Comparative proteomic analysis identifies exosomal Eps8 protein as a potential metastatic biomarker for pancreatic cancer

  • Authors:
    • Keiichi Ohshima
    • Keiichi Hatakeyama
    • Kaori Kanto
    • Tomomi Ide
    • Yuko Watanabe
    • Sachi Moromizato
    • Kanako Wakabayashi‑Nakao
    • Naoki Sakura
    • Ken Yamaguchi
    • Tohru Mochizuki
  • View Affiliations

  • Published online on: November 15, 2018     https://doi.org/10.3892/or.2018.6869
  • Pages: 1019-1034
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Exosomes are small vesicles found in extracellular environments including blood, urine, and cell culture medium. Their contents are cell‑type specific, and molecules embedded in exosomes can be useful fluid‑based clinical biomarkers. To identify proteins with metastatic marker potential, we conducted a comparative exosomal proteome analysis using human pancreatic cancer cell lines derived from metastasis, ascites, and primary tumors. Metastatic potential of cell lines was assessed by migratory and invasive activities. A pancreatic cancer cell line from metastasis (SU.86.86) revealed 23‑fold and 20‑fold increases in cell migratory and invasive activities, respectively, compared to the MIA PaCa‑2 cell line derived from primary tumor cells. Liquid chromatography‑mass spectrometry‑based proteome analysis and subsequent validation by immunoblot analysis revealed that epidermal growth factor receptor pathway substrate 8 (Eps8) was highly abundant in exosomes from metastasis‑derived SU.86.86 cells. Comparison of 12 pancreatic cancer cell lines derived from different stages of malignancy revealed a strong relationship between exosomal Eps8 protein levels and cell motile activities (migration: r=0.85, P=4.2x10‑4; invasion: r=0.60, P=3.2x10‑2). Conversely, relationships between intracellular Eps8 protein levels and cell motile activities were moderate (migration: r=0.65, P=2.0x10‑2; invasion: r=0.51, P=9.2x10‑2). It was therefore concluded that exosomal Eps8 protein levels were correlated with the migratory cell potential of human pancreatic cancer cells, indicating that exosomal Eps8 has the potential to be a metastatic biomarker for human pancreatic cancer.

Introduction

Exosomes are 30–100 nm membranous organelles that are released from cells into the extracellular microenvironment (1). Exosomes are vesicular carriers for intercellular communication, and they contain various signaling biomolecules, including proteins, metabolites, RNA, DNA, and lipids to target cells (24). Mass spectrometry and microarray technologies have been used to perform exosomal biomolecules profiling. These efforts have revealed that exosomal biomolecule composition varies depending on the cell type of origin (1,5). Since exosomes are found in biological fluids, including blood and urine, exosomal biomolecules with disease specificity are promising targets in liquid biopsies (6).

Cancer diagnosis at an early stage, before it has grown and spread to other organs by metastasis, is a prerequisite for successful treatment. Pancreatic cancer is one of the most deadly cancer forms and the third leading cause of cancer-related deaths in the United States (7), the EU (8) and Japan (http://ganjoho.jp/en/professional/statistics/brochure/2017_en.html). Early stage pancreatic cancer is difficult to diagnose since it is asymptomatic, making pancreatic cancer particularly challenging to treat and/or cure (9). In most cases, pancreatic cancer growth and metastasis have occurred by the time of diagnosis, leading to the poorest outcomes among the major types of cancer with a 5-year survival rate of <10% (7). While early diagnosis is essential for effective pancreatic cancer treatment and/or cure, there are currently no proven clinical tumor markers for the early stages of pancreatic cancer. However, recent developments in molecular profiling technologies have indicated that proteins and microRNAs identified in exosomes could be useful as fluid-based diagnostic and prognostic markers for pancreatic cancer (10,11).

Cell culture systems have been used for secretome analyses to identify the extracellular or exosomal proteins and microRNAs released into the medium (1214). Using cancer cells coupled with proteomics- or transcriptomics-based approaches, we have identified an abundance of polyadenylate binding protein 1 and let-7 family microRNAs in exosomes isolated from metastatic duodenal cancer cells (15,16). In the present study, we aimed to identify pancreatic cancer metastasis. We performed exosomal proteome analysis using pancreatic cancer cell lines derived from early (primary tumors), and late stages (ascites, and metastatic tumors) of tumor progression. Comparative analyses revealed that epidermal growth factor receptor pathway substrate 8 (Eps8) protein was abundant in exosomes derived from metastatic tumors and ascites and that the amount of exosomal Eps8 was quantitatively correlated with the in vitro cell migratory activity. These observations indicating that exosomal Eps8 is a predictive biomarker for pancreatic cancer metastasis.

Materials and methods

Cell culture

Cell lines used in the present study are listed in Table I. Cells were maintained in a humidified atmosphere (37°C, 5% CO2) in RPMI-1640 medium (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) supplemented with 2 mM L-glutamine (Nissui Pharmaceutical, Co., Ltd., Tokyo, Japan), 100 U/ml penicillin-100 mg/ml streptomycin, and 10% heat-inactivated (FBS) (all from Thermo Fisher Scientific, Inc., Waltham, MA, USA).

Table I.

List of human cancer cell lines used in the present study.

Table I.

List of human cancer cell lines used in the present study.

TissueCell lineHistologySupplierCatalogue no.Derivation
PancreasSU.86.86Ductal carcinomaATCCaCRL-1837Liver metastasis
CFPAC-1Ductal adenocarcinomaATCCCRL-1918Liver metastasis
KP-3Adenosquamous carcinomaJCRBbJCRB0178.0Liver metastasis
PK-45HCarcinomaRCBcRCB1973Liver metastasis, derived from the same patient as PK-45P
PK-1CarcinomaRCBRCB1972Liver metastasis, derived from the same patient as KLM-1
PK-8CarcinomaRCBRCB2700Liver metastasis
PK-59CarcinomaRCBRCB1901Liver metastasis
KLM-1CarcinomaRCBRCB2138Liver metastasis, derived from the same patient as PK-1
SW 1990AdenocarcinomaATCCCRL-2172Spleen metastasis
Hs 766TCarcinomaATCCHTB-134Lymph node metastasis
Capan-1AdenocarcinomaATCCHTB-79Liver metastasis
HuP-T3AdenocarcinomaECACCd93121055Ascites
HuP-T4AdenocarcinomaECACC93121056Ascites
HPAFIIAdenocarcinomaATCCCRL-1997Ascites
AsPC-1AdenocarcinomaATCCCRL-1682Ascites
MIA PaCa-2CarcinomaATCCCRL-1420Primary tumor
BxPC-3AdenocarcinomaATCCCRL-1687Primary tumor
PK-45PCarcinomaRCBRCB2141Primary tumor, derived from the same patient as PK-45H
Capan-2AdenocarcinomaATCCHTB-80Primary tumor
PSN1AdenocarcinomaECACC94060601Primary tumor
PANC-1Epithelioid carcinomaATCCCRL-1469Primary tumor
Panc 10.05AdenocarcinomaATCCCRL-2547Primary tumor
StomachKATOIIISignet ring cell carcinomaJCRBJCRB0611Pleural effusion and lymph node metastasis
SNU-1CarcinomaATCCCRL-5971Primary tumor
SNU-16CarcinomaATCCCRL-5974Ascites
NUGC-3Adenocarcinoma, poorly differentiatedJCRBJCRB0822Brachial muscle metastasis
NUGC-4Adenocarcinoma, poorly differentiated, signet ring cell carcinomaJCRBJCRB0834Lymph node metastasis
MKN45Adenocarcinoma, poorly differentiatedJCRBJCRB0254Primary tumor
MKN45PSubcloned from MKN45(38) Peritoneal metastasis in mice inoculated with MKN45
DuodenumAZ-521CarcinomaJCRBJCRB0061Primary tumor, identical genotype to HuTu 80 revealed by short Tandem repeat analysis
AZ-P7aSubcloned from AZ-521(39) Peritoneal metastasis in mice inoculated with MKN45
ColonCOLO 201Dukes' type D, colorectal adenocarcinomaJCRBJCRB0226Ascites, derived from the same patient as COLO 205
COLO 205Dukes' type D, colorectal adenocarcinomaATCCCCL-222Ascites, derived from the same patient as COLO 201
LoVoDukes' type C, grade IV, colorectal adenocarcinomaATCCCCL-229Left supraclavicular region metastasis
DLD-1Dukes' type C, colorectal adenocarcinomaECACC90102540Primary tumor
HT115CarcinomaECACC85061104Primary tumor
LungPC-10Squamous cell carcinomaIBLeIBL37013Primary tumor
LK-2Squamous cell carcinomaJCRBJCRB0829Primary tumor
EBC-1Squamous cell carcinomaJCRBJCRB0820Skin metastasis
NCI-H520Squamous cell carcinomaATCCHTB-182Primary tumor
SBC-1Small cell carcinomaJCRBJCRB0816Primary tumor
SBC-3Small cell carcinomaJCRBJCRB0818Bone marrow metastasis
NCI-H1581Large cell carcinomaATCCCRL-5878Primary tumor
NCI-H1650AdenocarcinomaATCCCRL-5883Pleural effusion metastasis
COR-L23Large cell carcinomaECACC92031919Pleural effusion metastasis
ThyroidTTMedullary carcinomaATCCCRL-1803Primary tumor
K1Papillary thyroid carcinomaECACC92030501Primary tumor
8505CUndifferentiated thyroid carcinomaJCRBJCRB0826Primary tumor
EsophagusKYSE30Squamous cell carcinomaJCRBJCRB0188Primary tumor
KYSE220Squamous cell carcinomaJCRBJCRB1086Primary tumor
EGJOE19AdenocarcinomaECACC96071721Primary tumor
ProstateLNCaPCarcinomaATCCCRL-1740Lymph node metastasis
DU145CarcinomaATCCHTB-81Brain metastasis
PC-3Grade IV, adenocarcinomaATCCCRL-1435Bone metastasis
BreastHs 578TCarcinomaECACC86082104Primary tumor
SK-BR-3AdenocarcinomaATCCHTB-30Pleural effusion metastasis
T-47DDuctal carcinomaATCCHTB-133Pleural effusion metastasis
Urinary bladder5637Grade II carcinomaATCCHTB-9Primary tumor
T24Transitional cell carcinomaATCCHTB-4Primary tumor
SCaBERSquamous cell carcinomaATCCHTB-3Primary tumor
LiverSK-HEP-1AdenocarcinomaECACC91091816Ascites
Hep G2 Hepatoblastoma(40)ATCCHB-8065Primary tumor

a American Type Culture Collection (Manassas, VA, USA)

b Japanese Collection of Research Bioresources (Osaka, Japan)

c RIKEN BioResourse Center (Ibaraki, Japan)

d European Collection of Cell Cultures (Salisbury, UK)

e Immuno-Biological Laboratories (Gunma, Japan). EGJ, esophagogastric junction.

Cell migration and invasion assays

Real-time cell analysis (RTCA) of in vitro cell migratory and invasive activities was performed using an xCELLigence RTCA DP instrument (Roche Diagnostics, Indianapolis, IN, USA) as previously described (15). Samples were analyzed in quadruplicate as technical replicates. Data analysis was performed using the RTCA software (version 1.2) supplied with the instrument.

Production and isolation of exosomes

Exosomes were isolated from the cell culture medium as previously described (15,16). Briefly, cells were cultured for 48 h at 37°C with 5% CO2 in complete RPMI-1640 medium containing 10% FBS depleted of contaminating microvesicles by centrifugation at 100,000 × g for 18 h. Culture medium (CM) was collected and centrifuged at 800 × g for 5 min and at an additional 2,000 × g for 10 min to remove detached cells. The supernatant was then filtered through a 0.1-µm pore polyethersulfone membrane filter (Thermo Fisher Scientific, Inc.) to remove cell debris and large vesicles, then concentrated using a Centricon Plus-70 with a 100,000-MW cut-off membrane (EMD Millipore; Billerica, MA, USA). Concentrated CM was ultracentrifuged at 100,000 × g for 2 h at 4°C using a 70Ti rotor (Beckman Coulter, Inc., Brea, CA, USA). Resultant pellets were resuspended in 6 ml phosphate-buffered saline (PBS) and ultracentrifuged at 100,000 × g for 1 h at 4°C using a 100Ti rotor (Beckman Coulter, Inc.).

Proteome analysis using mass spectrometry

Exosomal proteome analysis was performed by LC-MS/MS (liquid chromatography-mass spectrometry) as previously described (15,17). Proteins (200 µg) from isolated exosomes were dissolved in lysis buffer containing 7.5 M urea and 2.5 M thiourea (both from Sigma-Aldrich; Merck KGaA), 12.5% glycerol (Chemical Industries, Osaka, Japan), 50 mM Tris, 2.5% n-octyl-β-d-glucoside, 6.25 mM Tris(2-carboxyethyl)phosphine hydrochloride, and 1.25 mM protease inhibitor (all from Sigma-Aldrich; Merck KGaA) before being rotated at 4°C for 60 min. After centrifugation at 14,000 × g for 60 min at 4°C, the supernatant was fractionated using the Agilent 1200 HPLC system (Agilent Technologies, Inc., Santa Clara, CA, USA) with an Intrada WP-RP column (0.46×25 cm, 3-µm particle size and 30-nm pore size; Imtakt, Kyoto, Japan). Collected fractions were digested with trypsin (Promega Corp., Madison, WI, USA) and analyzed by LC-MS/MS using a nanoflow LC-ESI linear ion trap-TOF NanoFrontier L mass spectrometer (Hitachi High-Technologies, Tokyo, Japan). Raw LC-ESI data were converted to peak list files using NanoFrontier L Data Processing software (Hitachi High-Technologies). The peak list files were used for protein identification with the MASCOT MS/MS ion search (http://www.matrixscience.com) and X! Tandem software (http://www.thegpm.org). Upon peptide sequence annotation, the UniProtKB/Swiss-Prot database (version 2016_10; Homo sapiens; http://www.uniprot.org/statistics/Swiss-Prot) was used with the following parameters: enzyme, trypsin or none (when used with the home-made dataset only); maximum number of missed cleavage, 1; peptide tolerance, 0.2 Da; MS/MS tolerance, 0.2 Da; variable modification, oxidation of methionine; and peptide charge, (1+, 2+ and 3+). All identified proteins with MASCOT threshold scores < 95% confidence level and peptide numbers <2 were then removed from the protein list using Scaffold software (http://www.proteomesoftware.com/products/scaffold/).

Immunoblot analysis

Exosomes and cells were lysed with 7.5 M urea-based lysis buffer as described above. Protein concentrations were determined by the Bradford assay (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Proteins (5 or 10 µg) were subjected to 8% SDS-polyacrylamide gel electrophoresis (SDS-PAGE), and transferred onto an Immobilon-P polyvinylidene fluoride (PVDF) membrane (0.45-mm pore size; EMD Millipore). PVDF membranes were blocked for 1 h at room temperature in Tris-buffered saline (10 mM Tris-HCl, pH 7.5, 150 mM NaCl) containing 0.01% Tween-20 and 5% non-fat dried milk (Wako Pure Chemical Industries). Blocked membranes were then incubated overnight at 4°C with primary monoclonal antibodies (listed in Table II). Membranes were then incubated for 1 h at room temperature with anti-mouse IgG antibodies conjugated with horseradish peroxidase (Table II). Specific proteins were visualized using an ECL Plus western blotting detection system (GE Healthcare, Wauwatosa, WI, USA) and a Fujifilm Luminescent Image Analyzer LAS3000 (Fujifilm, Tokyo, Japan). The molecular weight of each protein was deduced using Precision Plus Protein All Blue Standards (Bio-Rad Laboratories, Inc.).

Table II.

List of antibodies used for western blot analysis.

Table II.

List of antibodies used for western blot analysis.

ProteinClonalityHostSupplierCatalogue no.DilutionDilution of 2nd Ab
Eps8MonoclonalMouseBD Biosciencesa6101441:5,000 1:10,000d
α-tubulinMonoclonalMouseEMD MilliporebCP061:5,000 1:10,000d
GAPDHMonoclonalMouseSCBTcsc-365621:1,000 1:5,000d

a BD Biosciences, San Jose, CA, USA

b EMD Millipore, Billerica, MA, USA

c Santa Cruz Biotechnology, Inc., Dallas, Texas, USA; dSecondary antibody conjugated with horseradish peroxidase: Goat anti-mouse IgG antibody (cat. no. 115-035-062, Jackson ImmunoResearch Laboratories). Eps8, epidermal growth factor receptor pathway substrate 8; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.

RNA isolation and quantitative RT-PCR analysis

Cells were cultured for 48 h, and total RNA was extracted using the miRNeasy Mini Kit (Qiagen, Hilden, Germany) as previously described (15). RNA samples were quantified with a NanoDrop spectrophotometer (Thermo Fisher Scientific, Inc.) and assessed using an Agilent 2100 Bioanalyzer and an RNA 6000 Nano Total RNA kit (both from Agilent Technologies, Inc.). Quantitative mRNA levels were determined using real-time RT-PCR using the Applied Biosystems 7900 HT Sequence Detection System, a TaqMan Gene Expression Assay for human EPS8 (assay ID Hs00610286_m1), and a Eukaryotic 18S rRNA Endogenous Control (Applied Biosystems; Thermo Fisher Scientific, Inc.). Only the probe sequence for EPS8 (TTGGATGAAAGCCAGAGCAGAGTGG) was provided by the manufacturer. The probes of EPS8 and 18S rRNA were labelled with FAM and VIC dyes, respectively. cDNA was generated using 100 ng of total RNA, and a High Capacity cDNA Reverse Transcription kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). RT-PCR was carried out in a total volume of 20 µl containing 100 ng of cDNA, TaqMan Fast Advanced Master Mix (Applied Biosystems), and the respective TaqMan target gene reagents. The amplification conditions were 95°C for 20 sec followed by 40 cycles of 95°C for 1 sec and 60°C for 20 sec. Samples were analyzed in triplicate as technical replicates. The EPS8 mRNA levels were defined from the cycle threshold (Ct), using the comparative Ct method (18), and each sample was normalized by comparison to 18S rRNA levels. The fold change of EPS8 mRNA levels in each cell line was determined using SU.86.86 cell EPS8 mRNA levels as a reference.

Statistical analysis

Student's t-test for comparison of cell motility between SU.86.86 and MIA PaCa-2 cell lines, the Pearson correlation coefficient to compare two variables in five analyses (exosomal Eps8 protein, intracellular Eps8 protein, intracellular Eps8 mRNA, migration, and invasion), and Multiple t-tests with Bonferroni-correction for comparison of three different cell origins of metastasis, ascites and primary tumors were used. P-values <0.05 were considered to indicate a statistically significant difference.

Results

In vitro migratory and invasive activities of SU.86.86 and MIA PaCa-2 cells

SU.86.86 cells were derived from a liver metastasis of a pancreatic ductal carcinoma (19). MIA PaCa-2 cells were derived from a primary pancreatic adenocarcinoma (20). Before proteome analysis, we first performed in vitro cell migration and invasion assays to evaluate if the two cell lines exhibited differences in metastatic-potential. The impedance-based RTCA has shown a strong correlation with the conventional Boyden chamber Transwell endpoint assay (15,21). Using the RTCA assay system, SU.86.86 cells had 23-fold greater cell migratory activity than did MIA PaCa-2 cells (Fig. 1A). Additionally, using a Matrigel barrier, SU.86.86 cells were found to be 20-times more invasive than MIA PaCa-2 cells (Fig. 1B). Collectively, these results indicated that the in vitro cell migratory and invasive behaviors of SU.86.86 and MIA PaCa-2 cells were correlated with their metastatic and primary tumor cell origins, respectively.

Exosomal proteome profiles of SU.86.86 and MIA PaCa-2 cells

The proteome profiles of SU.86.86 and MIA PaCa-2 cell-derived exosomes were analyzed using LC-MS/MS. After 48 h of cell growth, exosomes were isolated from culture media by a series of filtration and ultracentrifugation steps as previously described (15,16). Proteome data processing identified a total of 133 proteins from exosomes derived from both cell lines (Fig. 2A, Table III). Among them, 31 proteins were identified in the exosomes of both cell types. A total of 101 proteins were uniquely identified in SU.86.86 cell-derived exosomes, and a single unique protein, histone H2A type 2-B (H2A2B), was identified in MIA PaCa-2 cell-derived exosomes.

Table III.

List of proteins identified in exosomes derived from SU.86.86 and MIA PaCa-2 cells.

Table III.

List of proteins identified in exosomes derived from SU.86.86 and MIA PaCa-2 cells.

Number of matched peptides

Accession no. UniProtKB/Swiss-Prot entry nameProtein nameM.W. (kDa)SU.86.86MIA PaCa-2
Q7Z406MYH14_HUMANMyosin-14228290
Q12929EPS8_HUMANEpidermal growth factor receptor pathway substrate 892140
Q14764MVP_HUMANMajor vault protein99140
P05121PAI1_HUMANPlasminogen activator inhibitor 145100
P16104H2AX_HUMANHistone H2AX1580
P215895NTD_HUMAN 5′-nucleotidase6370
P84243H33_HUMANHistone H3.31570
Q9GZM7TINAL_HUMANTubulointerstitial nephritis antigen-like5270
P01023A2MG_HUMAN Alpha-2-macroglobulin16360
P80188NGAL_HUMANNeutrophil gelatinase-associated lipocalin2360
P23396RS3_HUMAN40S ribosomal protein S32750
Q9UQB8BAIP2_HUMANBrain-specific angiogenesis inhibitor 1-associated protein 26150
Q9UHR4BI2L1_HUMANBrain-specific angiogenesis inhibitor 1-associated protein 2-like protein 15750
P02458CO2A1_HUMANCollagen alpha-1 (II) chain14250
P23142FBLN1_HUMANFibulin-17750
P13647K2C5_HUMANKeratin, type II cytoskeletal 56250
P05787K2C8_HUMANKeratin, type II cytoskeletal 85450
P15880RS2_HUMAN40S ribosomal protein S23140
P08865RSSA_HUMAN40S ribosomal protein SA3340
P09525ANXA4_HUMANAnnexin A43640
P08133ANXA6_HUMANAnnexin A67640
O95994AGR2_HUMANAnterior gradient protein 2 homolog2040
O15335CHAD_HUMANChondroadherin4040
P12109CO6A1_HUMANCollagen alpha-1 (VI) chain10940
P39060COIA1_HUMANCollagen alpha-1 (XVIII) chain17840
P08238HS90B_HUMANHeat shock protein HSP 90-beta8340
Q16270IBP7_HUMANInsulin-like growth factor-binding protein 72940
Q08431MFGM_HUMANLactadherin4340
Q13753LAMC2_HUMANLaminin subunit gamma-213140
Q9NRN5OLFL3_HUMANOlfactomedin-like protein 34640
O00391QSOX1_HUMANSulfhydryl oxidase 18340
P00750TPA_HUMANTissue-type plasminogen activator6340
P29144TPP2_HUMAN Tripeptidyl-peptidase 213840
Q9H9H4VP37B_HUMANVacuolar protein sorting-associated protein 37B3140
P62241RS8_HUMAN40S ribosomal protein S82430
P46781RS9_HUMAN40S ribosomal protein S92330
Q07020RL18_HUMAN60S ribosomal protein L182230
P12429ANXA3_HUMANAnnexin A33630
O75531BAF_HUMAN Barrier-to-autointegration factor1030
P02462CO4A1_HUMANCollagen alpha-1(IV) chain16130
Q86YQ8CPNE8_HUMANCopine-86330
P68871HBB_HUMANHemoglobin subunit beta1630
P16403H12_HUMANHistone H1.22130
Q71UI9H2AV_HUMANHistone H2A.V1430
Q6ZNF0ACP7_HUMANAcid phosphatase type 75030
P02533K1C14_HUMANKeratin, type I cytoskeletal 145230
P05783K1C18_HUMANKeratin, type I cytoskeletal 184830
P04259K2C6B_HUMANKeratin, type II cytoskeletal 6B6030
Q16787LAMA3_HUMANLaminin subunit alpha-336730
P60660MYL6_HUMANMyosin light polypeptide 61730
P19105ML12A_HUMANMyosin regulatory light chain 12A2030
P19338NUCL_HUMANNucleolin7730
Q8WUM4PDC6I_HUMANProgrammed cell death 6-interacting protein9630
P00734THRB_HUMANProthrombin7030
P78371TCPB_HUMANT-complex protein 1 subunit beta5730
P68371TBB4B_HUMANTubulin beta-4B chain5030
P62195PRS8_HUMAN26S protease regulatory subunit 84620
P62847RS24_HUMAN40S ribosomal protein S241520
P50914RL14_HUMAN60S ribosomal protein L142320
P61313RL15_HUMAN60S ribosomal protein L152420
P18124RL7_HUMAN60S ribosomal protein L72920
P62424RL7A_HUMAN60S ribosomal protein L7a3020
P53999TCP4_HUMANActivated RNA polymerase II transcriptional coactivator p151420
P08758ANXA5_HUMANAnnexin A53620
P98160PGBM_HUMANBasement membrane-specific heparan sulfate proteoglycan core protein46920
P02749APOH_HUMANBeta-2-glycoprotein 13820
P62158CALM_HUMANCalmodulin1720
P15169CBPN_HUMANCarboxypeptidase N catalytic chain5220
P49747COMP_HUMANCartilage oligomeric matrix protein8320
Q9NZZ3CHMP5_HUMANCharged multivesicular body protein 52520
P08123CO1A2_HUMANCollagen alpha-2(I) chain12920
P01031CO5_HUMANComplement C518820
O75367H2AY_HUMANCore histone macro-H2A.14020
P26641EF1G_HUMANElongation factor 1-gamma5020
Q9H6S3ES8L2_HUMANEpidermal growth factor receptor kinase substrate 8-like protein 28120
P09972ALDOC_HUMAN Fructose-bisphosphate aldolase C3920
P23229ITA6_HUMANIntegrin alpha-612720
Q7Z794K2C1B_HUMANKeratin, type II cytoskeletal 1b6220
P19013K2C4_HUMANKeratin, type II cytoskeletal 45720
P02538K2C6A_HUMANKeratin, type II cytoskeletal 6A6020
P08729K2C7_HUMANKeratin, type II cytoskeletal 75120
P51884LUM_HUMANLumican3820
Q14112NID2_HUMANNidogen-215120
P30101PDIA3_HUMANProtein disulfide-isomerase A35720
P60903S10AA_HUMANProtein S100-A101120
P31949S10AB_HUMANProtein S100-A111220
P21980TGM2_HUMANProtein-glutamine gamma-glutamyltransferase 27720
Q92954PRG4_HUMANProteoglycan 415120
P50454SERPH_HUMANSerpin H14620
P24821TENA_HUMANTenascin24120
P35908K22E_HUMANKeratin, type II cytoskeletal 2 epidermal65161
P07437TBB5_HUMANTubulin beta chain50151
P0C0L4CO4A_HUMANComplement C4-A19391
P35580MYH10_HUMANMyosin-1022991
P68363TBA1B_HUMANTubulin alpha-1B chain5071
P36955PEDF_HUMANPigment epithelium-derived factor4661
P16401H15_HUMANHistone H1.52351
P23284PPIB_HUMANPeptidyl-prolyl cis-trans isomerase B2451
Q9H444CHM4B_HUMANCharged multivesicular body protein 4b2541
P05452TETN_HUMANTetranectin2341
P62854RS26_HUMAN40S ribosomal protein S261321
P35579MYH9_HUMANMyosin-92277811
P02751FINC_HUMANFibronectin263425
P04264K2C1_HUMANKeratin, type II cytoskeletal 166299
P07996TSP1_HUMAN Thrombospondin-1129272
P13645K1C10_HUMANKeratin, type I cytoskeletal 1059264
P35527K1C9_HUMANKeratin, type I cytoskeletal 962192
P01024CO3_HUMANComplement C3187172
P60709ACTB_HUMANActin, cytoplasmic 142154
P07355ANXA2_HUMANAnnexin A239142
P11142HSP7C_HUMANHeat shock cognate 71 kDa protein711313
P15311EZRI_HUMANEzrin69136
P62805H4_HUMANHistone H411106
P04406G3P_HUMAN Glyceraldehyde-3-phosphate dehydrogenase36103
P06396GELS_HUMANGelsolin86102
P12259FA5_HUMANCoagulation factor V25294
P14618KPYM_HUMANPyruvate kinase PKM5893
O00560SDCB1_HUMANSyntenin-13292
P33778H2B1B_HUMANHistone H2B type 1-B1483
P04083ANXA1_HUMANAnnexin A13982
P69905HBA_HUMANHemoglobin subunit alpha1563
P02768ALBU_HUMANSerum albumin6954
P68104EF1A1_HUMANElongation factor 1-alpha 15053
P04075ALDOA_HUMAN Fructose-bisphosphate aldolase A3953
P06733ENOA_HUMANAlpha-enolase4742
P03956MMP1_HUMANInterstitial collagenase5442
P02788TRFL_HUMAN Lactotransferrin7825
P62249RS16_HUMAN40S ribosomal protein S161624
P10643CO7_HUMANComplement component C79423
P02748CO9_HUMANComplement component C96323
Q71DI3H32_HUMANHistone H3.21522
Q06830PRDX1_HUMAN Peroxiredoxin-12222
Q8IUE6H2A2B_HUMANHistone H2A type 2-B1403
Identification of Eps8 in SU.86.86 cell-derived exosomes

To identify the SU.86.86 cell-specific exosomal proteins, we compared the 101 SU.86.86 cell-specific proteins with those that had been previously identified in exosomes derived from human duodenal cancer cell lines AZ-521 and AZ-P7a (Fig. 2A) (15). This comparison identified 82 proteins that were unique to SU.86.86 cell-derived exosomes (Table IV). Of the 82 proteins unique to SU.86.86, Eps8 revealed relatively high MS/MS values, including the number of matched peptides, the rate of sequence coverage, and the total spectral count. Furthermore, the Eps8 expression was elevated in pancreatic cancer cells derived from ascites and metastasis (22). Therefore, we chose to validate the presence of Eps8 in exosomes by western blot analysis.

Table IV.

List of proteins specifically identified in exosomes derived from SU.86.86 compared to other cancer cell lines.

Table IV.

List of proteins specifically identified in exosomes derived from SU.86.86 compared to other cancer cell lines.

Accession no. UniProtKB/Swiss-Prot entry nameProtein nameM.W. (kDa)Number of matched peptidesSequence coverage (%)Total spectral count
Q7Z406MYH14_HUMANMyosin-14228291971
Q12929EPS8_HUMANEpidermal growth factor receptor pathway substrate 892142932
Q14764MVP_HUMANMajor vault protein99142527
P05121PAI1_HUMANPlasminogen activator inhibitor 145103128
P16104H2AX_HUMANHistone H2AX1585246
P84243H33_HUMANHistone H3.31575228
Q9GZM7TINAL_HUMANTubulointerstitial nephritis antigen-like5272118
P215895NTD_HUMAN 5′-nucleotidase6371715
P80188NGAL_HUMANNeutrophil gelatinase-associated lipocalin2364424
P36955PEDF_HUMANPigment epithelium-derived factor4661713
P23284PPIB_HUMANPeptidyl-prolyl cis-trans isomerase B2452914
P13647K2C5_HUMANKeratin, type II cytoskeletal 56251722
P05787K2C8_HUMANKeratin, type II cytoskeletal 85451615
P16401H15_HUMANHistone H1.52351613
Q9UQB8BAIP2_HUMANBrain-specific angiogenesis inhibitor 1-associated protein 2615156
Q9UHR4BI2L1_HUMANBrain-specific angiogenesis inhibitor 1-associated protein 2-like protein 15751416
P23142FBLN1_HUMANFibulin-1775613
P02458CO2A1_HUMANCollagen alpha-1 (II) chain1425413
O95994AGR2_HUMANAnterior gradient protein 2 homolog204305
Q9H444CHM4B_HUMANCharged multivesicular body protein 4b254227
Q9H9H4VP37B_HUMANVacuolar protein sorting-associated protein 37B314224
P08865RSSA_HUMAN40S ribosomal protein SA334216
O15335CHAD_HUMANChondroadherin404168
P09525ANXA4_HUMANAnnexin A4364165
Q16270IBP7_HUMANInsulin-like growth factor-binding protein 72941511
Q08431MFGM_HUMANLactadherin434134
P00750TPA_HUMANTissue-type plasminogen activator634118
P08133ANXA6_HUMANAnnexin A676494
O00391QSOX1_HUMANSulfhydryl oxidase 183467
P12109CO6A1_HUMANCollagen alpha-1 (VI) chain109457
Q13753LAMC2_HUMANLaminin subunit gamma-2131458
P29144TPP2_HUMAN Tripeptidyl-peptidase 2138454
P39060COIA1_HUMANCollagen alpha-1 (XVIII) chain178446
O75531BAF_HUMAN Barrier-to-autointegration factor103435
P68371TBB4B_HUMANTubulin beta-4B chain5034145
Q71UI9H2AV_HUMANHistone H2A.V1433125
P19105ML12A_HUMANMyosin regulatory light chain 12A203273
P60660MYL6_HUMANMyosin light polypeptide 6173247
P02533K1C14_HUMANKeratin, type I cytoskeletal 145231840
P12429ANXA3_HUMANAnnexin A3363143
P05783K1C18_HUMANKeratin, type I cytoskeletal 18483137
P04259K2C6B_HUMANKeratin, type II cytoskeletal 6B6031238
P46781RS9_HUMAN40S ribosomal protein S9233126
Q6ZNF0ACP7_HUMANAcid phosphatase type 7503125
P78371TCPB_HUMANT-complex protein 1 subunit beta573113
Q86YQ8CPNE8_HUMANCopine-863383
P19338NUCL_HUMANNucleolin77369
Q16787LAMA3_HUMANLaminin subunit alpha-3367333
P02462CO4A1_HUMANCollagen alpha-1(IV) chain161325
P60903S10AA_HUMANProtein S100-A10112354
P62158CALM_HUMANCalmodulin172265
P31949S10AB_HUMANProtein S100-A11122244
P62854RS26_HUMAN40S ribosomal protein S261322112
P62847RS24_HUMAN40S ribosomal protein S24152207
Q9NZZ3CHMP5_HUMANCharged multivesicular body protein 5252182
P02538K2C6A_HUMANKeratin, type II cytoskeletal 6A6021325
P09972ALDOC_HUMAN Fructose-bisphosphate aldolase C392134
O75367H2AY_HUMANCore histone macro-H2A.1402126
P53999TCP4_HUMANActivated RNA polymerase II transcriptional coactivator p15142124
P18124RL7_HUMAN60S ribosomal protein L7292103
P08758ANXA5_HUMANAnnexin A5362103
P19013K2C4_HUMANKeratin, type II cytoskeletal 4572813
P62424RL7A_HUMAN60S ribosomal protein L7a30276
P08729K2C7_HUMANKeratin, type II cytoskeletal 7512711
P62195PRS8_HUMAN26S protease regulatory subunit 846272
P50454SERPH_HUMANSerpin H146272
P51884LUM_HUMANLumican38274
Q7Z794K2C1B_HUMANKeratin, type II cytoskeletal 1b622721
P15169CBPN_HUMANCarboxypeptidase N catalytic chain52262
P26641EF1G_HUMANElongation factor 1-gamma50264
P02749APOH_HUMANBeta-2-glycoprotein 138254
P30101PDIA3_HUMANProtein disulfide-isomerase A357253
P21980TGM2_HUMANProtein-glutamine gamma-glutamyltransferase 277245
P49747COMP_HUMANCartilage oligomeric matrix protein83244
Q9H6S3ES8L2_HUMANEpidermal growth factor receptor kinase substrate 8-like protein 281243
P23229ITA6_HUMANIntegrin alpha-6127232
Q14112NID2_HUMANNidogen-2151223
Q92954PRG4_HUMANProteoglycan 41512210
P08123CO1A2_HUMANCollagen alpha-2(I) chain129223
P01031CO5_HUMANComplement C5188213
P24821TENA_HUMANTenascin241212
P98160PGBM_HUMANBasement membrane-specific heparan sulfate proteoglycan core protein469203

Western blot analyses revealed that the Eps8 protein was abundant in SU.86.86 cell-derived exosomes, while no immunoreactive Eps8 signals were detected in MIA PaCa-2 cell-derived exosomes (Fig. 2B). Furthermore, intracellular Eps8 expression levels were much higher in SU.86.86 cells than in MIA PaCa-2 cells, indicating a positive correlation with tumor malignancy, as previously reported (22). Comparison of the house-keeping proteins in the exosomes of both cell types revealed variation in glyceraldehyde 3-phosphate dehydrogenase (GAPDH) levels and no immunoreactive signals for α-tubulin, making them unsuitable for normalizing exosomal protein levels previously described (15).

Exosomal Eps8 is abundant in metastasis- and ascites-derived pancreatic cancer cells

Eps8 was specifically detected in exosomes from metastatic-derived SU.86.86 cells. Therefore, we assessed exosomal Eps8 protein levels in other pancreatic cancer cell lines. Western blot analysis revealed positive immunoreactive Eps8 signals in exosomes from metastasis-derived pancreatic cancer cell lines, including CFPAC-1, KP-3, PK-45H, PK-8 and Capan-1 (Fig. 3A). Additionally, positive immunoreactive Eps8 signals were observed in exosomes from ascites-derived pancreatic cancer cell lines, including HuP-T3, HuP-T4 and AsPC-1. In contrast, Capan-2 was the only primary tumor cell line that exhibited Eps8 immunoreactivity. Densitometric analysis, using relative amounts of exosomal Eps8 protein, was used to quantify the Eps8 immunoreactivities observed (Fig. 4). The level of Eps8 immunoreactivity in the exosomes of different cell lines was assessed relative to that observed in SU.86.86 cell-derived exosomes, which was given a value of 1.0. The relative Eps8 immunoreactivity was 0.76, 0.15 and 0.17 in metastatic cell lines PK-45H, CFPAC-1 and KP-3, respectively. In ascites-derived cell lines HuP-T3, HuP-T4, and AsPC-1, and the Capan-2 primary tumor cell line, the relative Eps8 immunoreactivity was 0.49, 0.21, 0.15 and 0.55, respectively. Intracellular Eps8 levels varied among the cell lines, particularly those derived from metastasis (PK-1, PK-8, PK-59 and KLM-1) and primary tumors (MIA PaCa-2, BxPC-3, PANC-1 and Panc 10.05). Except for PK-8, there was either no Eps8 immunoreactivity, or less Eps8 immunoreactivity, relative to that observed intracellularly, observed in the exosomes of these cells. Also, distinct Eps8 immunoreactivity was observed in NUGC-4 and MKN45P stomach cancer cell lines and the LoVo colon cancer cell line (Fig. 3B). Furthermore, cells with relatively high intracellular Eps8 protein levels expressed greater amounts of EPS8 mRNA (Fig. 4). However, Eps8 protein and mRNA expression levels did not correlate with the amount of Eps8 in exosomes. Collectively, these results revealed that, particularly in pancreatic cancer cells derived from metastasis and ascites, Eps8 was secreted into the extracellular environment via exosomes.

In vitro migratory and invasive activities of pancreatic cancer cell lines

It was revealed that Eps8 protein is present in the exosomes of several pancreatic cancer cell lines in addition to SU.86.86. Therefore, we evaluated the in vitro cell migratory and invasive activities in 12 pancreatic cancer cell lines. The highest levels of migratory and invasive activities were observed in SU.86.86 cells, and these were set at a value of 1 to allow for comparison (Fig. 5). The metastatic PK-45H cell line, with the relative exosomal Eps8 protein level of 0.76, revealed relatively high levels of cell migratory (0.64) and invasive activities (0.56). Additionally, in ascites-derived HuP-T4 cells, with the relative exosomal Eps8 protein level of 0.21, relatively high levels of cell migratory (0.36) and invasive activities (0.84) were observed. However, no cell motility was detected in stomach cancer-derived NUGC-4 cells and colon cancer-derived LoVo cells, which exhibited moderate levels of exosomal Eps8 immunoreactivities (Fig. 3).

Integrative comparison of the data revealed that similar to SU.86.86 cells, PK-45H cells consistently had the highest levels of in vitro cell migratory and invasive activities, exosomal and intracellular Eps8 protein, and EPS8 mRNA expression (Fig. 6A and B). Furthermore, using the Pearson correlation coefficient, we identified that exosomal Eps8 levels were significantly correlated with migratory cell levels (r=0.85, P=4.2×10−4) (Fig. 6C). Therefore, we proposed that exosomal Eps8 protein level is indicative of metastatic potential in human pancreatic cancer cells.

Discussion

The present study revealed abundant levels of Eps8 protein in exosomes derived from pancreatic cancer cell lines. Furthermore, it was revealed that exosomal Eps8 levels were significantly correlated with migratory cell potential (Fig. 6C). Eps8 was initially identified as a substrate for the epidermal growth factor (EGF) receptor that enhances EGF-dependent mitogenic signals (23,24). Overexpression of Eps8 has been revealed to promote cellular proliferation and/or migration in various tumor types, including breast cancer (25), malignant glioma (26,27), pituitary tumors (28), oral squamous cell carcinoma (29), and cervical cancer (30). In Eps8-mediated tumorigenesis and proliferation, stimulated EGFR results in the activation of downstream pathways, including Eps8/Ras/MAPK, Eps8/Akt/FoxM1 and Eps8/mTOR/STAT3 were revealed (31).

Eps8 expression was enhanced in pancreatic cancer at both protein and mRNA levels (22), and Eps8 upregulation was immunohistochemically detected in 72% of paraffin-embedded clinical specimens (32). Welsch et al demonstrated that Eps8 expression levels were correlated with the degree of malignancy in pancreatic cancer cell lines (22). They found low levels of Eps8 expression in cell lines from primary pancreatic cancers (MIAPaCa-2, BxPC-3, and PANC-1), moderate Eps8 expression levels in cell lines from metastasis (SU.86.86 and Capan-1), and high Eps8 expression level in a cell line from malignant ascites (AsPC-1) (22). Additionally, their Eps8 expression levels were positively correlated with migratory potential (BxPC-3 < PANC-1<Capan-1<AsPC-1). In the present study, a moderate correlation between cell migratory capacity and intracellular Eps8 protein expression levels (r=0.65, P=2.0×10−2) was revealed but not between cell migratory capacity and intracellular EPS8 mRNA expression levels (r=0.44, P=1.5×10−1) (Fig. 6C). However, we identified a significant correlation between exosomal Eps8 protein levels and migratory cell capacity (r=0.85, P=4.2×10−4). The pancreatic cancer cell lines that exhibited relatively high exosomal Eps8 protein levels were SU.86.86 and PK-45H from metastasis, HuP-T3 from ascites, and Capan-2 from primary tumor cells (Figs. 3A and 6B). Despite originating from primary tumor cells, Capan-2 exhibited moderate cell migratory activity (Figs. 5A and 6B). These results were consistent with those revealing that Capan-2 possessed metastatic potential to the liver after being inoculated into nude mice (33). AsPC-1 cells, derived from ascites, have been previously revealed to have intracellular Eps8 protein and mRNA expression levels and migratory cell potential greater than those of metastasis-derived SU.86.86 cells (22). In our study, AsPC-1 cells had lower levels of intracellular and exosomal Eps8 protein and migratory cell levels than did SU.86.86 cells (Figs. 3A, 5A and 6B). We assume that these different results for AsPC-1 cells are at least due to culture conditions for maintenance of the cell line and preparation of samples. Additionally, among three groups of pancreatic cancer cell lines with different degrees of malignancy, intracellular Eps8 expression levels were significantly higher in cells from metastasis than in those from ascites (P=0.01628, the Bonferroni-corrected threshold for multiple t-tests =0.01667, α=0.05). Eps8 expression in ascites-derived cell lines did not significantly differ from that of primary tumor-derived cell lines (P=0.634). Eps8 expression levels in metastasis-derived cell lines were significantly higher than in primary tumor-derived cell lines (P=0.01662). We compared the levels of exosomal Eps8 protein, intracellular Eps8 mRNA, migratory and invasive activities of metastasis-, ascites-, and primary tumor-derived cell line groups and found no significant differences. Therefore, the present study indicated that there is a strong relationship between exosomal Eps8 protein level and migratory cell potential.

Exosomes play an essential role in tumor metastasis (34). Eps8 is involved in metastasis, and inhibiting Eps8 expression results in decreased levels of cell motility (26,30,32). The Eps8 protein localizes to lysosomes via the late endosomes, which function as a pre-degenerative compartment (35). The late endosomes also function as a recycling compartment, leading to extracellular secretion via fusion with the plasma membrane (1,36,37). Collectively, with the present results, it may be inferred that Eps8 protein is recruited to late endosomes, leading to either inclusion in lysosomes or extracellular secretion. Our results revealed that in pancreatic cancer cell lines with high migratory potential, Eps8 protein abundance in exosomes occurs through extracellular secretion. These observations indicated that exosomal Eps8 has a potential to be a metastatic biomarker for pancreatic cancer. Further studies need to be performed using clinical samples to validate this hypothesis. For validation, it is conceivable to use an ELISA system, which can easily detect secreted proteins in serum or plasma blood samples. For proteins embedded in exosomes, such as Eps8, it is challenging to develop an ELISA system, since detergents that may affect the assay are used for exosome lysis during the sample preparation.

Acknowledgements

Not applicable.

Funding

The present study was supported by the JSPS KAKENHI grant nos. JP26430150 and JP16K10523 to K.O.

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

KO made substantial contributions to the design of the study, drafting of the manuscript, cell culture and the preparation of the exosomes. KH, KWN and NS were responsible for the proteome analysis using LC-MS/MS. KK and TI performed the western blotting experiments. KK, YW and SM were involved in RNA preparation and RT-PCR analysis. KY and TM supervised the study and analyzed the data. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

This article contains no studies with human participants performed by any of the authors.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CM

culture medium

Ct

cycle threshold

EGF

epidermal growth factor

Eps8

epidermal growth factor receptor pathway substrate 8

FBS

fetal bovine serum

GAPDH

glyceraldehyde 3-phosphate dehydrogenase

LC-MS/MS

liquid chromatography-mass spectrometry

RTCA

real-time cell analysis

SDS-PAGE

SDS-polyacrylamide gel electrophoresis

References

1 

Colombo M, Raposo G and Théry C: Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 30:255–289. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Greening DW, Xu R, Gopal SK, Rai A and Simpson RJ: Proteomic insights into extracellular vesicle biology-defining exosomes and shed microvesicles. Expert Rev Proteomics. 14:69–95. 2007. View Article : Google Scholar

3 

Kalluri R: The biology and function of exosomes in cancer. J Clin Invest. 126:1208–1215. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Soung YH, Ford S, Zhang V and Chung J: Exosomes in cancer diagnostics. Cancers. 9:82017. View Article : Google Scholar :

5 

Choi DS, Kim DK, Kim YK and Gho YS: Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics. 13:1554–1571. 2013. View Article : Google Scholar : PubMed/NCBI

6 

EL Andaloussi S, Mäger I, Breakefield XO and Wood MJ: Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 12:347–357. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Ferlay J, Partensky C and Bray F: More deaths from pancreatic cancer than breast cancer in the EU by 2017. Acta Oncol. 55:1158–1160. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Hanada K, Okazaki A, Hirano N, Izumi Y, Teraoka Y, Ikemoto J, Kanemitsu K, Hino F, Fukuda T and Yonehara S: Diagnostic strategies for early pancreatic cancer. J Gastroenterol. 50:147–154. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Nuzhat Z, Kinhal V, Sharma S, Rice GE, Joshi V and Salomon C: Tumour-derived exosomes as a signature of pancreatic cancer-liquid biopsies as indicators of tumour progression. Oncotarget. 8:17279–17291. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Le Large TYS, Bijlsma MF, Kazemier G, van Laarhoven HWM, Giovannetti E and Jimenez CR: Key biological processes driving metastatic spread of pancreatic cancer as identified by multi-omics studies. Semin Cancer Biol. 44:153–169. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Sasaki K, Sato K, Akiyama Y, Yanagihara K, Oka M and Yamaguchi K: Peptidomics-based approach reveals the secretion of the 29-residue COOH-terminal fragment of the putative tumor suppressor protein DMBT1 from pancreatic adenocarcinoma cell lines. Cancer Res. 62:4894–4898. 2002.PubMed/NCBI

13 

Ogura S, Kaneko K, Miyajima S, Ohshima K, Yamaguchi K and Mochizuki T: Proneurotensin/neuromedin N secreted from small cell lung carcinoma cell lines as a potential tumor marker. Proteomics Clin Appl. 2:1620–1627. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Ohshima K, Kanto K, Hatakeyama K, Ide T, Wakabayashi-Nakao K, Watanabe Y, Sakura N, Terashima M, Yamaguchi K and Mochizuki T: Exosome-mediated extracellular release of polyadenylate-binding protein 1 in human metastatic duodenal cancer cells. Proteomics. 14:2297–2306. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Ohshima K, Inoue K, Fujiwara A, Hatakeyama K, Kanto K, Watanabe Y, Muramatsu K, Fukuda Y, Ogura S, Yamaguchi K and Mochizuki T: Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS One. 5:e132472010. View Article : Google Scholar : PubMed/NCBI

17 

Hatakeyama K, Ohshima K, Fukuda Y, Ogura S, Terashima M, Yamaguchi K and Mochizuki T: Identification of a novel protein isoform derived from cancer-related splicing variants using combined analysis of transcriptome and proteome. Proteomics. 11:2275–2282. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Drucker BJ, Marincola FM, Siao DY, Donlon TA, Bangs CD and Holder WD Jr: A new human pancreatic carcinoma cell line developed for adoptive immunotherapy studies with lymphokine-activated killer cells in nude mice. In Vitro Cell Dev Biol. 24:1179–1187. 1988. View Article : Google Scholar : PubMed/NCBI

20 

Yunis AA, Arimura GK and Russin DJ: Human pancreatic carcinoma (MIA PaCa-2) in continuous culture: Sensitivity to asparaginase. Int J Cancer. 19:128–135. 1977. View Article : Google Scholar : PubMed/NCBI

21 

Limame R, Wouters A, Pauwels B, Fransen E, Peeters M, Lardon F, De Wever O and Pauwels P: Comparative analysis of dynamic cell viability, migration and invasion assessments by novel real-time technology and classic endpoint assays. PLoS One. 7:e465362012. View Article : Google Scholar : PubMed/NCBI

22 

Welsch T, Endlich K, Giese T, Büchler MW and Schmidt J: Eps8 is increased in pancreatic cancer and required for dynamic actin-based cell protrusions and intercellular cytoskeletal organization. Cancer Lett. 255:205–218. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Fazioli F, Minichiello L, Matoska V, Castagnino P, Miki T, Wong WT and Di Fiore PP: Eps8, a substrate for the epidermal growth factor receptor kinase, enhances EGF-dependent mitogenic signals. EMBO J. 12:3799–3808. 1993. View Article : Google Scholar : PubMed/NCBI

24 

Castagnino P, Biesova Z, Wong WT, Fazioli F, Gill GN and Di Fiore PP: Direct binding of eps8 to the juxtamembrane domain of EGFR is phosphotyrosine- and SH2-independent. Oncogene. 10:723–729. 1995.PubMed/NCBI

25 

Chen C, Liang Z, Huang W, Li X, Zhou F, Hu X, Han M, Ding X and Xiang S: Eps8 regulates cellular proliferation and migration of breast cancer. Int J Oncol. 46:205–214. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Cattaneo MG, Cappellini E and Vicentini LM: Silencing of Eps8 blocks migration and invasion in human glioblastoma cell lines. Exp Cell Res. 318:1901–1912. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Ding X, Zhou F, Wang F, Yang Z, Zhou C, Zhou J, Zhang B, Yang J, Wang G, Wei Z, et al: Eps8 promotes cellular growth of human malignant gliomas. Oncol Rep. 29:697–703. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Xu M, Shorts-Cary L, Knox AJ, Kleinsmidt-DeMasters B, Lillehei K and Wierman ME: Epidermal growth factor receptor pathway substrate 8 is overexpressed in human pituitary tumors: Role in proliferation and survival. Endocrinology. 150:2064–2071. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Yap LF, Jenei V, Robinson CM, Moutasim K, Benn TM, Threadgold SP, Lopes V, Wei W, Thomas GJ and Paterson IC: Upregulation of Eps8 in oral squamous cell carcinoma promotes cell migration and invasion through integrin-dependent Rac1 activation. Oncogene. 28:2524–2534. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Li Q, Bao W, Fan Q, Shi WJ, Li ZN, Xu Y and Wu D: Epidermal growth factor receptor kinase substrate 8 promotes the metastasis of cervical cancer via the epithelial-mesenchymal transition. Mol Med Rep. 14:3220–3228. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Li YH, Xue TY, He YZ and Du JW: Novel oncoprotein EPS8: A new target for anticancer therapy. Future Oncol. 9:1587–1594. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Tod J, Hanley CJ, Morgan MR, Rucka M, Mellows T, Lopez MA, Kiely P, Moutasim KA, Frampton SJ, Sabnis D, et al: Pro-migratory and TGF-β-activating functions of αvβ6 integrin in pancreatic cancer are differentially regulated via an Eps8-dependent GTPase switch. J Pathol. 243:37–50. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Uchima Y, Sawada T, Nishihara T, Maeda K, Ohira M and Hirakawa K: Inhibition and mechanism of action of a protease inhibitor in human pancreatic cancer cells. Pancreas. 29:123–131. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Peinado H, Zhang H, Matei IR, Costa-Silva B, Hoshino A, Rodrigues G, Psaila B, Kaplan RN, Bromberg JF, Kang Y, et al: Pre-metastatic niches: Organ-specific homes for metastases. Nat Rev Cancer. 17:302–317. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Welsch T, Younsi A, Disanza A, Rodriguez JA, Cuervo AM, Scita G and Schmidt J: Eps8 is recruited to lysosomes and subjected to chaperone-mediated autophagy in cancer cells. Exp Cell Res. 316:1914–1924. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Théry C, Zitvogel L and Amigorena S: Exosomes: Composition, biogenesis and function. Nat Rev Immunol. 2:569–579. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Théry C, Ostrowski M and Segura E: Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. 9:581–593. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Yonemura Y, Endo Y, Yamaguchi T, Fujimura T, Obata T, Kawamura T, Nojima N, Miyazaki I and Sasaki T: Mechanisms of the formation of the peritoneal dissemination in gastric cancer. Int J Oncol. 8:795–802. 1996.PubMed/NCBI

39 

Nishimori H, Yasoshima T, Denno R, Shishido T, Hata F, Okada Y, Ura H, Yamaguchi K, Isomura H, Sato N, et al: A novel experimental mouse model of peritoneal dissemination of human gastric cancer cells: Different mechanisms in peritoneal dissemination and hematogenous metastasis. Jpn J Cancer Res. 91:715–722. 2000. View Article : Google Scholar : PubMed/NCBI

40 

López-Terrada D, Cheung SW, Finegold MJ and Knowles BB: Hep G2 is a hepatoblastoma-derived cell line. Hum Pathol. 40:1512–1515. 2009. View Article : Google Scholar

Related Articles

Journal Cover

February-2019
Volume 41 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ohshima K, Hatakeyama K, Kanto K, Ide T, Watanabe Y, Moromizato S, Wakabayashi‑Nakao K, Sakura N, Yamaguchi K, Mochizuki T, Mochizuki T, et al: Comparative proteomic analysis identifies exosomal Eps8 protein as a potential metastatic biomarker for pancreatic cancer . Oncol Rep 41: 1019-1034, 2019
APA
Ohshima, K., Hatakeyama, K., Kanto, K., Ide, T., Watanabe, Y., Moromizato, S. ... Mochizuki, T. (2019). Comparative proteomic analysis identifies exosomal Eps8 protein as a potential metastatic biomarker for pancreatic cancer . Oncology Reports, 41, 1019-1034. https://doi.org/10.3892/or.2018.6869
MLA
Ohshima, K., Hatakeyama, K., Kanto, K., Ide, T., Watanabe, Y., Moromizato, S., Wakabayashi‑Nakao, K., Sakura, N., Yamaguchi, K., Mochizuki, T."Comparative proteomic analysis identifies exosomal Eps8 protein as a potential metastatic biomarker for pancreatic cancer ". Oncology Reports 41.2 (2019): 1019-1034.
Chicago
Ohshima, K., Hatakeyama, K., Kanto, K., Ide, T., Watanabe, Y., Moromizato, S., Wakabayashi‑Nakao, K., Sakura, N., Yamaguchi, K., Mochizuki, T."Comparative proteomic analysis identifies exosomal Eps8 protein as a potential metastatic biomarker for pancreatic cancer ". Oncology Reports 41, no. 2 (2019): 1019-1034. https://doi.org/10.3892/or.2018.6869