3 | 23.558 | 2.13155137 | 0.02407208 |
Q9H9H4 | Vacuolar protein sorting-associated protein 37B | VPS37B | 9.2 | 20.654 | 2.09589466 | 0.02963136 |
P02461 | Collagen alpha-1(III) chain | COL3A1 | 1.6 | 138.56 | 2.09300108 | 0.02280738 |
P27635 | 60S ribosomal protein L10 | RPL10 | 40.7 | 12.285 | 2.08918466 | 0.03237894 |
Q8WUM4 | Programmed cell death 6-interacting protein | PDCD6IP | 39.6 | 96.022 | 2.05463106 | 0.00321507 |
P07237 | Protein disulfide-isomerase | P4HB | 14.2 | 16.215 | 2.04047326 | 0.0174391 |
Q9HD42 | Charged multivesicular body protein 1a | CHMP1A | 30.7 | 15.301 | 2.02701323 | 0.01108996 |
Table S2
Protein IDs | Protein names | Gene names | Sequence coverage [%] | Mol. weight [kDa] | SW620/SW480 | P. value |
---|---|---|---|---|---|---|
P11169 | Solute carrier family 2, facilitated glucose transporter member 3 | SLC2A3 | 20.8 | 31.874 | 0.011749168 | 0.0296785 |
O94832 | Unconventional myosin-Id | MYO1D | 28.2 | 116.2 | 0.038787995 | 0.00027188 |
P29374 | Retinol-binding protein 1 | RBP1 | 56 | 10.644 | 0.072787169 | 0.00388128 |
P98172 | Ephrin-B1 | EFNB1 | 16.2 | 38.006 | 0.074930021 | 0.00119442 |
P08133 | Annexin A6 | ANXA6 | 37.1 | 75.872 | 0.078012029 | 0.00031708 |
Q96TA1 | Niban-like protein 1 | C9orf88 | 10.9 | 82.682 | 0.126490412 | 0.01065253 |
Q9Y696 | Chloride intracellular channel protein 4 | CLIC4 | 26.6 | 26.694 | 0.137963444 | 0.03000887 |
P05556 | Integrin beta-1 | ITGB1 | 20.9 | 88.414 | 0.14864844 | 0.00146909 |
P16422 | Epithelial cell adhesion molecule | EPCAM | 34.8 | 37.893 | 0.163589346 | 0.00048008 |
P61081 | NEDD8-conjugating enzyme Ubc12 | UBE2M | 8.7 | 20.9 | 0.165887295 | 0.0115204 |
P14222 | Perforin-1 | PRF1 | 29.7 | 61.346 | 0.166465764 | 0.00887923 |
Q969P0 | Immunoglobulin superfamily member 8 | IGSF8 | 21.2 | 65.033 | 0.168737967 | 1.6776E-05 |
O43707 | Alpha-actinin-4 | ACTN4 | 63.7 | 104.85 | 0.184502626 | 0.00046581 |
O00159 | Unconventional myosin-Ic | MYO1C | 21.8 | 118.99 | 0.196118715 | 0.00091981 |
O00468 | Agrin | AGRN | 27 | 203.09 | 0.204624716 | 0.00258642 |
P11166 | Solute carrier family 2, facilitated glucose transporter member 1 | SLC2A1 | 12.5 | 45.869 | 0.227212304 | 0.00028607 |
P10909 | Clusterin | Clusterin | 17.4 | 57.832 | 0.231262489 | 0.01649402 |
O75083 | WD repeat-containing protein 1 | WDR1 | 25.7 | 66.227 | 0.235138519 | 0.01728513 |
P18564 | Integrin beta-6 | ITGB6 | 1.3 | 75.849 | 0.238701666 | 0.00190898 |
P43007 | Neutral amino acid transporter A | SLC1A4 | 2.4 | 47.726 | 0.248104871 | 0.00052163 |
Q01650 | Large neutral amino acids transporter small subunit 1 | SLC7A5 | 9.9 | 55.01 | 0.254952388 | 0.00514504 |
O00186 | Syntaxin-binding protein 3 | STXBP3 | 12 | 67.764 | 0.257123529 | 0.03321712 |
P63218 | Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-5 | GNG5 | 23.5 | 7.3184 | 0.258537813 | 0.01695074 |
P61026 | Ras-related protein Rab-10 | RAB10 | 20.5 | 22.541 | 0.260031307 | 0.0027144 |
P13987 | CD59 glycoprotein | CD59 | 29.6 | 11.985 | 0.274520883 | 0.000113 |
Q15758 | Neutral amino acid transporter B(0) | SLC1A5 | 25.9 | 56.583 | 0.275747651 | 7.5488E-05 |
Q9UHD8 | Septin-9 | Septin-9 | 22 | 23.971 | 0.279355157 | 0.00055609 |
P12814 | Alpha-actinin-1 | ACTN1 | 50.4 | 103.06 | 0.286041695 | 0.00051225 |
J3KPF3 | 4F2 cell-surface antigen heavy chain | SLC3A2 | 36.4 | 64.872 | 0.287544691 | 6.9295E-05 |
P55060 | Exportin-2 | CSE1L | 5.6 | 110.42 | 0.29415306 | 0.01109174 |
P31947 | 14–3–3 protein sigma | SFN | 43.1 | 27.774 | 0.294432959 | 0.00401826 |
Q16643 | Drebrin | DBN1 | 14.8 | 36.471 | 0.320680503 | 0.02215882 |
P59998 | Actin-related protein 2/3 complex subunit 4 | ARPC4 | 19.9 | 21.058 | 0.324678693 | 0.00407438 |
P61006 | Ras-related protein Rab-8A | RAB8A | 25.1 | 23.668 | 0.324702465 | 0.03330491 |
V9HWH9 | Protein S100-A11 | S100A11 | 42.9 | 11.74 | 0.335125777 | 0.00294128 |
Q01518 | Adenylyl cyclase-associated protein 1 | CAP1 | 16.2 | 47.39 | 0.337232915 | 0.03235735 |
P48509 | CD151 antigen | CD151 | 5.1 | 19.792 | 0.355641041 | 0.02287931 |
P17301 | Integrin alpha-2 | ITGA2 | 4.3 | 129.29 | 0.362271878 | 0.0435853 |
P26447 | Protein S100-A4 | S100A4 | 46.5 | 11.728 | 0.3624522 | 0.01947124 |
Q9H223 | EH domain-containing protein 4 | EHD4 | 35.1 | 61.155 | 0.370830425 | 0.00295808 |
Q1KLZ0 | Actin, cytoplasmic 2 | PS1TP5BP1 | 70.1 | 41.722 | 0.379410112 | 0.04247494 |
P27348 | 14–3–3 protein theta | YWHAQ | 53.9 | 27.764 | 0.391324245 | 0.00384362 |
Q92522 | Histone H1x | H1FX | 26.3 | 22.487 | 0.394994486 | 0.02046652 |
P54709 | Sodium/potassium-transporting ATPase subunit beta-3 | ATP1B3 | 31.2 | 31.512 | 0.39581471 | 0.00900052 |
V9HWF5 | Peptidyl-prolyl cis-trans isomerase A | PPIA | 48.5 | 18.012 | 0.397199254 | 0.04077159 |
Q12959 | Disks large homolog 1 | DLG1 | 4.9 | 75.96 | 0.398631587 | 0.01565711 |
P62879 | Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-2 | GNB2 | 48.5 | 37.331 | 0.402599067 | 0.00485033 |
P35241 | Radixin | RDX | 30 | 68.563 | 0.405935961 | 0.00974889 |
Q14847 | LIM and SH3 domain protein 1 | LASP1 | 26.4 | 29.645 | 0.406985226 | 0.01617477 |
P31431 | Syndecan-4 | SDC4 | 7.8 | 71.657 | 0.407307866 | 0.0159348 |
B6VEX4 | Abl interactor 1 | ABI1 | 9.3 | 42.637 | 0.407476812 | 0.02251877 |
Q9ULV4 | Coronin-1C | CORO1C | 21.9 | 53.28 | 0.40985014 | 0.00343122 |
Q86W92 | Liprin-beta-1 | PPFIBP1 | 7 | 114.02 | 0.41468217 | 0.02693422 |
P03971 | Macrophage migration inhibitory factor | MIF | 17.4 | 12.476 | 0.418381567 | 0.0001584 |
Q86VI3 | Ras GTPase-activating-like protein IQGAP3 | IQGAP3 | 4.5 | 179.48 | 0.420461641 | 0.02336381 |
P02786 | Transferrin receptor protein 1 | TFRC | 21.7 | 84.872 | 0.425257535 | 0.00214362 |
P04439 | HLA class I histocompatibility antigen, A alpha chain | HLA-A | 34.8 | 31.623 | 0.427826788 | 0.01397475 |
P62136 | Serine/threonine-protein phosphatase PP1-alpha catalytic subunit | PPP1CA | 21.1 | 38.631 | 0.432666807 | 0.00477209 |
Q9NRW3 | DNA dC->dU-editing enzyme APOBEC-3C | APOBEC3C | 6.8 | 22.796 | 0.440265332 | 0.04194558 |
Q86 × 29 | Lipolysis-stimulated lipoprotein receptor | LSR | 23.4 | 64.346 | 0.440534774 | 0.00496959 |
P02649 | Apolipoprotein E | APOE | 27.1 | 36.154 | 0.440589659 | 0.01600696 |
P18085 | ADP-ribosylation factor 4 | ARF4 | 41.1 | 20.511 | 0.443431257 | 0.01993945 |
P55011 | Solute carrier family 12 member 2 | SLC12A2 | 8.8 | 129.72 | 0.443858211 | 0.00511427 |
P36873 | Serine/threonine-protein phosphatase PP1-gamma catalytic subunit | PPP1CC | 24.5 | 33.773 | 0.445747816 | 0.00223503 |
Q5ZPR3 | CD276 antigen | CD276 | 10.3 | 41.709 | 0.447245301 | 0.00571664 |
V9HWB9 | L-lactate dehydrogenase;L-lactate dehydrogenase A chain | LDHA | 32.8 | 36.688 | 0.458180976 | 0.00041123 |
O15143 | Actin-related protein 2/3 complex subunit 1B | ARPC1B | 14.2 | 40.949 | 0.458231108 | 0.00228224 |
Q71UI9 | Histone H2A.V | H2AFV | 31.2 | 13.509 | 0.464114005 | 0.03317498 |
P21291 | Cysteine and glycine-rich protein 1 | CSRP1 | 33.8 | 16.94 | 0.468629793 | 0.02915667 |
V9HVZ4 | Glyceraldehyde-3-phosphate dehydrogenase | GAPDH | 46.9 | 36.053 | 0.46891796 | 0.00324459 |
Q8N5I2 | Arrestin domain-containing protein 1 | ARRDC1 | 9.2 | 29.254 | 0.470525512 | 0.0043285 |
P54707 | Potassium-transporting ATPase alpha chain 2 | ATP12A | 4.6 | 115.51 | 0.474831724 | 0.00997908 |
P15529 | Membrane cofactor protein | CD46 | 16.7 | 8.3916 | 0.476142784 | 0.00546069 |
Q9H5V8 | CUB domain-containing protein 1 | CDCP1 | 11.1 | 71.678 | 0.477411441 | 0.00845617 |
P05023 | Sodium/potassium-transporting ATPase subunit alpha-1 | ATP1A1 | 39.5 | 112.89 | 0.478365161 | 0.01275306 |
V9HWA6 | Destrin | DSTN | 46.7 | 18.506 | 0.484528666 | 0.03130172 |
P07355 | Annexin A2 | ANXA2 | 63.4 | 38.576 | 0.486185177 | 0.0028782 |
P46940 | Ras GTPase-activating-like protein IQGAP1 | IQGAP1 | 27.4 | 189.28 | 0.487387331 | 0.00379913 |
P63261 | Actin-related protein 2/3 complex subunit 1B | ACTG1 | 70.1 | 41.792 | 0.489096205 | 0.0144896 |
O15144 | Actin-related protein 2/3 complex subunit 2 | ARPC2 | 9 | 34.333 | 0.492376837 | 0.01353878 |
P09327 | Villin-1 | VIL1 | 38.5 | 92.724 | 0.492763587 | 0.00680854 |
Q9H4M9 | EH domain-containing protein 1 | EHD1 | 21.3 | 60.626 | 0.492878223 | 0.00197344 |
P63000 | Ras-related C3 botulinum toxin substrate 1 | RAC1 | 40.6 | 21.45 | 0.496595902 | 0.00756952 |
Table S3
protein IDs | Protein names | Gene names | Sequence coverage [%] | Mol. weight [kDa] | Sequence coverage 480-mean [%] | Sequence coverage 620-mean [%] | Intensity | LFQ intensity 480-mean | LFQ intensity 620-mean |
---|---|---|---|---|---|---|---|---|---|
Q04941 | Proteolipid protein 2 | PLP2 | 8.6 | 16.691 | 0 | 5.733333 | 80,528,000 | 0 | 41,380,666.7 |
P31942 | Heterogeneous nuclear ribonucleoprotein H3 | HNRPH3 | 7.9 | 22.322 | 0 | 7.9 | 67,404,000 | 0 | 33,249,433.3 |
P15328 | Folate receptor alpha | FOLR1 | 9.2 | 24.281 | 0 | 5.666667 | 37,123,000 | 0 | 19,172,000 |
Q05193 | Dynamin-1 | DNM1 | 12.2 | 35.248 | 0 | 8.133333 | 59,177,000 | 0 | 29,509,666.7 |
O00391 | Sulfhydryl oxidase 1 | QSOX1 | 4.1 | 66.821 | 2.2 | 1.333333 | 66,597,000 | 0 | 17,429,333.3 |
O00410 | Importin-5 | IPO5 | 4.2 | 81.23 | 0 | 2.8 | 24,252,000 | 0 | 12,211,666.7 |
Q96AY3 | Peptidyl-prolyl cis-trans isomerase FKBP10 | FKBP10 | 2.4 | 56.158 | 0 | 1.6 | 55,640,000 | 0 | 27,467,233.3 |
Q9UHB9 | Signal recognition particle subunit SRP68 | SRP68 | 4.2 | 60.226 | 0.833333 | 1.133333 | 14,730,000 | 0 | 7,536,333.33 |
Q13275 | Semaphorin-3F | SEMA3F | 7.2 | 30.259 | 0 | 4.8 | 39,158,000 | 0 | 19,862,666.7 |
P12830 | Cadherin-1 | CDH1 | 2.8 | 97.485 | 0.6 | 1 | 1.1E+08 | 0 | 49,516,333.3 |
P30408 | Transmembrane 4 L6 family member 1 | TM4SF1 | 8.9 | 10.989 | 0 | 8.9 | 50,396,000 | 0 | 25,707,000 |
P05067 | Amyloid beta A4 protein | APP | 8.5 | 54.981 | 0 | 5.033333 | 83,773,000 | 0 | 41,696,633.3 |
Q5HYI8 | Rab-like protein 3 | RABL3 | 20.8 | 5.5193 | 0 | 20.8 | 1.04E+08 | 0 | 50,064,666.7 |
P24821 | Tenascin | TNC | 34.3 | 220.85 | 12.43333 | 31.96667 | 8.61E+08 | 0 | 433,993,333 |
P63010 | AP-2 complex subunit beta-1 | AP2B1 | 7.3 | 18.606 | 0 | 4.866667 | 55,812,000 | 0 | 27,598,000 |
P26599 | Polypyrimidine tract-binding protein 1 | PTBP1 | 9 | 27.114 | 0 | 6 | 2.19E+08 | 0 | 108,196,667 |
P06241 | Tyrosine-protein kinase Fyn | FYN | 6.1 | 60.761 | 3.4 | 3.966667 | 40,373,000 | 0 | 20,373,000 |
P35326 | Small proline-rich protein 2A | SPRR2A | 43.1 | 7.9653 | 0 | 20.4 | 45,032,000 | 0 | 22,238,333.3 |
P25787 | Proteasome subunit alpha type-2 | PSMA2 | 13.2 | 25.84 | 1.433333 | 6 | 38,171,000 | 0 | 11,320,766.7 |
O00567 | Nucleolar protein 56 | NOP56 | 10.7 | 31.253 | 0 | 8.566667 | 36,208,000 | 0 | 18,588,333.3 |
Q5M9N0 | Coiled-coil domain-containing protein 158 | CCDC158 | 0.9 | 127.14 | 0.3 | 0.9 | 43,547,000 | 0 | 21,854,000 |
O94985 | Calsyntenin-1 | CLSTN1 | 2.7 | 88.039 | 0 | 2.033333 | 84,046,000 | 0 | 41,910,333.3 |
Q6P988 | Palmitoleoyl-protein carboxylesterase NOTUM | NOTUM | 4.8 | 55.699 | 0 | 3.8 | 73,588,000 | 0 | 37,186,666.7 |
P35232 | Prohibitin | PHB | 9.9 | 29.82 | 1.233333 | 4.133333 | 1.11E+08 | 0 | 55,058,533.3 |
O14986 | Phosphatidylinositol 4-phosphate 5-kinase type-1 beta | STM7 | 4.5 | 37.786 | 0 | 4.5 | 3.03E+08 | 0 | 153,100,000 |
Q8NCR9 | Clarin-3 | CLRN3 | 12.4 | 25.321 | 3.866667 | 6.6 | 79,843,000 | 0 | 34,011,666.7 |
Q8NDC0 | MAPK-interacting and spindle-stabilizing protein-like | MAPK1IP1L | 6.9 | 24.269 | 0 | 4.6 | 58,364,000 | 0 | 30,015,333.3 |
Q8WUJ3 | Cell migration-inducing and hyaluronan-binding protein | CEMIP | 11.2 | 153 | 0 | 7.2 | 5.88E+08 | 0 | 283,110,000 |
Q92896 | Golgi apparatus protein 1 | GLG1 | 3.4 | 134.55 | 0 | 2.766667 | 1.37E+08 | 0 | 68,899,333.3 |
Q9H3U7 | SPARC-related modular calcium-binding protein 2 | SMOC2 | 17.7 | 49.674 | 0 | 16.86667 | 8.83E+08 | 0 | 448,096,667 |
Table S4
Protein IDs | Protein names | Gene names | Sequence coverage [%] | Mol. weight [kDa] | Sequence coverage 480-mean [%] | Sequence coverage 620-mean [%] | Intensity | LFQ intensity 480-mean | LFQ intensity 620-mean |
---|---|---|---|---|---|---|---|---|---|
P05121 | Plasminogen activator inhibitor 1 | SERPINE1 | 2.5 | 45.059 | 1.666666667 | 0 | 36,502,000 | 11,615,333.33 | 0 |
P50151 | Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-10 | GNG10 | 42.6 | 7.2053 | 42.6 | 0 | 98,311,000 | 29,713,666.67 | 0 |
P41221 | Protein Wnt-5a | WNT5A | 9 | 40.886 | 7 | 0 | 213,720,000 | 62,549,666.67 | 0 |
P31146 | Coronin-1A | CORO1A | 3.9 | 51.026 | 1.866666667 | 0 | 25,254,000 | 8,137,000 | 0 |
Q92817 | Envoplakin | EVPL | 0.4 | 231.63 | 0.4 | 0.133333333 | 22,870,000 | 6,611,900 | 0 |
P11234 | Ras-related protein Ral-B | RALB | 15.5 | 23.408 | 11.63333333 | 5.3 | 140,580,000 | 41,686,666.67 | 0 |
P14324 | Farnesyl pyrophosphate synthase | FDPS | 3.6 | 28.594 | 2.4 | 0 | 23,921,000 | 7,485,933.333 | 0 |
Q07157 | Tight junction protein ZO-1 | TJP1 | 1.4 | 187.85 | 1.166666667 | 0 | 84,367,000 | 24,778,666.67 | 0 |
P36969 | Phospholipid hydroperoxide glutathione peroxidase | GPX4 | 10.4 | 15.438 | 6.933333333 | 0 | 12,080,000 | 3,787,700 | 0 |
P41250 | Glycine–tRNA ligase | GARS | 5 | 77.53 | 3.3 | 0 | 55,583,000 | 16,794,333.33 | 0 |
Q92598 | Heat shock protein 105 kDa | HSPH1 | 6.1 | 88.841 | 4.1 | 0 | 72,896,000 | 21,843,000 | 0 |
P07954 | Fumarate hydratase, mitochondrial | FH | 7.5 | 54.636 | 4.5 | 0.966666667 | 70,255,000 | 16,694,400 | 0 |
P02792 | Ferritin light chain | FTL | 24.6 | 20.019 | 11.06666667 | 0 | 99,553,000 | 32,634,000 | 0 |
P23469 | Receptor-type tyrosine-protein phosphatase epsilon | PTPRE | 3.1 | 74.564 | 1.666666667 | 0 | 17,773,000 | 5,656,600 | 0 |
P50895 | Basal cell adhesion molecule | BCAM | 15.8 | 8.162 | 10.53333333 | 0 | 29,297,000 | 9,369,333.333 | 0 |
O43242 | 26S proteasome non-ATPase regulatory subunit 3 | PSMD3 | 3 | 57.232 | 3 | 0 | 91,877,000 | 27,149,333.33 | 0 |
Q9NRY6 | Phospholipid scramblase 3 | PLSCR3 | 13.6 | 31.678 | 8.6 | 0.9 | 91,556,000 | 28,561,000 | 0 |
P43121 | Cell surface glycoprotein MUC18 | MCAM | 7.7 | 59.019 | 5.966666667 | 0 | 339,680,000 | 100,331,333.3 | 0 |
P21980 | Protein-glutamine gamma-glutamyltransferase 2 | TGM2 | 18.8 | 68.648 | 17.56666667 | 0 | 1,175,800,000 | 351,616,666.7 | 0 |
P56199 | Integrin alpha-1 | ITGA1 | 3.2 | 130 | 2.033333333 | 0 | 142,870,000 | 42,286,666.67 | 0 |
P48059 | LIM and senescent cell antigen-like-containing domain protein 1 | LIMS1 | 23 | 13.705 | 14.8 | 0 | 47,810,000 | 13,719,666.67 | 0 |
B7Z2Q0 | Actin filament-associated protein 1-like 2 | AFAP1L2 | 3 | 97.193 | 3 | 0 | 112,520,000 | 33,462,666.67 | 0 |
B9EG73 | Dedicator of cytokinesis protein 9 | DOCK9 | 3.4 | 233.83 | 2.566666667 | 0 | 307,260,000 | 89,682,666.67 | 0 |
P09758 | Tumor-associated calcium signal transducer 2 | TACSTD2 | 16.7 | 35.723 | 15.46666667 | 0 | 90,281,000 | 26,570,666.67 | 0 |
P35080 | Profilin-2 | PFN2 | 15.4 | 9.7982 | 15.4 | 0 | 61,479,000 | 18,287,333.33 | 0 |
P99999 | Cytochrome c | CYCS | 18.8 | 11.333 | 12.53333333 | 0 | 39,851,000 | 12,847,533.33 | 0 |
Q9H0Z9 | RNA-binding protein 38 | RBM38 | 8.9 | 13.604 | 8.9 | 0 | 184,370,000 | 55,261,333.33 | 0 |
O75390 | Citrate synthase | CS | 24.2 | 7.3535 | 16.13333333 | 0 | 51,753,000 | 14,528,333.33 | 0 |
O95456 | Proteasome assembly chaperone 1 | PSMG1 | 15.9 | 7.182 | 10.6 | 0 | 20,264,000 | 6,455,133.333 | 0 |
P09496 | Clathrin light chain A | CLTA | 3.5 | 27.818 | 3.5 | 0 | 72,129,000 | 21,608,000 | 0 |
P60983 | Glia maturation factor beta | GMFB | 33.3 | 17.512 | 15.96666667 | 0 | 95,964,000 | 30,852,666.67 | 0 |
Q9Y4F1 | FERM, RhoGEF and pleckstrin domain-containing protein 1 | FARP1 | 6 | 24.749 | 4 | 0 | 23,814,000 | 6,839,166.667 | 0 |
P62714 | Serine/threonine-protein phosphatase 2A catalytic subunit beta isoform | PPP2CB | 16.9 | 13.87 | 11.26666667 | 0 | 36,508,000 | 10,631,333.33 | 0 |
Q13330 | Metastasis-associated protein MTA1 | MTA1 | 21.8 | 5.8987 | 21.8 | 0 | 31,412,000 | 9,423,566.667 | 0 |
O95471 | Claudin-7 | CLDN7 | 25.6 | 13.649 | 21.46666667 | 0 | 189,410,000 | 56,309,666.67 | 0 |
Q13433 | Zinc transporter ZIP6 | SLC39A6 | 4.4 | 30.912 | 2.933333333 | 0 | 46,342,000 | 14,534,333.33 | 0 |
Q08828 | Adenylate cyclase type 1 | ADCY1 | 12.5 | 8.7119 | 12.5 | 0 | 32,293,000 | 9,449,100 | 0 |
O15230 | Laminin subunit alpha-5 | LAMA5 | 2.4 | 399.73 | 1.9 | 0 | 228,260,000 | 68,682,666.67 | 0 |
O60635 | Tetraspanin-1 | TSPAN1 | 5.4 | 26.301 | 5.4 | 0 | 162,040,000 | 48,687,000 | 0 |
P09758 | Tumor-associated calcium signal transducer 2 | TACSTD2 | 16.7 | 35.709 | 15.46666667 | 0 | 264,340,000 | 77,985,333.33 | 0 |
P62760 | Visinin-like protein 1 | VSNL1 | 19.9 | 22.142 | 10.63333333 | 0 | 207,320,000 | 60,677,000 | 0 |
Q03405 | Urokinase plasminogen activator surface receptor | PLAUR | 30.1 | 36.978 | 21.8 | 0 | 421,970,000 | 126,686,666.7 | 0 |
O75874 | Isocitrate dehydrogenase [NADP] cytoplasmic | IDH1 | 6.5 | 41.832 | 3.333333333 | 0 | 56,665,000 | 17,600,000 | 0 |
Q12846 | Syntaxin-4 | STX4 | 9.1 | 34.18 | 7.866666667 | 0 | 114,630,000 | 33,291,000 | 0 |
Q13200 | 26S proteasome non-ATPase regulatory subunit 2 | PSMD2 | 2.5 | 64.587 | 2.5 | 0 | 21,373,000 | 6,739,400 | 0 |
P19256 | Lymphocyte function-associated antigen 3 | CD58 | 10.1 | 13.732 | 8.966666667 | 0 | 45,701,000 | 13,166,000 | 0 |
O00487 | 26S proteasome non-ATPase regulatory subunit 14 | PSMD14 | 16.9 | 16.532 | 11.26666667 | 0 | 42,554,000 | 13,452,333.33 | 0 |
P36543 | V-type proton ATPase subunit E 1 | ATP6V1E1 | 5.8 | 26.145 | 3.866666667 | 0 | 13,687,000 | 3,852,366.667 | 0 |
Q54A15 | Fermitin family homolog 1 | FERMT1 | 9.9 | 77.408 | 6.633333333 | 0 | 170,830,000 | 50,556,666.67 | 0 |
Q562Z4 | Actin-like protein | ACT | 33 | 11.555 | 29.76666667 | 23.3 | 48,586,000 | 13,652,666.67 | 0 |
P08183 | Multidrug resistance protein 1 | ABCB1 | 3 | 116.72 | 2 | 0 | 82,775,000 | 24,464,700 | 0 |
O14936 | Peripheral plasma membrane protein CASK | CASK | 7.8 | 66.603 | 4.866666667 | 0 | 128,100,000 | 36,728,333.33 | 0 |
P05362 | Intercellular adhesion molecule 1 | ICAM1 | 11.3 | 57.768 | 11.3 | 0 | 292,830,000 | 87,657,666.67 | 0 |
Q14683 | Structural maintenance of chromosomes protein 1A | SMC1A | 6 | 48.887 | 3.266666667 | 1.366666667 | 95,721,000 | 23,194,000 | 0 |
Q6FHV6 | Gamma-enolase | ENO2 | 15.2 | 47.268 | 11.5 | 7.5 | 36,401,000 | 11,354,700 | 0 |
P00492 | Hypoxanthine-guanine phosphoribosyltransferase | HPRT1 | 16.1 | 24.588 | 9.033333333 | 0 | 49,736,000 | 14,969,666.67 | 0 |
P32004 | Neural cell adhesion molecule L1 | L1CAM | 4.5 | 59.802 | 2.133333333 | 0 | 17,346,000 | 4,990,466.667 | 0 |
Q13509 | Tubulin beta-3 chain | TUBB3 | 34 | 45.622 | 29.06666667 | 25.76666667 | 94,686,000 | 16,839,333.33 | 0 |
Q9UBT3 | Dickkopf-related protein 4 | DKK4 | 33.5 | 24.875 | 33.16666667 | 0 | 1,338,800,000 | 396,080,000 | 0 |
P30044 | Peroxiredoxin-5 | PRDX5 | 56.2 | 17.031 | 32.5 | 0 | 458,660,000 | 147,806,666.7 | 0 |
CRediT authorship contribution statement
Xinlu Liu: Methodology, Software, Validation, Formal analysis, Data curation, Writing – original draft, Writing – review & editing, Visualization, Supervision. Na Li: Software, Formal analysis, Data curation, Writing – original draft, Visualization. Chi Zhang: Formal analysis, Writing – original draft, Visualization. Xiaoyu Wu: Formal analysis, Writing – original draft. Shoujia Zhang: Writing – original draft. Gang Dong: Writing – original draft. Ge Liu: Methodology.
Declaration of Competing Interest
The authors declare no conflict of interest.
Acknowledgements
We acknowledge the technical support of this work by Shanghai Bioprofile Technology Company Ltd. We also acknowledge the financially supported in this work by Genetron Holdings Limited Corporation. We are also grateful for the English language editing from Editage (www.editage.cn).
References
- 1.Siegel R.L., Miller K.D., Fuchs H.E., Jemal A. Cancer statistics, 2021. CA Cancer J. Clin. 2021;71:7–33. doi: 10.3322/caac.21654. [DOI] [PubMed] [Google Scholar]
- 2.Fakih M.G. Metastatic colorectal cancer: current state and future directions. J. Clin. Oncol. 2015;33:1809–1824. doi: 10.1200/JCO.2014.59.7633. [DOI] [PubMed] [Google Scholar]
- 3.Basak D., Uddin M.N., Hancock J. The role of oxidative stress and its counteractive utility in colorectal cancer (CRC) Cancers. 2020;12 doi: 10.3390/cancers12113336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Deepak K.G.K., Vempati R., Nagaraju G.P., Dasari V.R., S N., Rao D.N., Malla R.R. Tumor microenvironment: challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol. Res. 2020;153 doi: 10.1016/j.phrs.2020.104683. [DOI] [PubMed] [Google Scholar]
- 5.Al-Nedawi K., Meehan B., Rak J. Microvesicles: messengers and mediators of tumor progression. Cell Cycle. 2009;8:2014–2018. doi: 10.4161/cc.8.13.8988. [DOI] [PubMed] [Google Scholar]
- 6.Shao H., Im H., Castro C.M., Breakefield X., Weissleder R., Lee H. New technologies for analysis of extracellular vesicles. Chem. Rev. 2018;118:1917–1950. doi: 10.1021/acs.chemrev.7b00534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Guo A.J., Wang F.J., Ji Q., Geng H.W., Yan X., Wang L.Q., Tie W.W., Liu X.Y., Thorne R.F., Liu G., Xu A.M. Proteome analyses reveal S100A11, S100P, and RBM25 are tumor biomarkers in colorectal cancer. Proteomics Clin. Appl. 2020 doi: 10.1002/prca.202000056. [DOI] [PubMed] [Google Scholar]
- 8.Shannon P., Markiel A., Ozier O., Baliga N.S., Wang J.T., Ramage D., Amin N., Schwikowski B., Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13:2498–2504. doi: 10.1101/gr.1239303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Liu X., Weng Y., Liu P., Sui Z., Zhou L., Huang Y., Zhang L., Zhang Y., Tan X. Identification of PGAM1 as a putative therapeutic target for pancreatic ductal adenocarcinoma metastasis using quantitative proteomics. Onco Targets Ther. 2018;11:3345–3357. doi: 10.2147/OTT.S162470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Tassi E., Henke R.T., Bowden E.T., Swift M.R., Kodack D.P., Kuo A.H., Maitra A., Wellstein A. Expression of a fibroblast growth factor-binding protein during the development of adenocarcinoma of the pancreas and colon. Cancer Res. 2006;66:1191–1198. doi: 10.1158/0008-5472.CAN-05-2926. [DOI] [PubMed] [Google Scholar]
- 11.Okamoto T., Tanaka Y., Kan M., Sakamoto A., Takada K., Sato J.D. Expression of fibroblast growth factor binding protein HBp17 in normal and tumor cells. In Vitro Cell. Dev. Biol. Anim. 1996;32:69–71. doi: 10.1007/BF02723035. [DOI] [PubMed] [Google Scholar]
- 12.Aigner A., Renneberg H., Bojunga J., Apel J., Nelson P.S., Czubayko F. Ribozyme-targeting of a secreted FGF-binding protein (FGF-BP) inhibits proliferation of prostate cancer cells in vitro and in vivo. Oncogene. 2002;21:5733–5742. doi: 10.1038/sj.onc.1205560. [DOI] [PubMed] [Google Scholar]
- 13.Kagan B.L., Henke R.T., Cabal-Manzano R., Stoica G.E., Nguyen Q., Wellstein A., Riegel A.T. Complex regulation of the fibroblast growth factor-binding protein in MDA- MB-468 breast cancer cells by CCAAT/enhancer-binding protein beta. Cancer Res. 2003;63:1696–1705. [PubMed] [Google Scholar]
- 14.Chen J., Liu Q.M., Du P.C., Ning D., Mo J., Zhu H.D., Wang C., Ge Q.Y., Cheng Q., Zhang X.W., Fan Y.W., Liang H.F., Chu L., Chen X.P., Zhang B.X., Jiang L. Type-2 11beta-hydroxysteroid dehydrogenase promotes the metastasis of colorectal cancer via the Fgfbp1-AKT pathway. Am. J. Cancer Res. 2020;10:662–673. [PMC free article] [PubMed] [Google Scholar]
- 15.Zhang Z., Liu M., Hu Q., Xu W., Liu W., Sun Q., Ye Z., Fan G., Xu X., Yu X., Ji S., Qin Y. FGFBP1, a downstream target of the FBW7/c-Myc axis, promotes cell proliferation and migration in pancreatic cancer. Am. J. Cancer Res. 2019;9:2650–2664. [PMC free article] [PubMed] [Google Scholar]
- 16.Tsygankova O.M., Ma C., Tang W., Korch C., Feldman M.D., Lv Y., Brose M.S., Meinkoth J.L. Downregulation of Rap1GAP in human tumor cells alters cell/matrix and cell/cell adhesion. Mol. Cell. Biol. 2010;30:3262–3274. doi: 10.1128/MCB.01345-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kometani K., Ishida D., Hattori M., Minato N. Rap1 and SPA-1 in hematologic malignancy. Trends Mol. Med. 2004;10:401–408. doi: 10.1016/j.molmed.2004.06.004. [DOI] [PubMed] [Google Scholar]
- 18.Hattori M., Minato N. Rap1 GTPase: functions, regulation, and malignancy. J. Biochem. 2003;134:479–484. doi: 10.1093/jb/mvg180. [DOI] [PubMed] [Google Scholar]
- 19.Shimizu Y., Hamazaki Y., Hattori M., Doi K., Terada N., Kobayashi T., Toda Y., Yamasaki T., Inoue T., Kajita Y., Maeno A., Kamba T., Mikami Y., Kamoto T., Yamada T., Kanno T., Yoshikawa K., Ogawa O., Minato N., Nakamura E. SPA-1 controls the invasion and metastasis of human prostate cancer. Cancer Sci. 2011;102:828–836. doi: 10.1111/j.1349-7006.2011.01876.x. [DOI] [PubMed] [Google Scholar]
- 20.Kodama J., Hashimoto I., Seki N., Hongo A., Yoshinouchi M., Okuda H., Kudo T. Thrombospondin-1 and -2 messenger RNA expression in invasive cervical cancer: correlation with angiogenesis and prognosis. Clin. Cancer Res. 2001;7:2826–2831. [PubMed] [Google Scholar]
- 21.Yamaguchi M., Sugio K., Ondo K., Yano T., Sugimachi K. Reduced expression of thrombospondin-1 correlates with a poor prognosis in patients with non-small cell lung cancer. Lung Cancer. 2002;36:143–150. doi: 10.1016/s0169-5002(01)00470-6. [DOI] [PubMed] [Google Scholar]
- 22.Ioachim E., Damala K., Tsanou E., Briasoulis E., Papadiotis E., Mitselou A., Charhanti A., Doukas M., Lampri L., Arvanitis D.L. Thrombospondin-1 expression in breast cancer: prognostic significance and association with p53 alterations, tumour angiogenesis and extracellular matrix components. Histol. Histopathol. 2012;27:209–216. doi: 10.14670/HH-27.209. [DOI] [PubMed] [Google Scholar]
- 23.Teraoku H., Morine Y., Ikemoto T., Saito Y., Yamada S., Yoshikawa M., Takasu C., Higashijima J., Imura S., Shimada M. Role of thrombospondin-1 expression in colorectal liver metastasis and its molecular mechanism. J. Hepatobiliary Pancreat. Sci. 2016;23:565–573. doi: 10.1002/jhbp.376. [DOI] [PubMed] [Google Scholar]
- 24.Chiavarina B., Costanza B., Ronca R., Blomme A., Rezzola S., Chiodelli P., Giguelay A., Belthier G., Doumont G., Van Simaeys G., Lacroix S., Yokobori T., Erkhem-Ochir B., Balaguer P., Cavailles V., Fabbrizio E., Di Valentin E., Gofflot S., Detry O., Jerusalem G., Goldman S., Delvenne P., Bellahcene A., Pannequin J., Castronovo V., Turtoi A. Metastatic colorectal cancer cells maintain the TGFbeta program and use TGFBI to fuel angiogenesis. Theranostics. 2021;11:1626–1640. doi: 10.7150/thno.51507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Li Z., Luo R.T., Mi S., Sun M., Chen P., Bao J., Neilly M.B., Jayathilaka N., Johnson D.S., Wang L., Lavau C., Zhang Y., Tseng C., Zhang X., Wang J., Yu J., Yang H., Wang S.M., Rowley J.D., Chen J., Thirman M.J. Consistent deregulation of gene expression between human and murine MLL rearrangement leukemias. Cancer Res. 2009;69:1109–1116. doi: 10.1158/0008-5472.CAN-08-3381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kang S., Dong S.M., Park N.H. Frequent promoter hypermethylation of TGFBI in epithelial ovarian cancer. Gynecol. Oncol. 2010;118:58–63. doi: 10.1016/j.ygyno.2010.03.025. [DOI] [PubMed] [Google Scholar]
- 27.Shah J.N., Shao G., Hei T.K., Zhao Y. Methylation screening of the TGFBI promoter in human lung and prostate cancer by methylation-specific PCR. BMC Cancer. 2008;8:284. doi: 10.1186/1471-2407-8-284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tomioka H., Morita K., Hasegawa S., Omura K. Gene expression analysis by cDNA microarray in oral squamous cell carcinoma. J. Oral Pathol. Med. 2006;35:206–211. doi: 10.1111/j.1600-0714.2006.00410.x. [DOI] [PubMed] [Google Scholar]
- 29.Owusu-Ansah K.G., Song G., Chen R., Edoo M.I.A., Li J., Chen B., Wu J., Zhou L., Xie H., Jiang D., Zheng S. COL6A1 promotes metastasis and predicts poor prognosis in patients with pancreatic cancer. Int. J. Oncol. 2019;55:391–404. doi: 10.3892/ijo.2019.4825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hou T., Tong C., Kazobinka G., Zhang W., Huang X., Huang Y., Zhang Y. Expression of COL6A1 predicts prognosis in cervical cancer patients. Am. J. Transl. Res. 2016;8:2838–2844. [PMC free article] [PubMed] [Google Scholar]
- 31.Chiu K.H., Chang Y.H., Wu Y.S., Lee S.H., Liao P.C. Quantitative secretome analysis reveals that COL6A1 is a metastasis-associated protein using stacking gel-aided purification combined with iTRAQ labeling. J. Proteome Res. 2011;10:1110–1125. doi: 10.1021/pr1008724. [DOI] [PubMed] [Google Scholar]
- 32.Jian Z., Cheng T., Zhang Z., Raulefs S., Shi K., Steiger K., Maeritz N., Kleigrewe K., Hofmann T., Benitz S., Bruns P., Lamp D., Jastroch M., Akkan J., Jager C., Huang P., Nie S., Shen S., Zou X., Ceyhan G.O., Michalski C.W., Friess H., Kleeff J., Kong B. Glycemic Variability Promotes Both Local Invasion and Metastatic Colonization by Pancreatic Ductal Adenocarcinoma. Cell Mol. Gastroenterol. Hepatol. 2018;6:429–449. doi: 10.1016/j.jcmgh.2018.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Nguyen Y.H., Mills A.A., Stanbridge E.J. Assembly of the QM protein onto the 60S ribosomal subunit occurs in the cytoplasm. J. Cell. Biochem. 1998;68:281–285. [PubMed] [Google Scholar]
- 34.Shi J., Zhang L., Zhou D., Zhang J., Lin Q., Guan W., Zhang J., Ren W., Xu G. Biological Function of Ribosomal Protein L10 on Cell Behavior in Human Epithelial Ovarian Cancer. J. Cancer. 2018;9:745–756. doi: 10.7150/jca.21614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Aras S., Zaidi M.R. TAMeless traitors: macrophages in cancer progression and metastasis. Br. J. Cancer. 2017;117:1583–1591. doi: 10.1038/bjc.2017.356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Altinok G., Powell I.J., Che M., Hormont K., Sarkar F.H., Sakr W.A., Grignon D., Liao D.J. Reduction of QM protein expression correlates with tumor grade in prostatic adenocarcinoma. Prostate Cancer Prostatic Dis. 2006;9:77–82. doi: 10.1038/sj.pcan.4500848. [DOI] [PubMed] [Google Scholar]
- 37.Girardi T., Vereecke S., Sulima S.O., Khan Y., Fancello L., Briggs J.W., Schwab C., de Beeck J.O., Verbeeck J., Royaert J., Geerdens E., Vicente C., Bornschein S., Harrison C.J., Meijerink J.P., Cools J., Dinman J.D., Kampen K.R., De Keersmaecker K. The T-cell leukemia-associated ribosomal RPL10 R98S mutation enhances JAK-STAT signaling. Leukemia. 2018;32:809–819. doi: 10.1038/leu.2017.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Graziano F., Ruzzo A., Giacomini E., Ricciardi T., Aprile G., Loupakis F., Lorenzini P., Ongaro E., Zoratto F., Catalano V., Sarti D., Rulli E., Cremolini C., De Nictolis M., De Maglio G., Falcone A., Fiorentini G., Magnani M. Glycolysis gene expression analysis and selective metabolic advantage in the clinical progression of colorectal cancer. Pharmacogenomics J. 2017;17:258–264. doi: 10.1038/tpj.2016.13. [DOI] [PubMed] [Google Scholar]
- 39.Kim E., Jung S., Park W.S., Lee J.H., Shin R., Heo S.C., Choe E.K., Lee J.H., Kim K., Chai Y.J. Upregulation of SLC2A3 gene and prognosis in colorectal carcinoma: analysis of TCGA data. BMC Cancer. 2019;19:302. doi: 10.1186/s12885-019-5475-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Yao X., He Z., Qin C., Deng X., Bai L., Li G., Shi J. SLC2A3 promotes macrophage infiltration by glycolysis reprogramming in gastric cancer. Cancer Cell Int. 2020;20:503. doi: 10.1186/s12935-020-01599-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Napoli J.L. Cellular retinoid binding-proteins, CRBP, CRABP, FABP5: effects on retinoid metabolism, function and related diseases. Pharmacol. Ther. 2017;173:19–33. doi: 10.1016/j.pharmthera.2017.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Silvaroli J.A., Arne J.M., Chelstowska S., Kiser P.D., Banerjee S., Golczak M. Ligand binding induces conformational changes in human cellular retinol-binding protein 1 (CRBP1) revealed by atomic resolution crystal structures. J. Biol. Chem. 2016;291:8528–8540. doi: 10.1074/jbc.M116.714535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dobrotkova V., Chlapek P., Mazanek P., Sterba J., Veselska R. Traffic lights for retinoids in oncology: molecular markers of retinoid resistance and sensitivity and their use in the management of cancer differentiation therapy. BMC Cancer. 2018;18:1059. doi: 10.1186/s12885-018-4966-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Silvaroli J.A., Widjaja-Adhi M.A.K., Trischman T., Chelstowska S., Horwitz S., Banerjee S., Kiser P.D., Blaner W.S., Golczak M. Abnormal Cannabidiol Modulates Vitamin A Metabolism by Acting as a Competitive Inhibitor of CRBP1. ACS Chem. Biol. 2019;14:434–448. doi: 10.1021/acschembio.8b01070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chou A.P., Chowdhury R., Li S., Chen W., Kim A.J., Piccioni D.E., Selfridge J.M., Mody R.R., Chang S., Lalezari S., Lin J., Sanchez D.E., Wilson R.W., Garrett M.C., Harry B., Mottahedeh J., Nghiemphu P.L., Kornblum H.I., Mischel P.S., Prins R.M., Yong W.H., Cloughesy T., Nelson S.F., Liau L.M., Lai A. Identification of retinol binding protein 1 promoter hypermethylation in isocitrate dehydrogenase 1 and 2 mutant gliomas. J. Natl. Cancer Inst. 2012;104:1458–1469. doi: 10.1093/jnci/djs357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Toki K., Enokida H., Kawakami K., Chiyomaru T., Tatarano S., Yoshino H., Uchida Y., Kawahara K., Nishiyama K., Seki N., Nakagawa M. CpG hypermethylation of cellular retinol-binding protein 1 contributes to cell proliferation and migration in bladder cancer. Int. J. Oncol. 2010;37:1379–1388. doi: 10.3892/ijo_00000789. [DOI] [PubMed] [Google Scholar]
- 47.Peralta R., Valdivia A., Alvarado-Cabrero I., Gallegos F., Apresa T., Hernandez D., Mendoza M., Romero P., Paniagua L., Ibanez M., Cabrera L., Salcedo M. Correlation between expression of cellular retinol-binding protein 1 and its methylation status in larynx cancer. J. Clin. Pathol. 2012;65:46–50. doi: 10.1136/jclinpath-2011-200304. [DOI] [PubMed] [Google Scholar]
- 48.Gao L., Wang Q., Ren W., Zheng J., Li S., Dou Z., Kong X., Liang X., Zhi K. The RBP1-CKAP4 axis activates oncogenic autophagy and promotes cancer progression in oral squamous cell carcinoma. Cell Death. Dis. 2020;11:488. doi: 10.1038/s41419-020-2693-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Shvab A., Haase G., Ben-Shmuel A., Gavert N., Brabletz T., Dedhar S., Ben-Ze'ev A. Induction of the intestinal stem cell signature gene SMOC-2 is required for L1-mediated colon cancer progression. Oncogene. 2016;35:549–557. doi: 10.1038/onc.2015.127. [DOI] [PubMed] [Google Scholar]
- 50.Liu P., Lu J., Cardoso W.V., Vaziri C. The SPARC-related factor SMOC-2 promotes growth factor-induced cyclin D1 expression and DNA synthesis via integrin-linked kinase. Mol. Biol. Cell. 2008;19:248–261. doi: 10.1091/mbc.E07-05-0510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Su J.R., Kuai J.H., Li Y.Q. Smoc2 potentiates proliferation of hepatocellular carcinoma cells via promotion of cell cycle progression. World J. Gastroenterol. 2016;22:10053–10063. doi: 10.3748/wjg.v22.i45.10053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Brady J.J., Chuang C.H., Greenside P.G., Rogers Z.N., Murray C.W., Caswell D.R., Hartmann U., Connolly A.J., Sweet-Cordero E.A., Kundaje A., Winslow M.M. An Arntl2-driven secretome enables lung adenocarcinoma metastatic self-sufficiency. Cancer Cell. 2016;29:697–710. doi: 10.1016/j.ccell.2016.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Jang B.G., Kim H.S., Bae J.M., Kim W.H., Kim H.U., Kang G.H. SMOC2, an intestinal stem cell marker, is an independent prognostic marker associated with better survival in colorectal cancers. Sci. Rep. 2020;10:14591. doi: 10.1038/s41598-020-71643-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Esposito M., Mondal N., Greco T.M., Wei Y., Spadazzi C., Lin S.C., Zheng H., Cheung C., Magnani J.L., Lin S.H., Cristea I.M., Sackstein R., Kang Y. Bone vascular niche E-selectin induces mesenchymal-epithelial transition and Wnt activation in cancer cells to promote bone metastasis. Nat. Cell Biol. 2019;21:627–639. doi: 10.1038/s41556-019-0309-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Morisaki T., Yashiro M., Kakehashi A., Inagaki A., Kinoshita H., Fukuoka T., Kasashima H., Masuda G., Sakurai K., Kubo N., Muguruma K., Ohira M., Wanibuchi H., Hirakawa K. Comparative proteomics analysis of gastric cancer stem cells. PLoS One. 2014;9 doi: 10.1371/journal.pone.0110736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kuscu C., Evensen N., Kim D., Hu Y.J., Zucker S., Cao J. Transcriptional and epigenetic regulation of KIAA1199 gene expression in human breast cancer. PLoS One. 2012;7:e44661. doi: 10.1371/journal.pone.0044661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Terashima M., Fujita Y., Togashi Y., Sakai K., De Velasco M.A., Tomida S., Nishio K. KIAA1199 interacts with glycogen phosphorylase kinase beta-subunit (PHKB) to promote glycogen breakdown and cancer cell survival. Oncotarget. 2014;5:7040–7050. doi: 10.18632/oncotarget.2220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Sabates-Bellver J., Van der Flier L.G., de Palo M., Cattaneo E., Maake C., Rehrauer H., Laczko E., Kurowski M.A., Bujnicki J.M., Menigatti M., Luz J., Ranalli T.V., Gomes V., Pastorelli A., Faggiani R., Anti M., Jiricny J., Clevers H., Marra G. Transcriptome profile of human colorectal adenomas. Mol. Cancer Res. 2007;5:1263–1275. doi: 10.1158/1541-7786.MCR-07-0267. [DOI] [PubMed] [Google Scholar]
- 59.LaPointe L.C., Pedersen S.K., Dunne R., Brown G.S., Pimlott L., Gaur S., McEvoy A., Thomas M., Wattchow D., Molloy P.L., Young G.P. Discovery and validation of molecular biomarkers for colorectal adenomas and cancer with application to blood testing. PLoS One. 2012;7:e29059. doi: 10.1371/journal.pone.0029059. [DOI] [PMC free article] [PubMed] [Google Scholar]