Skip to main content
Erschienen in: Molecular Cancer 1/2018

Open Access 01.12.2018 | Letter to the Editor

Tumor-originated exosomal lncUEGC1 as a circulating biomarker for early-stage gastric cancer

verfasst von: Ling-Yun Lin, Li Yang, Qiang Zeng, Lin Wang, Mao-Li Chen, Ze-Hang Zhao, Guo-Dong Ye, Qi-Cong Luo, Pei-Yu Lv, Qi-Wei Guo, Bo-An Li, Jian-Chun Cai, Wang-Yu Cai

Erschienen in: Molecular Cancer | Ausgabe 1/2018

Abstract

Conventional tumor markers for non-invasive diagnosis of gastric cancer (GC) exhibit insufficient sensitivity and specificity to facilitate detection of early gastric cancer (EGC). We aimed to identify EGC-specific exosomal lncRNA biomarkers that are highly sensitive and stable for the non-invasive diagnosis of EGC. Hence, in the present study, exosomes from the plasma of five healthy individuals and ten stage I GC patients and from culture media of four human primary stomach epithelial cells and four gastric cancer cells (GCCs) were isolated. Exosomal RNA profiling was performed using RNA sequencing to identify EGC-specific exosomal lncRNAs. A total of 79 and 285 exosomal RNAs were expressed at significantly higher levels in stage I GC patients and GCCs, respectively, than that in normal controls. Through combinational analysis of the RNA sequencing results, we found two EGC-specific exosomal lncRNAs, lncUEGC1 and lncUEGC2, which were further confirmed to be remarkably up-regulated in exosomes derived from EGC patients and GCCs. Furthermore, stability testing demonstrates that almost all the plasma lncUEGC1 was encapsulated within exosomes and thus protected from RNase degradation. The diagnostic accuracy of exosomal lncUEGC1 was evaluated, and lncUEGC1 exhibited AUC values of 0.8760 and 0.8406 in discriminating EGC patients from healthy individuals and those with premalignant chronic atrophic gastritis, respectively, which was higher than the diagnostic accuracy of carcinoembryonic antigen. Consequently, exosomal lncUEGC1 may be promising in the development of highly sensitive, stable, and non-invasive biomarkers for EGC diagnosis.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12943-018-0834-9) contains supplementary material, which is available to authorized users.
Abkürzungen
CAG
Chronic atrophic gastritis
CEA
Carcinoembryonic antigen
CM
Culture media
EGC
Early gastric cancer
GC
Gastric cancer
GCCs
Gastric cancer cells
HP
Helicobacter pylori
HPSECs
Human primary stomach epithelial cells
IM
Intestinal metaplasia
lncRNAs
Long non-coding RNAs
lncUEGC
LncRNA up-regulated in the exosomes of gastric cancer
ROC
Receiver operating characteristic
TEM
Transmission electron microscopy

Main text

Gastric cancer (GC) is the fourth most common cancer and the second leading cause of cancer-related death worldwide, and it remains the most lethal cancer in Asia, including China [1]. GC patient survival rates can be improved by early diagnosis and treatment. Currently, EGC diagnosis involves invasive and non-invasive methods, which are painful or exhibit low sensitivity. Worse still, some gastric premalignant and precursor lesions, such as Helicobacter pylori (HP) infection, intestinal metaplasia (IM) and chronic atrophic gastritis (CAG), increase the difficulty of detecting EGC through existing non-invasive diagnostic methods [2].
Circulating RNAs in serum or plasma have emerged as novel non-invasive diagnostic biomarkers [3]. However, a large proportion of the circulating RNAs are easily affected by RNAs released by circulating dysfunctional cells. Previous studies have confirmed that cancerous cells secrete exosomes into the peripheral circulation, and exosomal RNAs can more accurately reflect changes in cancer cells during tumor progression [4]. In addition, exosomes can protect RNA from degradation by endogenous RNases, thereby enhancing the stability of exosomal RNAs in circulating blood [5]. Therefore, circulating exosomal RNAs, especially miRNAs, have emerged as promising biomarkers for the detection of early-stage cancers [68]; however, it remains uncertain whether tumor cell-originated exosomal lncRNAs in plasma can effectively diagnose early-stage cancer. Thus, in this study, we focused on lncRNAs in circulating exosomes that originated from cancer cells to determine their potential value for EGC diagnosis.

RNA sequencing-based screening for identification of EGC-specific exosomal lncRNA

The screening strategy for identification of EGC-specific exosomal lncRNAs is illustrated in Fig. 1. Plasma samples from patients diagnosed with stage I GC (n = 10) and healthy individuals (n = 5) (Additional file 1: Table S1) and culture media (CM) from different GCCs (AGS, KATO III, NCI-N87, and Hs 746 T; n = 4) and HPSECs (n = 4) were collected. Then, exosome-enriched fractions were prepared using step-wise centrifugation-ultracentrifugation and discontinuous iodixanol gradient methods. Transmission electron microscopy (TEM) and NanoSight particle tracking were used to characterize and quantify the isolated exosomes (Additional file 2: Figure S1A-D). Moreover, western blotting revealed the presence of specific exosome marker proteins (CD9 and CD63) and the absence of the negative control protein tubulin in the exosome-enriched fractions (Additional file 2: Figure S1E-F).
Next, exosomal RNA was purified and analyzed via RNA sequencing on an HiSeq3000 instrument. Based on the RNA sequencing data, 79 exosomal lncRNAs and mRNAs were up-regulated by more than 2-fold in stage I GC patients (n = 10) compared with healthy individuals (n = 5) (Q < 0.00001) (Additional file 2: Figure S1G, Additional file 3: Table S2). Previous studies have demonstrated that circulating exosomal lncRNA ZFAS1, LINC00152 and lncRNA HOTTIP levels were significantly elevated in GC patients compared with those in healthy controls [911]; however, these lncRNAs were not found in our up-regulated exosomal RNA profile, likely because they are not present in EGC. In our previous study, we could not exclude the possibility that these RNAs are secreted from other cells, including immune and inflammatory cells, which are coincidentally present at primary tissue lesions. Thus, exosomal RNA from the CM of four HPSECs and four GCCs was also analyzed via RNA sequencing. Two hundred eighty-five exosomal lncRNAs and mRNAs were up-regulated by more than 2-fold in culture medium from GCCs (n = 4) compared with HPSECs (n = 4) (Q < 0.00001) (Additional file 2: Figure S1H, Additional file 4: Table S3). We combined the results and found that six genes, ENST00000568893.1 (lncUEGC1), ENST00000378432.1 (lncUEGC2), DNM1P18, GSC, LRRC4B, and PKD1P2, were significantly up-regulated in both of the groups (Fig. 1). Hence, we focused on the two potential EGC-specific exosomal lncRNAs with the highest up-regulated fold changes and basic expression levels among the six genes and named them lncUEGC1 and lncUEGC2 (lncRNA up-regulated in the exosomes of gastric cancer).

Verification of exosomal lncUEGC1 and lncUEGC2 up-regulation in EGC specimens and GCCs

To further study the robustness of the RNA-seq data, quantitative PCR (qPCR) was performed to analyze exosomal lncUEGC1 and lncUEGC2 expression in EGC specimens from the validation set (Additional file 1: Table S1) and GCCs. As shown in Fig. 2a and b, the relative plasma exosomal lncUEGC1 and lncUEGC2 expression levels were both significantly up-regulated in stage I and II GC patients (n = 51) compared with healthy controls (n = 60) (fold change > 5, P < 0.0001). Moreover, culture medium from 12 different GCCs and four HPSECs obtained via primary culture from four different healthy donors were used to prepare exosome-enriched fractions. The qPCR analysis results revealed that exosomal lncUEGC1 and lncUEGC2 were significantly up-regulated in GCC (n = 12) culture medium compared with those in HPSEC culture medium (n = 4) (lncUEGC1, fold change = 11.27, P = 0.0024; lncUEGC1, fold change = 4.01, P = 0.0054) (Fig. 2c-d). Collectively, these results were consistent with the RNA-seq data and suggested that lncUEGC1 and lncUEGC2 may have potential as EGC-specific exosomal lncRNAs for early non-invasive GC diagnosis.

The stability testing of circulating lncUEGC1 and lncUEGC2 in EGC specimens

Although identification of circulating RNA in plasma is an emerging field for non-invasive diagnostic applications, the majority of long RNA molecules in plasma exhibit poor stability. To examine the stability of lncUEGC1 and lncUEGC2 in plasma, twenty stage I GC specimen plasma samples were directly treated with RNase. The relative plasma levels of total circulating lncUEGC1 were not significantly different between plasma treated with or without RNase (P = 0.4154) (Fig. 2e). However, the relative plasma levels of total circulating lncUEGC2 were down-regulated by approximately two-fold in plasma under RNase treatment (P = 0.0305) (Fig. 2f). We also evaluated lncUEGC1 and lncUEGC2 levels in exosomes-depleted plasma supernatants after ultracentrifugation. The expression of lncUEGC1 were abundant in exosomes instead of exosomes-depleted plasma, but the expression of lncUEGC2 were not significantly different between exosomes and exosomes-depleted plasma (Additional file 5: Figure S2), which revealed that a proportion of lncUEGC2 was not encapsulated within exosomes.
Moreover, we analyzed the correlation between the relative lncUEGC1 and lncUEGC2 levels in plasma exosomal RNA and plasma total circulating RNA. The relative lncUEGC1 and lncUEGC2 levels were both closely related in plasma exosomal RNA and plasma total circulating RNA according to the calculated correlation coefficients (Fig. 2g-h). Taken together, the above results suggest that almost all the plasma lncUEGC1 was encapsulated within exosomes and thus protected from RNase degradation rather than circulating freely in the plasma.

Expression pattern and diagnostic accuracy of CEA, lncUEGC1 and lncUEGC2 in the validation set

To determine the diagnostic power of exosomal lncUEGC1 and lncUEGC2 and the conventional tumor marker CEA for EGC detection, we examined plasma exosomal lncUEGC1 and lncUEGC2 expression levels and serum CEA levels for their capacity to distinguish EGC patients from those with premalignant CAG lesions as well as from healthy controls. As shown in Fig. 3a-c, the relative plasma exosomal lncUEGC1 and lncUEGC2 expression levels and serum CEA levels were all significantly up-regulated in stage I (n = 23) or II (n = 28) GC patients compared with healthy controls (n = 60), but only plasma exosomal lncUEGC1 relative expression levels were significantly up-regulated in stage I (n = 23) GC patients compared with CAG patients (n = 18).
Furthermore, the receiver operating characteristic (ROC) curve (AUC) was generated to evaluate the diagnostic accuracy. Plasma exosomal lncUEGC1 exhibited an AUC value of 0.8760 (P < 0.0001) in distinguishing stage I and II GC patients (n = 51) from healthy controls (n = 60), while plasma exosomal lncUEGC2 exhibited an AUC = 0.7582 (P < 0.0001) and serum CEA exhibited an AUC = 0.6614 (P = 0.0035) (Fig. 3d, Additional file 6: Table S4). The AUC value of exosomal lncUEGC1 was significantly higher than those of the combined serum tumor markers (CEA, CA19-9, CA72-4 and CA12-5) and potential plasma miRNA biomarkers identified in recent studies of EGC diagnosis [12, 13]. More importantly, our diagnostic accuracy study selectively restricted GC patients to those with stage I and II GC, excluding advanced stages, which indicated that lncUEGC1 may be more convincing for EGC diagnosis than markers identified in previous studies investigating diagnostic accuracy in GC patients at all stages [1416]. Similar results were achieved in distinguishing stage I GC patients (n = 23) from healthy controls (n = 60); plasma exosomal lncUEGC1 still showed better diagnostic efficiency, with an AUC = 0.8500 (P < 0.0001), compared with plasma exosomal lncUEGC2, with an AUC = 0.7486 (P = 0.0005), and serum CEA, with an AUC = 0.6424 (P = 0.0456) (Fig. 3e, Additional file 6: Table S4). Because it is difficult to distinguish between early-stage gastric cancer and premalignant lesions using the current liquid biopsy index, we further generated the ROC curve of plasma exosomal lncUEGC1 and lncUEGC2 and serum CEA in distinguishing stage I GC patients (n = 23) from CAG patients (n = 18). As shown in Fig. 3f and Additional file 6: Table S4, plasma exosomal lncUEGC1 exhibited a high AUC value of 0.8406 (P = 0.0002) compared with plasma exosomal lncUEGC2, with an AUC = 0.6522 (P = 0.0980), and serum CEA, with an AUC = 0.6123 (P = 0.2219). Collectively, these results provide evidence that plasma exosomal lncUEGC1 may serve as a primary diagnostic GC biomarker. Moreover, this index might effectively distinguish between EGC and premalignant lesions. All methods and meterials used during this study are included in Additional file 7.

Conclusions

Our research is the first use to use exosomal long chain RNA sequencing to systematically and thoroughly screen potential EGC biomarkers. The unique properties of exosomes, including their ability to embed cancer-originated RNAs, their stability in circulation and their reproducible detection, were revealed in this study. EGC-originated exosomal lncUEGC1 may be promising for the development of highly sensitive, stable, and non-invasive biomarkers for EGC diagnosis.

Funding

This work was supported by grants from the National Natural Science Foundation of China (grant number 81602148 to Wang-Yu Cai; grant number 81472457 to Bo-An Li; grant number 81602563 to Guo-Dong Ye; grant number 81672871 to Qi-Cong Luo); the Natural Science Foundation of Fujian Province (grant number 2015 J01523 to Wang-Yu Cai); and the Science and Technology Projects of Xiamen City (grant number 3502Z20154025 to Wang-Yu Cai).

Availability of data and materials

All data generated or analysed during this study are included in this published article and its additional files.
The studies were conducted in accordance with the International Ethical Guidelines for Biomedical Research Involving Human Subjects (CIOMS), and the research protocols were approved by the Clinical Research Ethics Committee of Zhongshan Hospital of Xiamen University. Written informed consents were obtained from the participants before sampling.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Anhänge

Additional files

Literatur
1.
2.
Zurück zum Zitat Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomark Prev. 2014;23(5):700–13.CrossRef Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomark Prev. 2014;23(5):700–13.CrossRef
3.
Zurück zum Zitat Huang YK, Yu JC. Circulating microRNAs and long non-coding RNAs in gastric cancer diagnosis: an update and review. World J Gastroenterol. 2015;21(34):9863–86.CrossRefPubMedPubMedCentral Huang YK, Yu JC. Circulating microRNAs and long non-coding RNAs in gastric cancer diagnosis: an update and review. World J Gastroenterol. 2015;21(34):9863–86.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Tkach M, Thery C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164(6):1226–32.CrossRefPubMed Tkach M, Thery C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164(6):1226–32.CrossRefPubMed
5.
Zurück zum Zitat Liu T, Zhang X, Gao S, Jing F, Yang Y, Du L, Zheng G, Li P, Li C, Wang C. Exosomal long noncoding RNA CRNDE-h as a novel serum-based biomarker for diagnosis and prognosis of colorectal cancer. Oncotarget. 2016;7(51):85551–63.PubMedPubMedCentral Liu T, Zhang X, Gao S, Jing F, Yang Y, Du L, Zheng G, Li P, Li C, Wang C. Exosomal long noncoding RNA CRNDE-h as a novel serum-based biomarker for diagnosis and prognosis of colorectal cancer. Oncotarget. 2016;7(51):85551–63.PubMedPubMedCentral
6.
Zurück zum Zitat Wang J, Yan F, Zhao Q, Zhan F, Wang R, Wang L, Zhang Y, Huang X. Circulating exosomal miR-125a-3p as a novel biomarker for early-stage colon cancer. Sci Rep. 2017;7(1):4150.CrossRefPubMedPubMedCentral Wang J, Yan F, Zhao Q, Zhan F, Wang R, Wang L, Zhang Y, Huang X. Circulating exosomal miR-125a-3p as a novel biomarker for early-stage colon cancer. Sci Rep. 2017;7(1):4150.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Cazzoli R, Buttitta F, Di Nicola M, Malatesta S, Marchetti A, Rom WN, Pass HI. microRNAs derived from circulating exosomes as noninvasive biomarkers for screening and diagnosing lung cancer. J Thorac Oncol. 2013;8(9):1156–62.CrossRefPubMedPubMedCentral Cazzoli R, Buttitta F, Di Nicola M, Malatesta S, Marchetti A, Rom WN, Pass HI. microRNAs derived from circulating exosomes as noninvasive biomarkers for screening and diagnosing lung cancer. J Thorac Oncol. 2013;8(9):1156–62.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Jin X, Chen Y, Chen H, Fei S, Chen D, Cai X, Liu L, Lin B, Su H, Zhao L, et al. Evaluation of tumor-derived exosomal miRNA as potential diagnostic biomarkers for early-stage non-small cell lung Cancer using next-generation sequencing. Clin Cancer Res. 2017;23(17):5311–9.CrossRefPubMed Jin X, Chen Y, Chen H, Fei S, Chen D, Cai X, Liu L, Lin B, Su H, Zhao L, et al. Evaluation of tumor-derived exosomal miRNA as potential diagnostic biomarkers for early-stage non-small cell lung Cancer using next-generation sequencing. Clin Cancer Res. 2017;23(17):5311–9.CrossRefPubMed
9.
Zurück zum Zitat Pan L, Liang W, Fu M, Huang ZH, Li X, Zhang W, Zhang P, Qian H, Jiang PC, Xu WR, et al. Exosomes-mediated transfer of long noncoding RNA ZFAS1 promotes gastric cancer progression. J Cancer Res Clin Oncol. 2017;143(6):991–1004.CrossRefPubMed Pan L, Liang W, Fu M, Huang ZH, Li X, Zhang W, Zhang P, Qian H, Jiang PC, Xu WR, et al. Exosomes-mediated transfer of long noncoding RNA ZFAS1 promotes gastric cancer progression. J Cancer Res Clin Oncol. 2017;143(6):991–1004.CrossRefPubMed
10.
Zurück zum Zitat Li Q, Shao Y, Zhang X, Zheng T, Miao M, Qin L, Wang B, Ye G, Xiao B, Guo J. Plasma long noncoding RNA protected by exosomes as a potential stable biomarker for gastric cancer. Tumour Biol. 2015;36(3):2007–12.CrossRefPubMed Li Q, Shao Y, Zhang X, Zheng T, Miao M, Qin L, Wang B, Ye G, Xiao B, Guo J. Plasma long noncoding RNA protected by exosomes as a potential stable biomarker for gastric cancer. Tumour Biol. 2015;36(3):2007–12.CrossRefPubMed
11.
Zurück zum Zitat Zhao R, Zhang Y, Zhang X, Yang Y, Zheng X, Li X, Liu Y, Zhang Y. Exosomal long noncoding RNA HOTTIP as potential novel diagnostic and prognostic biomarker test for gastric cancer. Mol Cancer. 2018;17(1):68.CrossRefPubMed Zhao R, Zhang Y, Zhang X, Yang Y, Zheng X, Li X, Liu Y, Zhang Y. Exosomal long noncoding RNA HOTTIP as potential novel diagnostic and prognostic biomarker test for gastric cancer. Mol Cancer. 2018;17(1):68.CrossRefPubMed
12.
Zurück zum Zitat Hung PS, Chen CY, Chen WT, Kuo CY, Fang WL, Huang KH, Chiu PC, Lo SS. miR-376c promotes carcinogenesis and serves as a plasma marker for gastric carcinoma. PLoS One. 2017;12(5):e0177346.CrossRefPubMedPubMedCentral Hung PS, Chen CY, Chen WT, Kuo CY, Fang WL, Huang KH, Chiu PC, Lo SS. miR-376c promotes carcinogenesis and serves as a plasma marker for gastric carcinoma. PLoS One. 2017;12(5):e0177346.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Li C, Li JF, Cai Q, Qiu QQ, Yan M, Liu BY, Zhu ZG. MiRNA-199a-3p: a potential circulating diagnostic biomarker for early gastric cancer. J Surg Oncol. 2013;108(2):89–92.CrossRefPubMed Li C, Li JF, Cai Q, Qiu QQ, Yan M, Liu BY, Zhu ZG. MiRNA-199a-3p: a potential circulating diagnostic biomarker for early gastric cancer. J Surg Oncol. 2013;108(2):89–92.CrossRefPubMed
14.
Zurück zum Zitat Wang H, Wang L, Wu Z, Sun R, Jin H, Ma J, Liu L, Ling R, Yi J, Wang L, et al. Three dysregulated microRNAs in serum as novel biomarkers for gastric cancer screening. Medical oncology (Northwood, London, England). 2014;31(12):298.CrossRef Wang H, Wang L, Wu Z, Sun R, Jin H, Ma J, Liu L, Ling R, Yi J, Wang L, et al. Three dysregulated microRNAs in serum as novel biomarkers for gastric cancer screening. Medical oncology (Northwood, London, England). 2014;31(12):298.CrossRef
15.
Zurück zum Zitat Cai H, Yuan Y, Hao YF, Guo TK, Wei X, Zhang YM. Plasma microRNAs serve as novel potential biomarkers for early detection of gastric cancer. Medical oncology (Northwood, London, England). 2013;30(1):452.CrossRef Cai H, Yuan Y, Hao YF, Guo TK, Wei X, Zhang YM. Plasma microRNAs serve as novel potential biomarkers for early detection of gastric cancer. Medical oncology (Northwood, London, England). 2013;30(1):452.CrossRef
16.
Zurück zum Zitat Shin VY, Ng EK, Chan VW, Kwong A, Chu KM. A three-miRNA signature as promising non-invasive diagnostic marker for gastric cancer. Mol Cancer. 2015;14:202.CrossRefPubMedPubMedCentral Shin VY, Ng EK, Chan VW, Kwong A, Chu KM. A three-miRNA signature as promising non-invasive diagnostic marker for gastric cancer. Mol Cancer. 2015;14:202.CrossRefPubMedPubMedCentral
Metadaten
Titel
Tumor-originated exosomal lncUEGC1 as a circulating biomarker for early-stage gastric cancer
verfasst von
Ling-Yun Lin
Li Yang
Qiang Zeng
Lin Wang
Mao-Li Chen
Ze-Hang Zhao
Guo-Dong Ye
Qi-Cong Luo
Pei-Yu Lv
Qi-Wei Guo
Bo-An Li
Jian-Chun Cai
Wang-Yu Cai
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2018
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0834-9

Weitere Artikel der Ausgabe 1/2018

Molecular Cancer 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.