Next Article in Journal
Highly Metastatic Subpopulation of TNBC Cells Has Limited Iron Metabolism and Is a Target of Iron Chelators
Next Article in Special Issue
A Comprehensive Review of Prognostic Factors in Patients with Gastric Adenocarcinoma
Previous Article in Journal
The Use of Artificial Intelligence (AI) in the Radiology Field: What Is the State of Doctor–Patient Communication in Cancer Diagnosis?
Previous Article in Special Issue
Secondary Primary Cancer after Primary Gastric Cancer: Literature Review and Big Data Analysis Using the Health Insurance Review and Assessment Service (HIRA) Database of Republic of Korea
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Exosomes and Their Potential as Biomarkers in Epstein-Barr Virus-Associated Gastric Cancer

1
Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44610, Republic of Korea
2
Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
3
Center of Companion Diagnostics, Samsung Medical Center, Seoul 06351, Republic of Korea
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(2), 469; https://doi.org/10.3390/cancers15020469
Submission received: 15 November 2022 / Revised: 5 January 2023 / Accepted: 10 January 2023 / Published: 12 January 2023

Abstract

:

Simple Summary

Exosomes are considered to be involved in the pathogenesis of various diseases, including cancer, viral infections, and autoimmune diseases. Research on exosomes is critical, since understanding the pathogenesis of diseases is crucial for their diagnosis and treatment. Epstein-Barr virus (EBV)-infected cells have been found to secrete exosomes for intercellular communication. Exosomal pathways play vital roles in the pathogenesis of EBV-related malignancies. This review aims to summarize the role of exosomes in EBV-associated gastric cancer and to serve as a basis for future diagnostic and therapeutic development.

Abstract

Exosomes are a subtype of extracellular vesicles ranging from 30 to 150 nm and comprising many cellular components, including DNA, RNA, proteins, and metabolites, encapsulated in a lipid bilayer. Exosomes are secreted by many cell types and play important roles in intercellular communication in cancer. Viruses can hijack the exosomal pathway to regulate viral propagation, cellular immunity, and the microenvironment. Cells infected with Epstein-Barr virus (EBV), one of the most common oncogenic viruses, have also been found to actively secrete exosomes, and studies on their roles in EBV-related malignancies are ongoing. In this review, we focus on the role of exosomes in EBV-associated gastric cancer and their clinical applicability in diagnosis and treatment.

1. Introduction

Extracellular vesicles are lipid bilayer-delimited particles released from almost all cell types [1]. As they are known to transport biological information through proteins, nucleic acids, lipids, metabolites, and organelles, research on extracellular vesicles has been invigorated [1]. Extracellular vesicles exist in a wide variety of subtypes that are defined by size, biogenesis pathway, cargo, cellular source, and function; however, they are generally categorized into three types: exosomes, microvesicles, and apoptotic bodies [1]. Exosomes are endosomal in origin and are released from multivesicular bodies by exocytosis, with a size range of 30–150 nm [1]. Microvesicles, also known as ectosomes or microparticles, are of plasma membrane origin and are released from the cell surface with a size range of 30–1000 nm [1]. Apoptotic bodies originate from the plasma membrane and are released by dying cells undergoing apoptosis; they are large vesicles with diameters in the range of microns [1].
Exosomes are released from all cell types and play vital roles in cell-to-cell communication as carriers of genetic material [1]. The blood of healthy individuals contains more than 2000 trillion exosomes, whereas that of patients with cancer contains more than twice as much—indicating highly active intercellular communication through exosomes in cancer and their significance [2,3]. Indeed, many studies have revealed that intercellular interactions via exosomes significantly affect the tumor microenvironment, tumor angiogenesis, tumor growth, metastasis, and drug resistance [4].
Viruses are thought to hijack the exosomal pathway for regulating viral propagation, cellular immunity, and the microenvironment [5]. In the exosomal pathway, cytokines and signaling pathways are altered, leading to cell growth or migration of recipient cells for spreading infection [6]. Exosomes secreted from Epstein-Barr virus (EBV)-infected cells contain viral miRNAs and proteins that increase cytokine and chemokine production by target cells to alter the microenvironment [6,7,8,9,10,11,12,13,14] and significantly impact the pathogenesis of EBV-related malignancies [15,16,17]. Several studies have suggested that exosomes secreted from EBV-infected cells may affect tumorigenesis, metastasis, tumor microenvironment, immune escape, and viral physiology [18,19,20,21]. Polakovicova et al. reported that Helicobacter pylori, a major risk factor for gastric cancer, and EBV alter the expression of host miRNAs and cause immune evasion, epithelial–mesenchymal transition (EMT), and maintenance of infection [16]. Although the roles of exosomes in EBV-related malignancies are being actively researched, little is known about EBV-associated gastric cancer (EBVaGC) compared to B-cell lymphoma or nasopharyngeal cancer (NPC) [15]. In this review, we focused on the role of exosomes in EBVaGC and their clinical applicability in diagnosis and treatment.

2. Balance between Lytic and Latent Cycles

EBV has two phases in its life cycle: lytic replication and latency [17]. A latent infection occurs first upon de novo EBV infection and exhibits one of three patterns depending on the infected cell type [17]. EBVaGC and Burkitt’s lymphoma belong to latency I; NPC and NK/T lymphoma belong to latency II; and lymphoblastoid cell lines belong to latency III [17]. In the latent state, the EBV genome exists in the nucleus as an episome, where closed circular plasmid DNA is incorporated with histones to produce only a limited number of latent viral genes [22,23]. The transfected latent viral genome is amplified once every host cell division and delivered to daughter cells along with the host genome [23]. This silent mode can facilitate persistence by minimizing exposure to the host immune system [23]. In the lytic state, all lytic genes of EBV are expressed simultaneously, resulting in an over 100-fold genome amplification and cell death [23]. The switch from latency to the lytic cycle is critical for viral persistence and the pathogenesis of EBV-related cancers; exosomes are also involved in the balance between these two modes [23]. Unlike B cells, EBV preferentially undergoes lytic replication in epithelial cells to increase the peripheral viral titer and B-cell reactivation, promoting further infection of epithelial cells through reactivated B cells [14]. The miR-200 family, which is known to form a double-negative feedback cycle with zinc finger E-box binding homeobox 1/2 (ZEB1/ZEB2), has been experimentally demonstrated to promote cell transition between the latent and lytic cycles [24,25]; these molecules are secreted from oral epithelial cells via exosomes to induce a lytic cascade in EBV-positive B cells, promoting viral transfer to the oral epithelium [26]. In conclusion, exosomes are involved in the switch from latency to the lytic cycle for viral persistence and play crucial roles in the pathogenesis of EBV-related cancers.

3. EBV Infection in Normal Gastric Cells

EBV infects both B cells and epithelial cells [17]. Gastric epithelial cells can be infected by EBV-infected B cells or directly by EBV [17]. Integrins, ephrin receptor A2, and non-muscle myosin heavy chain IIA are important cofactors for EBV epithelial cell entry [27,28,29,30]. However, gastric mucosal cells are mainly thought to be infected by EBV through EBV-infected B cells rather than by direct infection [31]. EBV-infected lymphocytes come into contact with epithelial cells via integrin β1/β2 and translocate intracellular adhesion molecule-1 to the cell surface, promoting cell-to-cell contact, especially in latency type III cells [7]. Finally, viral particles are transmitted via the clathrin-mediated endocytosis pathway [32]. During this step, exosome-assisted communication between EBV-infected B cells and epithelial cells facilitates cell-to-cell contact-mediated EBV transmission [32]. When exosomal secretions from EBV-positive B cells and epithelial cells are blocked, viral transmission is partly inhibited [33]. This finding indicates that exosomes are involved in EBV transmission into epithelial cells. Moreover, when TGF-β was blocked using an antibody, the lytic cycle in B cells and viral transmission were suppressed in a dose-dependent manner, suggesting that TGF-β spontaneously released from epithelial cells promotes induction of the lytic cycle in B cells, leading to efficient viral transmission into epithelial cells [33]. After endocytosis, EBV-DNA is transported to the nucleus and undergoes circulation, where it is assembled into a circular mini-chromosome [34]; further, a chromatinization process after circulation allows for stable episome formation [34]. The two steps, circulation and chromatinization, are considered crucial for stabilizing latent infection [34]; latent infection is successfully established via extensive CpG methylation [17]. These findings demonstrate that exosomes are involved in EBV transmission into epithelial cells. The molecular processes are depicted in Figure 1.

4. EBV Genes and Their Roles in EBVaGC

EBV infection is found in a small fraction of the non-neoplastic gastric mucosa both in vitro and in vivo [35], suggesting that clonal growth of EBV-infected gastric epithelial cells results in carcinoma rather than augmenting EBV infection of gastric epithelial cells per se [36,37]. DNA methylation machinery is considered to be involved in this process, transforming infected cells into clonal growth [20] and altering mRNA expression, which results in apoptosis inhibition, EMT, and immune evasion [21]. Furthermore, EBV induces exosome secretion by infected epithelial cells to alter the microenvironment in a manner favorable to the tumor [21].
EBVaGC is known to express Epstein-Barr virus-encoded small RNA (EBER) 1 and 2 (EBERs), EBV nuclear antigen 1 (EBNA1), and microRNA BamHI-A rightward transcripts (miR-BARTs), and to express approximately 40% of the latent membrane protein 2A (LMP2A) [35]. Further, the expression of small nucleolar RNA host gene 8 (SNHG8), a long non-coding RNA, is markedly increased in EBVaGC compared with that in EBV-negative gastric cancers or normal gastric mucosa [38,39,40]. Moreover, the knockdown of SNHG8 inhibits cell proliferation and colony formation, arrests the G0/G1 phase in vitro, and suppresses tumor growth in vivo [40]. Although exosomal SNHG8 has not been reported in EBVaGC, it has been found in exosomes obtained from the serum of patients with breast cancer and the breast milk of healthy mothers [41,42]. These genetic materials are believed to contribute to tumorigenesis, and their functions in EBVaGC are summarized in Table 1. In conclusion, exosomes are involved in EBV transmission into epithelial cells and alteration of the microenvironment.

5. Roles of Exosomes in EBVaGC

EBV-infected cells actively release exosomes loaded with viral proteins and miRNAs [7,8,9,10,11,12,13] and EBERs are secreted primarily in complex with the protein La via exosomes [89,90,91]. A previous study characterized exosomes released from EBV-infected cells, including latency I and III cell lines [92]. Compared with the detection of a large number of miRNAs in the exosomes of a latency III cell line, miR-BART1 and miR-BART3 were detected at low levels, and no EBV-mRNA was detected in the exosomes of a latency I cell line [92]. Further, EBV infection alters exosome contents [93]. When exosomes from gastric cancer cell lines with and without artificial EBV infection were analyzed, the expression levels of CD63 and CD81 proteins were increased with EBV infection, indicating an increase in exosome delivery to the microenvironment [93]. The expression level of miR-155 in recipient epithelial cells was also increased in the presence of EBV [94].
These genetic materials are incorporated into cells via exosomes, suggesting that they might function through exosomal transfer [95,96]. Furthermore, accumulating evidence suggests that this exosomal transfer affects the phenotype of recipient cells and influences the microenvironment surrounding the infected cells [92,95]. EBV miR-BART15, which targets the NLRP3 3′-UTR, can be secreted from infected B cells via exosomes to inhibit the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome in non-infected cells, suggesting that EBV may trigger inflammasome activity [12]. EBV latent-infected cells also trigger antiviral immunity in dendritic cells through the selective release and delivery of RNA via exosomes [97]; furthermore, exosomes derived from EBV-positive gastric cancer cell lines suppress dendritic cell maturation [93]. EBER induces type I interferons and inflammatory cytokines via retinoic acid-induced gene I (RIG-I) activation, which are transported to exosomes and function similarly in EBV-infected latency III epithelial cells [95]. In oral squamous cell carcinoma, exosomes carrying EBER-1 can induce indoleamine 2,3-dioxygenase (IDO1) expression in monocyte-derived macrophages, with the help of interleukin (IL)-6 and tumor necrosis factor (TNF)-α-dependent mechanisms via the RIG-I signaling pathway [98]. They can further create an immunosuppressive microenvironment that influences the T-cell immune response [98]. IDO1, an effective immunosuppressive enzyme, is upregulated in EBVaGC, indicating the possibility of an immune evasion strategy [99]. Evidence of inflammasome responses to the EBV genome has been obtained, and this seems to be constitutive during latencies I, II, and III in B cells and epithelial cells [100]. Interferon gamma inducible protein 16 (IFI16) and cleaved interleukins have been detected in exosomes from latency I and III epithelial cells, which could be a strategy for EBV to escape host immunity [100]. Excessive stimulation and uncontrolled proliferation of EBV-infected B cells have been suggested to induce T-cell exhaustion, allowing escape from T-cell surveillance [101]. Furthermore, exosomal IL-1β may promote EBV persistence owing to its inability to recruit neutrophils [100]. In summary, EBV may trigger inflammasome activity through exosomal transfer in the pathogenesis of EBVaGC.

6. EBV Genetic Material and Exosomes

A total of 22 miRNA precursors have been found in EBV, which were then processed into 40 mature miRNAs (miR-BARTs); the spliced and polyadenylated exons then form nuclear non-coding RNAs [20,102]. Four additional miRNAs are derived from BamHI fragment H rightward open reading frame 1 (BHRF-1), but these are only expressed during lytic infections [20]. In EBVaGC, most virus-derived polyadenylated transcripts are from BARTs and these are more abundant in epithelial tumor cells than in lymphoid cells [103]. EBV miR-BART can reduce the lytic replication of viruses by regulating expression of the viral genes BZLF1 and BRLF1, suggesting that it may play a regulatory role between lytic and latent cycles [103]. In latent AGS cells, host cell transcription analysis by RNA-seq revealed that many downregulated genes were BART miRNA targets [102]; this was also observed when a full-length cDNA clone of one of the BART isoforms was artificially expressed in EBV-negative AGS cells [102]. EBV miR-BART cluster 1 and miR-BHRF 1–3 transferred via exosomes also silenced target gene expression in uninfected recipient cells, suggesting that exosomal miRNA transfer from EBV-infected cells to uninfected cells acts as a gene-silencing mechanism [9,16]. miR-BART7-3p shows the highest expression levels in EBVaGC tissues [59]. In NPC, miR-BART7-3p is hypothesized to play a role in chemoresistance, EMT, and metastasis by suppressing phosphatase and tensin homolog (PTEN) and suppressor of mothers against decapentaplegic (SMAD) 7 expression [104,105], but there have been no reports on the function of this miRNA in EBVaGC. miR-BART has various forms that either favorably or adversely affect tumors; however, most of these functions work in favor of tumors. This suggests a complex interaction between miR-BARTs and their targeting mRNA [20]. Further, as miR-BART functions only in EBVaGC, further functional studies are needed to show that miR-BARTs transfer via exosomes and induce substantial changes in recipient cells.
As shown in Table 1, several roles of EBNA1 have been revealed in EBVaGC; however, the role of EBNA1 via exosomes is not well understood. High levels of EBNA1 expression have been observed in the exosomes of patients with multiple sclerosis [106], and the inflammatory cascade induced by the EBNA1-DNA complex from apoptotic EBV-positive B cells has been suggested to underlie the pathogenesis of multiple sclerosis [101]. Although further studies are needed in this regard, as EBNA1 is known to play a crucial role in regulating EBV genome persistence and cell division [107], its role via exosomes is expected to be related to this aspect.
The relationship between LMP2A and exosomes is also unclear. LMP2A is secreted in the form of exosomes, and cholesterol is considered to be involved in the trafficking and stability of LMP2A [108]. In a comprehensive proteomic analysis of LMP1 and LMP2A, these proteins were found to affect proteasome subunits, ubiquitin-specific conjugates, peptidase, and vesicle-trafficking proteins, indicating that they are also involved in regulating exosome trafficking [109]. Further, LMP2A in exosomes can be used for diagnosing NPC with high accuracy [110]. Unlike B-cell lymphoma and NPC, EBVaGC does not express LMP1. In one study, co-culture of LMP1-positive and -negative cells using the gastric cancer cell line AGS resulted in a gradual decrease in the number of LMP1-positive cells at each cell passage; further, LMP1-positive cells stimulated proliferation of the surrounding LMP1-negative cells with exosome-mediated epidermal growth factor receptor (EGFR) activation [111]. This could explain the downregulated LMP1 expression in patients with EBVaGC. Although little is known about the relationship between EBV genetic material and exosomes, exosomal miRNA transfer from EBV-infected cells to uninfected cells acts as a gene-silencing mechanism.

7. Clinical Applicability of Exosomes

An ideal biomarker should have the following features: it should be stable in the body fluid for at least 24 h; its concentration should be determined quantitatively; and the measurement method should be easy, rapid, and affordable [112]. Exosomes, which reflect the donor cell characteristics, are present in almost all body fluids and can safely migrate long distances while being surrounded by lipid bilayers [113]; in particular, as the concentration of exosomes in cancer increases [3], exosomes in liquid biopsies are expected to facilitate non-invasive cancer diagnosis [113].
Some studies have evaluated the potential of exosomes as biomarkers using patient samples in EBV-related malignancy, but all of them have focused on NPC [114,115,116]. Extracellular vesicle-bound BART 13-3p appears to be the optimal selective marker to differentiate NPC from healthy cases, other types of cancer, and EBV-associated disease as controls [114]. The level of cyclophilin A, a member of the immunophilin family, was found to be significantly higher in both serum and exosomes from patients with NPC than from healthy individuals; further, when exosomal cyclophilin A and the antibody for EBV capsid antigen immunoglobulin A were combined, the diagnostic sensitivity and specificity were enhanced [115].
Although not exosomes, cell-free EBV DNAs were detected in the plasma of patients with EBVaGC and showed 71.4% sensitivity and 97.1% specificity [117]. When the plasma EBV ratio was repeatedly evaluated, a decrease or increase in plasma EBV DNA was observed after treatment and during tumor progression/relapse, respectively [117], suggesting that plasma EBV DNA monitoring could be important for patients with EBVaGC [118]. Further, candidate substances that could be potential exosomal targets, including LMP1, LMP2A, BARF1, EBV DNAs, EBV mRNAs, and EBV miRNAs, have been found in serum, saliva, and tumors, but most of these experiments were conducted on lymphoma or NPC [15,116]. Although more research is needed, these genetic products of EBV may be useful biomarkers for EBVaGC, as well as other EBV-related malignancies [21].
EBV-related oncoproteins and miRNAs have also been suggested as potential prognostic markers for EBVaGC. Upregulated BART20-5p expression in EBVaGC has been reported to be associated with poor recurrence-free survival [119]; further, the EBV-encoded circular RNA LMP2A plays crucial roles in inducing and maintaining the stemness phenotype [120] and is associated with distant metastasis and poor prognosis by promoting angiogenesis under hypoxia [74,120]. A high EBV copy number per genome (>10 copies) has also been reported to be correlated with programmed death ligand-1 (PD-L1) expression in cancer cells and poor disease-specific survival in EBVaGC [121]. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway can be activated by LMP1 and LMP2A, resulting in resistance to chemotherapy [70]. Treatment with a PI3K inhibitor can augment the effect of chemotherapy in gastric cancer cell lines [70], suggesting that LMP1 and LMP2A are also involved in drug resistance. EBNA1 transfection reduces the phosphorylation of ataxia telangiectasia and Rad3 related (ATR) with phosphorylation of p38 mitogen-activated protein kinase (MAPK), resulting in increased susceptibility to olaparib; the same result was obtained when an ATR kinase or p38 MAPK inhibitor was used [122]. The potential diagnostic and prognostic biomarkers of EBVaGC are summarized in Table 2.
Exosomes are being extensively studied as important biological mediators, as well as vehicles for drug delivery, because they are well tolerated and do not induce a significant immune reaction or toxicity, as observed even after repeated injections in mice [124,125,126,127,128,129,130]. One study reported that exosomes derived from phosphoantigen-expanded Vδ2-T cells can target and kill EBV-associated tumor cells through the Fas ligand and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathways and promote EBV antigen-specific CD4 and CD8 expansion in vitro; their antitumor activity was also confirmed in humanized mice [131]. Exosomes derived from γδ-T cells preserved their antitumor activities, such as tumor killing and T-cell promotion in the microenvironment of immunosuppressive NPC [132]. Although no experimental data have been obtained using exosomes as therapeutic agents in EBVaGC, these results suggest the potential of exosomes as therapeutic agents.
There are several limitations to using exosomes as biomarkers for EBVaGC. First, they may not be specific to a particular type of cancer and have a low sensitivity, particularly in the early stages of cancer [19]. Ultracentrifugation and density gradient centrifugation are the most common methods for isolating exosomes; however, these do not have a high purity [14]. Therefore, for exosomes to be used in clinical practice, more reliable isolation methods must be developed and sufficient validation is required. In conclusion, exosomes are expected to facilitate non-invasive cancer diagnosis and treatment through optimal isolation methods and applications in drug delivery and immune modulation.

8. Immunotherapy in EBVaGC

Immunotherapy has recently become a topic of much interest for many types of malignancies. Efforts are being made to find an appropriate biomarker for selecting a patient group that is likely to respond to immunotherapy, and the most used biomarker is PD-L1 immunohistochemistry. One of the molecular characteristics of EBVaGC is the overexpression of PD-L1/PD-L2, which is expected to respond well to immunotherapy and has shown a good response in several studies [133,134]. One study reported that six stage IV EBVaGC cases exhibited partial responses after immunotherapy [133], while another study reported that nine stage IV EBVaGC cases showed partial responses (3/9, 33.3%) and stable disease (5/9, 55.6%) [134]. Two cases of overly pretreated patients with refractory metastatic EBVaGC, who experienced a significant and sustained response to pembrolizumab monotherapy, were published as case reports [135].
Although the exact mechanism of PD-L1 overexpression in EBVaGC has not yet been elucidated, it has been addressed in several papers [61,136,137]. One study showed that EBV-miR-BART11 and EBV-miR-BART17-3p upregulated the expression of PD-L1 in EBVaGC and NPC, resulting in the promotion of tumor immune escape [136]. Another study showed that PD-L1 expression was dramatically increased by activating Janus kinase 2 (JAK2)/signal transducer and activator of transcription 1 (STAT1)/interferon regulatory factor 1 (IRF-1) signaling in EBV-positive GC cells compared with that in EBV-negative GC cells after interferon (IFN)γ treatment [137]. In another study, transfection of miR-BART5-5p into EBV-negative GC cells induced protein inhibitor of activated STAT3 (PIAS3)/pSTAT3-dependent PD-L1 upregulation, while PD-L1 knockdown increased apoptosis and decreased cell proliferation, invasion ability, and migration [61]. However, the correlation between exosomes and PD-L1 expression in EBVaGC has not yet been reported. However, as exosomal PD-L1 plays a major role in tumor progression in several other malignancies and has attracted attention as a promising therapeutic target, it is expected to exert similar effects in EBVaGC [138].

9. Conclusions

In EBVaGC, similar to many other types of malignancies, exosomes may be involved in tumorigenesis, tumor microenvironment, immune escape, and the maintenance of viral physiology [18,19,20,21]. Further, exosomes are also considered to be involved in the balance between the lytic and latent cycles, which is important for viral persistence and the pathogenesis of EBV-related cancers [23].
Gastric epithelial cells can be infected through EBV-infected B cells or directly by EBV [17,31]. Integrins, ephrin receptor A2, and non-muscle myosin heavy chain IIA act as important cofactors for epithelial infection by EBV [27,28,29,30], while integrin β1/β2 and intracellular adhesion molecule-1 are involved in epithelial infection by EBV-infected B cells [7]. TGF-β spontaneously released from epithelial cells promotes induction of the lytic cycle in B cells and causes efficient viral transmission into epithelial cells [33].
Genetic materials from EBV-infected epithelial cells are released through exosomes and affect the tumor microenvironment. EBNA1 is thought to be involved in EBV genome persistence and cell division [107], LMP2A in regulation of exosome trafficking [109], miR-BART in target gene-silencing and balancing between the viral cycles [9,16,103], and EBER in modulation of the inflammatory response [95,98,99]. While IFI16 and cleaved interleukins are involved in escaping host immunity, IL-1β is thought to promote EBV persistence [100,101].
To date, there has been no paper showing the diagnostic usefulness of exosomes in EBVaGC; however, cell-free EBV DNA shows a high sensitivity and specificity in patients with EBVaGC [117]. Since a decrease or increase in plasma EBV DNA correlates with the clinical course, its usefulness in clinical practice is expected [117]. There are several papers showing that EBV-related oncoproteins and miRNAs are associated with prognosis in EBVaGC [70,74,119,120,122]. LMP2A and EBNA1 are considered to be associated with chemotherapy response [70,122], and are thought to be potential biomarkers for the diagnosis and prognosis of EBVaGC even within exosomes.
One of the molecular characteristics of EBVaGC is the overexpression of PD-L1/PD-L2, which has shown a good response for immunotherapy [133,134]. Although the exact mechanism of PD-L1 overexpression in EBVaGC has not been elucidated, EBV miR-BART11, EBV-BART17-3p, JAK2/STAT1/IRF-1 pathway, and PIAS3/pSTAT3 pathway induced by miR-BART5-5p are thought to be involved in PD-L1 overexpression in EBVaGC [61,136,137]. As exosomal PD-L1 has attracted attention as a promising diagnostic and therapeutic target in other malignancies, similar attention should be paid to its role in EBVaGC [138].
Compared with NPC or EBV-related B-cell lymphomas, there are fewer published articles on exosomes in EBVaGC. Therefore, further research is warranted to prove the functions of exosomes in EBVaGC. Although this review was based on a limited number of papers and information, we found that exosomes may be involved in tumorigenesis, tumor microenvironment, immune escape, and viral persistence in EBVaGC. By developing reliable isolation methods and validation studies, exosomes can be applied in clinical practice to diagnose and treat EBVaGC.

Author Contributions

Conceptualization, B.K.; writing—original draft preparation, B.K.; writing—review and editing, B.K. and K.-M.K.; supervision, K.-M.K.; project administration, K.-M.K.; funding acquisition, K.-M.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from the Korea Health Technology R&D Project via the Korea Health Industry Development Institute, funded by the Ministry of Health and Welfare, Republic of Korea (grant numbers: HR20C0025 and HI21C1137).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
  2. Zhang, L.; Yu, D. Exosomes in cancer development, metastasis, and immunity. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 455–468. [Google Scholar] [CrossRef] [PubMed]
  3. Zhong, Y.; Li, H.; Li, P.; Chen, Y.; Zhang, M.; Yuan, Z.; Zhang, Y.; Xu, Z.; Luo, G.; Fang, Y.; et al. Exosomes: A New Pathway for Cancer Drug Resistance. Front. Oncol. 2021, 11, 743556. [Google Scholar] [CrossRef] [PubMed]
  4. Paskeh, M.D.A.; Entezari, M.; Mirzaei, S.; Zabolian, A.; Saleki, H.; Naghdi, M.J.; Sabet, S.; Khoshbakht, M.A.; Hashemi, M.; Hushmandi, K.; et al. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J. Hematol. Oncol. 2022, 15, 83. [Google Scholar] [CrossRef]
  5. Saad, M.H.; Badierah, R.; Redwan, E.M.; El-Fakharany, E.M. A Comprehensive Insight into the Role of Exosomes in Viral Infection: Dual Faces Bearing Different Functions. Pharmaceutics 2021, 13, 1405. [Google Scholar] [CrossRef]
  6. Schwab, A.; Meyering, S.S.; Lepene, B.; Iordanskiy, S.; van Hoek, M.L.; Hakami, R.M.; Kashanchi, F. Extracellular vesicles from infected cells: Potential for direct pathogenesis. Front. Microbiol. 2015, 6, 1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Nanbo, A.; Kawanishi, E.; Yoshida, R.; Yoshiyama, H. Exosomes derived from Epstein-Barr virus-infected cells are internalized via caveola-dependent endocytosis and promote phenotypic modulation in target cells. J. Virol. 2013, 87, 10334–10347. [Google Scholar] [CrossRef] [Green Version]
  8. Meckes, D.G., Jr.; Shair, K.H.; Marquitz, A.R.; Kung, C.P.; Edwards, R.H.; Raab-Traub, N. Human tumor virus utilizes exosomes for intercellular communication. Proc. Natl. Acad. Sci. USA 2010, 107, 20370–20375. [Google Scholar] [CrossRef] [Green Version]
  9. Pegtel, D.M.; Cosmopoulos, K.; Thorley-Lawson, D.A.; van Eijndhoven, M.A.; Hopmans, E.S.; Lindenberg, J.L.; de Gruijl, T.D.; Würdinger, T.; Middeldorp, J.M. Functional delivery of viral miRNAs via exosomes. Proc. Natl. Acad. Sci. USA 2010, 107, 6328–6333. [Google Scholar] [CrossRef] [Green Version]
  10. Verweij, F.J.; van Eijndhoven, M.A.; Hopmans, E.S.; Vendrig, T.; Wurdinger, T.; Cahir-McFarland, E.; Kieff, E.; Geerts, D.; van der Kant, R.; Neefjes, J.; et al. LMP1 association with CD63 in endosomes and secretion via exosomes limits constitutive NF-κB activation. Embo J. 2011, 30, 2115–2129. [Google Scholar] [CrossRef]
  11. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Haneklaus, M.; Gerlic, M.; Kurowska-Stolarska, M.; Rainey, A.A.; Pich, D.; McInnes, I.B.; Hammerschmidt, W.; O’Neill, L.A.; Masters, S.L. Cutting edge: miR-223 and EBV miR-BART15 regulate the NLRP3 inflammasome and IL-1β production. J. Immunol. 2012, 189, 3795–3799. [Google Scholar] [CrossRef] [Green Version]
  13. Aromseree, S.; Middeldorp, J.M.; Pientong, C.; van Eijndhoven, M.; Ramayanti, O.; Lougheed, S.M.; Pegtel, D.M.; Steenbergen, R.D.; Ekalaksananan, T. High Levels of EBV-Encoded RNA 1 (EBER1) Trigger Interferon and Inflammation-Related Genes in Keratinocytes Expressing HPV16 E6/E7. PLoS ONE 2017, 12, e0169290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Zhao, M.; Nanbo, A.; Sun, L.; Lin, Z. Extracellular Vesicles in Epstein-Barr Virus’ Life Cycle and Pathogenesis. Microorganisms 2019, 7, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Teow, S.Y.; Liew, K.; Khoo, A.S.; Peh, S.C. Pathogenic Role of Exosomes in Epstein-Barr Virus (EBV)-Associated Cancers. Int. J. Biol. Sci. 2017, 13, 1276–1286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Polakovicova, I.; Jerez, S.; Wichmann, I.A.; Sandoval-Bórquez, A.; Carrasco-Véliz, N.; Corvalán, A.H. Role of microRNAs and Exosomes in Helicobacter pylori and Epstein-Barr Virus Associated Gastric Cancers. Front. Microbiol. 2018, 9, 636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Zhao, Y.; Zhang, J.; Cheng, A.S.L.; Yu, J.; To, K.F.; Kang, W. Gastric cancer: Genome damaged by bugs. Oncogene 2020, 39, 3427–3442. [Google Scholar] [CrossRef] [Green Version]
  18. Zhang, J.; Li, X.; Hu, J.; Cao, P.; Yan, Q.; Zhang, S.; Dang, W.; Lu, J. Long noncoding RNAs involvement in Epstein-Barr virus infection and tumorigenesis. Virol. J. 2020, 17, 51. [Google Scholar] [CrossRef] [Green Version]
  19. AbuSalah, M.A.H.; Gan, S.H.; Al-Hatamleh, M.A.I.; Irekeola, A.A.; Shueb, R.H.; Yean Yean, C. Recent Advances in Diagnostic Approaches for Epstein-Barr Virus. Pathogens 2020, 9, 226. [Google Scholar] [CrossRef] [Green Version]
  20. Fukayama, M.; Abe, H.; Kunita, A.; Shinozaki-Ushiku, A.; Matsusaka, K.; Ushiku, T.; Kaneda, A. Thirty years of Epstein-Barr virus-associated gastric carcinoma. Virchows Arch. 2020, 476, 353–365. [Google Scholar] [CrossRef]
  21. Abe, H.; Kaneda, A.; Fukayama, M. Epstein-Barr Virus-Associated Gastric Carcinoma: Use of Host Cell Machineries and Somatic Gene Mutations. Pathobiology 2015, 82, 212–223. [Google Scholar] [CrossRef] [PubMed]
  22. Odumade, O.A.; Hogquist, K.A.; Balfour, H.H., Jr. Progress and problems in understanding and managing primary Epstein-Barr virus infections. Clin. Microbiol. Rev. 2011, 24, 193–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Murata, T.; Tsurumi, T. Switching of EBV cycles between latent and lytic states. Rev. Med. Virol. 2014, 24, 142–153. [Google Scholar] [CrossRef] [PubMed]
  24. Lin, Z.; Wang, X.; Fewell, C.; Cameron, J.; Yin, Q.; Flemington, E.K. Differential expression of the miR-200 family microRNAs in epithelial and B cells and regulation of Epstein-Barr virus reactivation by the miR-200 family member miR-429. J. Virol. 2010, 84, 7892–7897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ellis-Connell, A.L.; Iempridee, T.; Xu, I.; Mertz, J.E. Cellular microRNAs 200b and 429 regulate the Epstein-Barr virus switch between latency and lytic replication. J. Virol. 2010, 84, 10329–10343. [Google Scholar] [CrossRef] [Green Version]
  26. Lin, Z.; Swan, K.; Zhang, X.; Cao, S.; Brett, Z.; Drury, S.; Strong, M.J.; Fewell, C.; Puetter, A.; Wang, X.; et al. Secreted Oral Epithelial Cell Membrane Vesicles Induce Epstein-Barr Virus Reactivation in Latently Infected B Cells. J. Virol. 2016, 90, 3469–3479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Zhang, H.; Li, Y.; Wang, H.B.; Zhang, A.; Chen, M.L.; Fang, Z.X.; Dong, X.D.; Li, S.B.; Du, Y.; Xiong, D.; et al. Ephrin receptor A2 is an epithelial cell receptor for Epstein-Barr virus entry. Nat. Microbiol. 2018, 3, 1–8. [Google Scholar] [CrossRef]
  28. Chesnokova, L.S.; Hutt-Fletcher, L.M. Fusion of Epstein-Barr virus with epithelial cells can be triggered by αvβ5 in addition to αvβ6 and αvβ8, and integrin binding triggers a conformational change in glycoproteins gHgL. J. Virol. 2011, 85, 13214–13223. [Google Scholar] [CrossRef] [Green Version]
  29. Chesnokova, L.S.; Nishimura, S.L.; Hutt-Fletcher, L.M. Fusion of epithelial cells by Epstein-Barr virus proteins is triggered by binding of viral glycoproteins gHgL to integrins alphavbeta6 or alphavbeta8. Proc. Natl. Acad. Sci. USA 2009, 106, 20464–20469. [Google Scholar] [CrossRef] [Green Version]
  30. Xiong, D.; Du, Y.; Wang, H.B.; Zhao, B.; Zhang, H.; Li, Y.; Hu, L.J.; Cao, J.Y.; Zhong, Q.; Liu, W.L.; et al. Nonmuscle myosin heavy chain IIA mediates Epstein-Barr virus infection of nasopharyngeal epithelial cells. Proc. Natl. Acad. Sci. USA 2015, 112, 11036–11041. [Google Scholar] [CrossRef]
  31. Imai, S.; Nishikawa, J.; Takada, K. Cell-to-cell contact as an efficient mode of Epstein-Barr virus infection of diverse human epithelial cells. J. Virol. 1998, 72, 4371–4378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Nanbo, A.; Kachi, K.; Yoshiyama, H.; Ohba, Y. Epstein-Barr virus exploits host endocytic machinery for cell-to-cell viral transmission rather than a virological synapse. J. Gen. Virol. 2016, 97, 2989–3006. [Google Scholar] [CrossRef] [PubMed]
  33. Nanbo, A.; Ohashi, M.; Yoshiyama, H.; Ohba, Y. The Role of Transforming Growth Factor β in Cell-to-Cell Contact-Mediated Epstein-Barr Virus Transmission. Front. Microbiol. 2018, 9, 984. [Google Scholar] [CrossRef] [PubMed]
  34. Lieberman, P.M. Keeping it quiet: Chromatin control of gammaherpesvirus latency. Nat. Rev. Microbiol. 2013, 11, 863–875. [Google Scholar] [CrossRef]
  35. Iizasa, H.; Nanbo, A.; Nishikawa, J.; Jinushi, M.; Yoshiyama, H. Epstein-Barr Virus (EBV)-associated gastric carcinoma. Viruses 2012, 4, 3420–3439. [Google Scholar] [CrossRef] [Green Version]
  36. Fukayama, M.; Hayashi, Y.; Iwasaki, Y.; Chong, J.; Ooba, T.; Takizawa, T.; Koike, M.; Mizutani, S.; Miyaki, M.; Hirai, K. Epstein-Barr virus-associated gastric carcinoma and Epstein-Barr virus infection of the stomach. Lab. Investig. 1994, 71, 73–81. [Google Scholar]
  37. Hayashi, K.; Teramoto, N.; Akagi, T.; Sasaki, Y.; Suzuki, T. In situ detection of Epstein-Barr virus in the gastric glands with intestinal metaplasia. Am. J. Gastroenterol. 1996, 91, 1481. [Google Scholar]
  38. Chen, B.Z.; Lin, X.D.; Chen, G.; Hu, D.; Zhu, Q.; Shi, Y.; Wang, X.J.; Jin, S.F.; Wang, H.F.; Zheng, X.W. [Expression of long non-coding RNA SNHG8 in Epstein-Barr virus-related gastric cancer and clinical outcome]. Zhonghua Bing Li Xue Za Zhi 2017, 46, 84–87. [Google Scholar] [CrossRef]
  39. Huang, T.; Ji, Y.; Hu, D.; Chen, B.; Zhang, H.; Li, C.; Chen, G.; Luo, X.; Zheng, X.W.; Lin, X. SNHG8 is identified as a key regulator of epstein-barr virus (EBV)-associated gastric cancer by an integrative analysis of lncRNA and mRNA expression. Oncotarget 2016, 7, 80990–81002. [Google Scholar] [CrossRef] [Green Version]
  40. Liu, J.; Yang, C.; Gu, Y.; Li, C.; Zhang, H.; Zhang, W.; Wang, X.; Wu, N.; Zheng, C. Knockdown of the lncRNA SNHG8 inhibits cell growth in Epstein-Barr virus-associated gastric carcinoma. Cell. Mol. Biol. Lett. 2018, 23, 17. [Google Scholar] [CrossRef]
  41. Zhao, X.; Guo, X.; Jiao, D.; Zhu, J.; Xiao, H.; Yang, Y.; Zhao, S.; Zhang, J.; Jiao, F.; Liu, Z. Analysis of the expression profile of serum exosomal lncRNA in breast cancer patients. Ann. Transl. Med. 2021, 9, 1382. [Google Scholar] [CrossRef] [PubMed]
  42. Karlsson, O.; Rodosthenous, R.S.; Jara, C.; Brennan, K.J.; Wright, R.O.; Baccarelli, A.A.; Wright, R.J. Detection of long non-coding RNAs in human breastmilk extracellular vesicles: Implications for early child development. Epigenetics 2016, 11, 721–729. [Google Scholar] [CrossRef] [PubMed]
  43. Iwakiri, D.; Eizuru, Y.; Tokunaga, M.; Takada, K. Autocrine growth of Epstein-Barr virus-positive gastric carcinoma cells mediated by an Epstein-Barr virus-encoded small RNA. Cancer Res. 2003, 63, 7062–7067. [Google Scholar] [PubMed]
  44. Banerjee, A.S.; Pal, A.D.; Banerjee, S. Epstein-Barr virus-encoded small non-coding RNAs induce cancer cell chemoresistance and migration. Virology 2013, 443, 294–305. [Google Scholar] [CrossRef] [Green Version]
  45. Shinozaki, A.; Sakatani, T.; Ushiku, T.; Hino, R.; Isogai, M.; Ishikawa, S.; Uozaki, H.; Takada, K.; Fukayama, M. Downregulation of microRNA-200 in EBV-associated gastric carcinoma. Cancer Res. 2010, 70, 4719–4727. [Google Scholar] [CrossRef] [Green Version]
  46. Nishikawa, H.; Christiany, P.; Hayashi, T.; Iizasa, H.; Yoshiyama, H.; Hatakeyama, M. Kinase Activity of PAR1b, Which Mediates Nuclear Translocation of the BRCA1 Tumor Suppressor, Is Potentiated by Nucleic Acid-Mediated PAR1b Multimerization. Int. J. Mol. Sci. 2022, 23, 6634. [Google Scholar] [CrossRef]
  47. Cheng, T.C.; Hsieh, S.S.; Hsu, W.L.; Chen, Y.F.; Ho, H.H.; Sheu, L.F. Expression of Epstein-Barr nuclear antigen 1 in gastric carcinoma cells is associated with enhanced tumorigenicity and reduced cisplatin sensitivity. Int. J. Oncol. 2010, 36, 151–160. [Google Scholar]
  48. Sivachandran, N.; Dawson, C.W.; Young, L.S.; Liu, F.F.; Middeldorp, J.; Frappier, L. Contributions of the Epstein-Barr virus EBNA1 protein to gastric carcinoma. J. Virol. 2012, 86, 60–68. [Google Scholar] [CrossRef] [Green Version]
  49. Leight, E.R.; Sugden, B. EBNA-1: A protein pivotal to latent infection by Epstein-Barr virus. Rev. Med. Virol. 2000, 10, 83–100. [Google Scholar] [CrossRef]
  50. Li, N.; Thompson, S.; Schultz, D.C.; Zhu, W.; Jiang, H.; Luo, C.; Lieberman, P.M. Discovery of selective inhibitors against EBNA1 via high throughput in silico virtual screening. PLoS ONE 2010, 5, e10126. [Google Scholar] [CrossRef]
  51. Armero, V.E.S.; Tremblay, M.P.; Allaire, A.; Boudreault, S.; Martenon-Brodeur, C.; Duval, C.; Durand, M.; Lapointe, E.; Thibault, P.; Tremblay-Létourneau, M.; et al. Transcriptome-wide analysis of alternative RNA splicing events in Epstein-Barr virus-associated gastric carcinomas. PLoS ONE 2017, 12, e0176880. [Google Scholar] [CrossRef] [Green Version]
  52. Song, H.; Zhang, Y.; Liu, J.; Liu, W.; Luo, B. Activation of DNA methyltransferase 3a by Epstein-Barr nuclear antigen 1 in gastric carcinoma. Dig. Liver Dis. 2022, 54, 973–983. [Google Scholar] [CrossRef] [PubMed]
  53. Marquitz, A.R.; Mathur, A.; Chugh, P.E.; Dittmer, D.P.; Raab-Traub, N. Expression profile of microRNAs in Epstein-Barr virus-infected AGS gastric carcinoma cells. J. Virol. 2014, 88, 1389–1393. [Google Scholar] [CrossRef] [PubMed]
  54. Marquitz, A.R.; Mathur, A.; Shair, K.H.; Raab-Traub, N. Infection of Epstein-Barr virus in a gastric carcinoma cell line induces anchorage independence and global changes in gene expression. Proc. Natl. Acad. Sci. USA 2012, 109, 9593–9598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Min, K.; Kim, J.Y.; Lee, S.K. Epstein-Barr virus miR-BART1-3p suppresses apoptosis and promotes migration of gastric carcinoma cells by targeting DAB2. Int. J. Biol. Sci. 2020, 16, 694–707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Jing, J.J.; Wang, Z.Y.; Li, H.; Sun, L.P.; Yuan, Y. Key elements involved in Epstein-Barr virus-associated gastric cancer and their network regulation. Cancer Cell Int. 2018, 18, 146. [Google Scholar] [CrossRef] [Green Version]
  57. Liu, J.; Zhang, Y.; Liu, W.; Zhang, Q.; Xiao, H.; Song, H.; Luo, B. MiR-BART1-5p targets core 2β-1,6-acetylglucosaminyltransferase GCNT3 to inhibit cell proliferation and migration in EBV-associated gastric cancer. Virology 2020, 541, 63–74. [Google Scholar] [CrossRef]
  58. Wang, J.; Zheng, X.; Qin, Z.; Wei, L.; Lu, Y.; Peng, Q.; Gao, Y.; Zhang, X.; Zhang, X.; Li, Z.; et al. Epstein-Barr virus miR-BART3-3p promotes tumorigenesis by regulating the senescence pathway in gastric cancer. J. Biol. Chem. 2019, 294, 4854–4866. [Google Scholar] [CrossRef] [Green Version]
  59. Shinozaki-Ushiku, A.; Kunita, A.; Isogai, M.; Hibiya, T.; Ushiku, T.; Takada, K.; Fukayama, M. Profiling of Virus-Encoded MicroRNAs in Epstein-Barr Virus-Associated Gastric Carcinoma and Their Roles in Gastric Carcinogenesis. J. Virol. 2015, 89, 5581–5591. [Google Scholar] [CrossRef] [Green Version]
  60. Zheng, X.; Wang, J.; Wei, L.; Peng, Q.; Gao, Y.; Fu, Y.; Lu, Y.; Qin, Z.; Zhang, X.; Lu, J.; et al. Epstein-Barr Virus MicroRNA miR-BART5-3p Inhibits p53 Expression. J. Virol. 2018, 92, e01022-18. [Google Scholar] [CrossRef] [Green Version]
  61. Yoon, C.J.; Chang, M.S.; Kim, D.H.; Kim, W.; Koo, B.K.; Yun, S.C.; Kim, S.H.; Kim, Y.S.; Woo, J.H. Epstein-Barr virus-encoded miR-BART5-5p upregulates PD-L1 through PIAS3/pSTAT3 modulation, worsening clinical outcomes of PD-L1-positive gastric carcinomas. Gastric Cancer 2020, 23, 780–795. [Google Scholar] [CrossRef]
  62. Choi, H.; Lee, H.; Kim, S.R.; Gho, Y.S.; Lee, S.K. Epstein-Barr virus-encoded microRNA BART15-3p promotes cell apoptosis partially by targeting BRUCE. J. Virol. 2013, 87, 8135–8144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Choi, H.; Lee, S.K. TAX1BP1 downregulation by EBV-miR-BART15-3p enhances chemosensitivity of gastric cancer cells to 5-FU. Arch. Virol. 2017, 162, 369–377. [Google Scholar] [CrossRef]
  64. Yoon, J.H.; Min, K.; Lee, S.K. Epstein-Barr Virus miR-BART17-5p Promotes Migration and Anchorage-Independent Growth by Targeting Kruppel-Like Factor 2 in Gastric Cancer. Microorganisms 2020, 8, 258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Tsai, C.Y.; Liu, Y.Y.; Liu, K.H.; Hsu, J.T.; Chen, T.C.; Chiu, C.T.; Yeh, T.S. Comprehensive profiling of virus microRNAs of Epstein-Barr virus-associated gastric carcinoma: Highlighting the interactions of ebv-Bart9 and host tumor cells. J. Gastroenterol. Hepatol. 2017, 32, 82–91. [Google Scholar] [CrossRef] [PubMed]
  66. Jung, Y.J.; Choi, H.; Kim, H.; Lee, S.K. MicroRNA miR-BART20-5p stabilizes Epstein-Barr virus latency by directly targeting BZLF1 and BRLF1. J. Virol. 2014, 88, 9027–9037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Kim, H.; Choi, H.; Lee, S.K. Epstein-Barr virus miR-BART20-5p regulates cell proliferation and apoptosis by targeting BAD. Cancer Lett. 2015, 356, 733–742. [Google Scholar] [CrossRef]
  68. Hooykaas, M.J.G.; van Gent, M.; Soppe, J.A.; Kruse, E.; Boer, I.G.J.; van Leenen, D.; Groot Koerkamp, M.J.A.; Holstege, F.C.P.; Ressing, M.E.; Wiertz, E.; et al. EBV MicroRNA BART16 Suppresses Type I IFN Signaling. J. Immunol. 2017, 198, 4062–4073. [Google Scholar] [CrossRef] [Green Version]
  69. Marquitz, A.R.; Mathur, A.; Nam, C.S.; Raab-Traub, N. The Epstein-Barr Virus BART microRNAs target the pro-apoptotic protein Bim. Virology 2011, 412, 392–400. [Google Scholar] [CrossRef]
  70. Chen, J. Roles of the PI3K/Akt pathway in Epstein-Barr virus-induced cancers and therapeutic implications. World J. Virol. 2012, 1, 154–161. [Google Scholar] [CrossRef]
  71. Scholle, F.; Bendt, K.M.; Raab-Traub, N. Epstein-Barr virus LMP2A transforms epithelial cells, inhibits cell differentiation, and activates Akt. J. Virol. 2000, 74, 10681–10689. [Google Scholar] [CrossRef] [Green Version]
  72. Fukuda, M.; Longnecker, R. Epstein-Barr virus latent membrane protein 2A mediates transformation through constitutive activation of the Ras/PI3-K/Akt Pathway. J. Virol. 2007, 81, 9299–9306. [Google Scholar] [CrossRef] [Green Version]
  73. Xiang, T.; Lin, Y.X.; Ma, W.; Zhang, H.J.; Chen, K.M.; He, G.P.; Zhang, X.; Xu, M.; Feng, Q.S.; Chen, M.Y.; et al. Vasculogenic mimicry formation in EBV-associated epithelial malignancies. Nat. Commun. 2018, 9, 5009. [Google Scholar] [CrossRef] [PubMed]
  74. Du, Y.; Zhang, J.Y.; Gong, L.P.; Feng, Z.Y.; Wang, D.; Pan, Y.H.; Sun, L.P.; Wen, J.Y.; Chen, G.F.; Liang, J.; et al. Hypoxia-induced ebv-circLMP2A promotes angiogenesis in EBV-associated gastric carcinoma through the KHSRP/VHL/HIF1α/VEGFA pathway. Cancer Lett. 2022, 526, 259–272. [Google Scholar] [CrossRef] [PubMed]
  75. Li, W.; Okabe, A.; Usui, G.; Fukuyo, M.; Matsusaka, K.; Rahmutulla, B.; Mano, Y.; Hoshii, T.; Funata, S.; Hiura, N.; et al. Activation of EHF via STAT3 phosphorylation by LMP2A in Epstein-Barr virus-positive gastric cancer. Cancer Sci. 2021, 112, 3349–3362. [Google Scholar] [CrossRef] [PubMed]
  76. Hino, R.; Uozaki, H.; Murakami, N.; Ushiku, T.; Shinozaki, A.; Ishikawa, S.; Morikawa, T.; Nakaya, T.; Sakatani, T.; Takada, K.; et al. Activation of DNA methyltransferase 1 by EBV latent membrane protein 2A leads to promoter hypermethylation of PTEN gene in gastric carcinoma. Cancer Res. 2009, 69, 2766–2774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Liu, X.; Gao, Y.; Luo, B.; Zhao, Y. Construction and Antiapoptosis Activities of Recombinant Adenoviral Expression Vector Carrying EBV Latent Membrane Protein 2A. Gastroenterol. Res. Pract. 2011, 2011, 182832. [Google Scholar] [CrossRef] [PubMed]
  78. Pal, A.D.; Basak, N.P.; Banerjee, A.S.; Banerjee, S. Epstein-Barr virus latent membrane protein-2A alters mitochondrial dynamics promoting cellular migration mediated by Notch signaling pathway. Carcinogenesis 2014, 35, 1592–1601. [Google Scholar] [CrossRef] [Green Version]
  79. Deb Pal, A.; Banerjee, S. Epstein-Barr virus latent membrane protein 2A mediated activation of Sonic Hedgehog pathway induces HLA class Ia downregulation in gastric cancer cells. Virology 2015, 484, 22–32. [Google Scholar] [CrossRef] [Green Version]
  80. Singh, S.; Banerjee, S. Downregulation of HLA-ABC expression through promoter hypermethylation and downmodulation of MIC-A/B surface expression in LMP2A-positive epithelial carcinoma cell lines. Sci. Rep. 2020, 10, 5415. [Google Scholar] [CrossRef] [Green Version]
  81. Wang, W.; Zhang, Y.; Liu, W.; Zhang, X.; Xiao, H.; Zhao, M.; Luo, B. CXCR4 induces cell autophagy and maintains EBV latent infection in EBVaGC. Theranostics 2020, 10, 11549–11561. [Google Scholar] [CrossRef] [PubMed]
  82. Yasui, M.; Kunita, A.; Numakura, S.; Uozaki, H.; Ushiku, T.; Fukayama, M. Cancer stem cells in Epstein-Barr virus-associated gastric carcinoma. Cancer Sci. 2020, 111, 2598–2607. [Google Scholar] [CrossRef]
  83. Seo, J.S.; Jun, S.M.; Kwon, S.W.; Oh, I.H.; Kim, T.G.; Lee, S.K. Establishment and characterization of gastric carcinoma cell clones expressing LMP2A of Epstein-Barr virus. Int. J. Mol. Med. 2010, 25, 11–16. [Google Scholar] [PubMed]
  84. Liu, J.; Zhang, Y.; Yu, C.; Li, J.; Liu, W.; Luo, B. Epstein-Barr Virus-Encoded Latent Membrane Protein 2A Downregulates GCNT3 via the TGF-β1/Smad-mTORC1 Signaling Axis. J. Virol. 2021, 95, e02481-20. [Google Scholar] [CrossRef] [PubMed]
  85. Jiang, Y.; Xiao, H.; Sun, L.; Zhang, Y.; Liu, S.; Luo, B. LMP2A suppresses the role of AHR pathway through ERK signal pathway in EBV-associated gastric cancer. Virus Res. 2021, 297, 198399. [Google Scholar] [CrossRef]
  86. Liu, D.; Shi, D.; Xu, L.; Sun, L.; Liu, S.; Luo, B. LMP2A inhibits the expression of KLF5 through the mTORC1 pathway in EBV-associated gastric carcinoma. Virus Res. 2022, 315, 198792. [Google Scholar] [CrossRef]
  87. Shi, Q.; Zhang, Y.; Liu, W.; Xiao, H.; Qi, Y.; Li, J.; Luo, B. Latent membrane protein 2A inhibits expression level of Smad2 through regulating miR-155-5p in EBV-associated gastric cancer cell lines. J. Med. Virol. 2020, 92, 96–106. [Google Scholar] [CrossRef]
  88. Wang, J.; Liu, W.; Zhang, X.; Zhang, Y.; Xiao, H.; Luo, B. LMP2A induces DNA methylation and expression repression of AQP3 in EBV-associated gastric carcinoma. Virology 2019, 534, 87–95. [Google Scholar] [CrossRef]
  89. Iwakiri, D.; Zhou, L.; Samanta, M.; Matsumoto, M.; Ebihara, T.; Seya, T.; Imai, S.; Fujieda, M.; Kawa, K.; Takada, K. Epstein-Barr virus (EBV)-encoded small RNA is released from EBV-infected cells and activates signaling from Toll-like receptor 3. J. Exp. Med. 2009, 206, 2091–2099. [Google Scholar] [CrossRef] [Green Version]
  90. Ahmed, W.; Philip, P.S.; Tariq, S.; Khan, G. Epstein-Barr virus-encoded small RNAs (EBERs) are present in fractions related to exosomes released by EBV-transformed cells. PLoS ONE 2014, 9, e99163. [Google Scholar] [CrossRef]
  91. Ahmed, W.; Tariq, S.; Khan, G. Tracking EBV-encoded RNAs (EBERs) from the nucleus to the excreted exosomes of B-lymphocytes. Sci. Rep. 2018, 8, 15438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Canitano, A.; Venturi, G.; Borghi, M.; Ammendolia, M.G.; Fais, S. Exosomes released in vitro from Epstein-Barr virus (EBV)-infected cells contain EBV-encoded latent phase mRNAs. Cancer Lett. 2013, 337, 193–199. [Google Scholar] [CrossRef] [PubMed]
  93. Hinata, M.; Kunita, A.; Abe, H.; Morishita, Y.; Sakuma, K.; Yamashita, H.; Seto, Y.; Ushiku, T.; Fukayama, M. Exosomes of Epstein-Barr Virus-Associated Gastric Carcinoma Suppress Dendritic Cell Maturation. Microorganisms 2020, 8, 1776. [Google Scholar] [CrossRef]
  94. Yoon, C.; Kim, J.; Park, G.; Kim, S.; Kim, D.; Hur, D.Y.; Kim, B.; Kim, Y.S. Delivery of miR-155 to retinal pigment epithelial cells mediated by Burkitt’s lymphoma exosomes. Tumor Biol. 2016, 37, 313–321. [Google Scholar] [CrossRef]
  95. Iwakiri, D. Multifunctional non-coding Epstein-Barr virus encoded RNAs (EBERs) contribute to viral pathogenesis. Virus Res. 2016, 212, 30–38. [Google Scholar] [CrossRef]
  96. Chaudhari, P.; Ghate, V.; Nampoothiri, M.; Lewis, S. Multifunctional role of exosomes in viral diseases: From transmission to diagnosis and therapy. Cell Signal. 2022, 94, 110325. [Google Scholar] [CrossRef] [PubMed]
  97. Baglio, S.R.; van Eijndhoven, M.A.; Koppers-Lalic, D.; Berenguer, J.; Lougheed, S.M.; Gibbs, S.; Léveillé, N.; Rinkel, R.N.; Hopmans, E.S.; Swaminathan, S.; et al. Sensing of latent EBV infection through exosomal transfer of 5′pppRNA. Proc. Natl. Acad. Sci. USA 2016, 113, E587–E596. [Google Scholar] [CrossRef] [Green Version]
  98. Burassakarn, A.; Srisathaporn, S.; Pientong, C.; Wongjampa, W.; Vatanasapt, P.; Patarapadungkit, N.; Ekalaksananan, T. Exosomes-carrying Epstein-Barr virus-encoded small RNA-1 induces indoleamine 2, 3-dioxygenase expression in tumor-infiltrating macrophages of oral squamous-cell carcinomas and suppresses T-cell activity by activating RIG-I/IL-6/TNF-α pathway. Oral Oncol. 2021, 117, 105279. [Google Scholar] [CrossRef]
  99. Strong, M.J.; Xu, G.; Coco, J.; Baribault, C.; Vinay, D.S.; Lacey, M.R.; Strong, A.L.; Lehman, T.A.; Seddon, M.B.; Lin, Z.; et al. Differences in gastric carcinoma microenvironment stratify according to EBV infection intensity: Implications for possible immune adjuvant therapy. PLoS Pathog. 2013, 9, e1003341. [Google Scholar] [CrossRef]
  100. Ansari, M.A.; Singh, V.V.; Dutta, S.; Veettil, M.V.; Dutta, D.; Chikoti, L.; Lu, J.; Everly, D.; Chandran, B. Constitutive interferon-inducible protein 16-inflammasome activation during Epstein-Barr virus latency I, II, and III in B and epithelial cells. J. Virol. 2013, 87, 8606–8623. [Google Scholar] [CrossRef] [Green Version]
  101. Meier, U.C.; Cipian, R.C.; Karimi, A.; Ramasamy, R.; Middeldorp, J.M. Cumulative Roles for Epstein-Barr Virus, Human Endogenous Retroviruses, and Human Herpes Virus-6 in Driving an Inflammatory Cascade Underlying MS Pathogenesis. Front. Immunol. 2021, 12, 757302. [Google Scholar] [CrossRef] [PubMed]
  102. Marquitz, A.R.; Mathur, A.; Edwards, R.H.; Raab-Traub, N. Host Gene Expression Is Regulated by Two Types of Noncoding RNAs Transcribed from the Epstein-Barr Virus BamHI A Rightward Transcript Region. J. Virol. 2015, 89, 11256–11268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. De Re, V.; Caggiari, L.; De Zorzi, M.; Fanotto, V.; Miolo, G.; Puglisi, F.; Cannizzaro, R.; Canzonieri, V.; Steffan, A.; Farruggia, P.; et al. Epstein-Barr virus BART microRNAs in EBV- associated Hodgkin lymphoma and gastric cancer. Infect. Agents Cancer 2020, 15, 42. [Google Scholar] [CrossRef] [PubMed]
  104. Cai, L.; Long, Y.; Chong, T.; Cai, W.; Tsang, C.M.; Zhou, X.; Lin, Y.; Ding, T.; Zhou, W.; Zhao, H.; et al. EBV-miR-BART7-3p Imposes Stemness in Nasopharyngeal Carcinoma Cells by Suppressing SMAD7. Front. Genet. 2019, 10, 939. [Google Scholar] [CrossRef] [Green Version]
  105. Cai, L.M.; Lyu, X.M.; Luo, W.R.; Cui, X.F.; Ye, Y.F.; Yuan, C.C.; Peng, Q.X.; Wu, D.H.; Liu, T.F.; Wang, E.; et al. EBV-miR-BART7-3p promotes the EMT and metastasis of nasopharyngeal carcinoma cells by suppressing the tumor suppressor PTEN. Oncogene 2015, 34, 2156–2166. [Google Scholar] [CrossRef] [Green Version]
  106. Mrad, M.F.; Saba, E.S.; Nakib, L.; Khoury, S.J. Exosomes from Subjects with Multiple Sclerosis Express EBV-Derived Proteins and Activate Monocyte-Derived Macrophages. Neurol. Neuroimmunol. Neuroinflamm. 2021, 8, 18. [Google Scholar] [CrossRef] [PubMed]
  107. Raab-Traub, N. Nasopharyngeal Carcinoma: An Evolving Role for the Epstein-Barr Virus. Curr. Top. Microbiol. Immunol. 2015, 390, 339–363. [Google Scholar] [CrossRef]
  108. Ikeda, M.; Longnecker, R. Cholesterol is critical for Epstein-Barr virus latent membrane protein 2A trafficking and protein stability. Virology 2007, 360, 461–468. [Google Scholar] [CrossRef]
  109. DeKroon, R.M.; Gunawardena, H.P.; Edwards, R.; Raab-Traub, N. Global Proteomic Changes Induced by the Epstein-Barr Virus Oncoproteins Latent Membrane Protein 1 and 2A. mBio 2018, 9, e00959-18. [Google Scholar] [CrossRef] [Green Version]
  110. Hu, Y.; Tian, Y.; Di, H.; Xue, C.; Zheng, Y.; Hu, B.; Lin, Q.; Yan, X. Noninvasive Diagnosis of Nasopharyngeal Carcinoma Based on Phenotypic Profiling of Viral and Tumor Markers on Plasma Extracellular Vesicles. Anal. Chem. 2022, 94, 9740–9749. [Google Scholar] [CrossRef]
  111. Sato, Y.; Ochiai, S.; Murata, T.; Kanda, T.; Goshima, F.; Kimura, H. Elimination of LMP1-expressing cells from a monolayer of gastric cancer AGS cells. Oncotarget 2017, 8, 39345–39355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Ng, P.C.; Lam, H.S. Biomarkers for late-onset neonatal sepsis: Cytokines and beyond. Clin. Perinatol. 2010, 37, 599–610. [Google Scholar] [CrossRef]
  113. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef] [PubMed]
  114. Ramayanti, O.; Verkuijlen, S.; Novianti, P.; Scheepbouwer, C.; Misovic, B.; Koppers-Lalic, D.; van Weering, J.; Beckers, L.; Adham, M.; Martorelli, D.; et al. Vesicle-bound EBV-BART13-3p miRNA in circulation distinguishes nasopharyngeal from other head and neck cancer and asymptomatic EBV-infections. Int. J. Cancer 2019, 144, 2555–2566. [Google Scholar] [CrossRef] [Green Version]
  115. Liu, L.; Zuo, L.; Yang, J.; Xin, S.; Zhang, J.; Zhou, J.; Li, G.; Tang, J.; Lu, J. Exosomal cyclophilin A as a novel noninvasive biomarker for Epstein-Barr virus associated nasopharyngeal carcinoma. Cancer Med. 2019, 8, 3142–3151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Teow, S.-Y.; Peh, S.-C. Exosomes as the Promising Biomarker for Epstein-Barr Virus (EBV)-Associated Cancers. In Novel Implications of Exosomes in Diagnosis and Treatment of Cancer and Infectious Diseases; BoD: Paris, France, 2017. [Google Scholar]
  117. Shoda, K.; Ichikawa, D.; Fujita, Y.; Masuda, K.; Hiramoto, H.; Hamada, J.; Arita, T.; Konishi, H.; Kosuga, T.; Komatsu, S.; et al. Clinical utility of circulating cell-free Epstein-Barr virus DNA in patients with gastric cancer. Oncotarget 2017, 8, 28796–28804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Lo, Y.M. Quantitative analysis of Epstein-Barr virus DNA in plasma and serum: Applications to tumor detection and monitoring. Ann. N. Y. Acad. Sci. 2001, 945, 68–72. [Google Scholar] [CrossRef]
  119. Kang, B.W.; Choi, Y.; Kwon, O.K.; Lee, S.S.; Chung, H.Y.; Yu, W.; Bae, H.I.; Seo, A.N.; Kang, H.; Lee, S.K.; et al. High level of viral microRNA-BART20-5p expression is associated with worse survival of patients with Epstein-Barr virus-associated gastric cancer. Oncotarget 2017, 8, 14988–14994. [Google Scholar] [CrossRef] [Green Version]
  120. Gong, L.P.; Chen, J.N.; Dong, M.; Xiao, Z.D.; Feng, Z.Y.; Pan, Y.H.; Zhang, Y.; Du, Y.; Zhang, J.Y.; Bi, Y.H.; et al. Epstein-Barr virus-derived circular RNA LMP2A induces stemness in EBV-associated gastric cancer. EMBO Rep. 2020, 21, e49689. [Google Scholar] [CrossRef]
  121. Nakayama, A.; Abe, H.; Kunita, A.; Saito, R.; Kanda, T.; Yamashita, H.; Seto, Y.; Ishikawa, S.; Fukayama, M. Viral loads correlate with upregulation of PD-L1 and worse patient prognosis in Epstein-Barr Virus-associated gastric carcinoma. PLoS ONE 2019, 14, e0211358. [Google Scholar] [CrossRef]
  122. Moon, S.H.; Park, N.S.; Noh, M.H.; Kim, Y.S.; Cheong, S.H.; Hur, D.Y. Olaparib-induced Apoptosis Through EBNA1-ATR-p38 MAPK Signaling Pathway in Epstein-Barr Virus-positive Gastric Cancer Cells. Anticancer Res. 2022, 42, 555–563. [Google Scholar] [CrossRef] [PubMed]
  123. Shin, J.Y.; Kim, J.O.; Lee, S.K.; Chae, H.S.; Kang, J.H. LY294002 may overcome 5-FU resistance via down-regulation of activated p-AKT in Epstein-Barr virus-positive gastric cancer cells. BMC Cancer 2010, 10, 425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef] [PubMed]
  125. Mendt, M.; Kamerkar, S.; Sugimoto, H.; McAndrews, K.M.; Wu, C.C.; Gagea, M.; Yang, S.; Blanko, E.V.R.; Peng, Q.; Ma, X.; et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight 2018, 3, e99263. [Google Scholar] [CrossRef] [PubMed]
  126. Zhu, X.; Badawi, M.; Pomeroy, S.; Sutaria, D.S.; Xie, Z.; Baek, A.; Jiang, J.; Elgamal, O.A.; Mo, X.; Perle, K.; et al. Comprehensive toxicity and immunogenicity studies reveal minimal effects in mice following sustained dosing of extracellular vesicles derived from HEK293T cells. J. Extracell. Vesicles 2017, 6, 1324730. [Google Scholar] [CrossRef] [Green Version]
  127. Fitts, C.A.; Ji, N.; Li, Y.; Tan, C. Exploiting Exosomes in Cancer Liquid Biopsies and Drug Delivery. Adv. Healthc. Mater. 2019, 8, e1801268. [Google Scholar] [CrossRef]
  128. Barile, L.; Vassalli, G. Exosomes: Therapy delivery tools and biomarkers of diseases. Pharmacol. Ther. 2017, 174, 63–78. [Google Scholar] [CrossRef] [Green Version]
  129. Ferguson, S.W.; Nguyen, J. Exosomes as therapeutics: The implications of molecular composition and exosomal heterogeneity. J. Control. Release 2016, 228, 179–190. [Google Scholar] [CrossRef]
  130. Liao, W.; Du, Y.; Zhang, C.; Pan, F.; Yao, Y.; Zhang, T.; Peng, Q. Exosomes: The next generation of endogenous nanomaterials for advanced drug delivery and therapy. Acta Biomater. 2019, 86, 1–14. [Google Scholar] [CrossRef]
  131. Wang, X.; Xiang, Z.; Liu, Y.; Huang, C.; Pei, Y.; Wang, X.; Zhi, H.; Wong, W.H.; Wei, H.; Ng, I.O.; et al. Exosomes derived from Vδ2-T cells control Epstein-Barr virus-associated tumors and induce T cell antitumor immunity. Sci. Transl. Med. 2020, 12, eaaz3426. [Google Scholar] [CrossRef]
  132. Wang, X.; Zhang, Y.; Mu, X.; Tu, C.R.; Chung, Y.; Tsao, S.W.; Chan, G.C.; Leung, W.H.; Lau, Y.L.; Liu, Y.; et al. Exosomes derived from γδ-T cells synergize with radiotherapy and preserve antitumor activities against nasopharyngeal carcinoma in immunosuppressive microenvironment. J. Immunother. Cancer 2022, 10, e003832. [Google Scholar] [CrossRef] [PubMed]
  133. Kim, S.T.; Cristescu, R.; Bass, A.J.; Kim, K.M.; Odegaard, J.I.; Kim, K.; Liu, X.Q.; Sher, X.; Jung, H.; Lee, M.; et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat. Med. 2018, 24, 1449–1458. [Google Scholar] [CrossRef] [PubMed]
  134. Xie, T.; Liu, Y.; Zhang, Z.; Zhang, X.; Gong, J.; Qi, C.; Li, J.; Shen, L.; Peng, Z. Positive Status of Epstein-Barr Virus as a Biomarker for Gastric Cancer Immunotherapy: A Prospective Observational Study. J. Immunother. 2020, 43, 139–144. [Google Scholar] [CrossRef]
  135. Masetti, M.; Lindinger, M.; Lorenzen, S. Programmed Cell Death Protein 1 Blockade Elicits Ongoing Remission in 2 Cases of Refractory Epstein-Barr Virus-Associated Metastatic Gastric Carcinoma. Oncol. Res. Treat. 2022, 45, 375–379. [Google Scholar] [CrossRef]
  136. Wang, J.; Ge, J.; Wang, Y.; Xiong, F.; Guo, J.; Jiang, X.; Zhang, L.; Deng, X.; Gong, Z.; Zhang, S.; et al. EBV miRNAs BART11 and BART17-3p promote immune escape through the enhancer-mediated transcription of PD-L1. Nat. Commun. 2022, 13, 866. [Google Scholar] [CrossRef]
  137. Moon, J.W.; Kong, S.K.; Kim, B.S.; Kim, H.J.; Lim, H.; Noh, K.; Kim, Y.; Choi, J.W.; Lee, J.H.; Kim, Y.S. IFNγ induces PD-L1 overexpression by JAK2/STAT1/IRF-1 signaling in EBV-positive gastric carcinoma. Sci. Rep. 2017, 7, 17810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Ye, L.; Zhu, Z.; Chen, X.; Zhang, H.; Huang, J.; Gu, S.; Zhao, X. The Importance of Exosomal PD-L1 in Cancer Progression and Its Potential as a Therapeutic Target. Cells 2021, 10, 3247. [Google Scholar] [CrossRef]
Figure 1. The process of epithelial EBV infection and impacts of exosomes in EBV-associated gastric cancer. (1) EBV itself or EBV-infected B cells cause EBV infection in the gastric epithelial cells. (2) TGF-β released from EBV-infected gastric epithelial cells induces expression of TGF-β receptors on B cells and (3) causes lysis of B cells leading to efficient viral transmission into epithelial cells. (4) EBV DNA transported to the nucleus undergoes (5) circulation and chromatinization. (6) Interferon gamma inducible protein 16 (IFI16) detects EBV DNA, and (7) recruits ASC and Pro-Casp-1 to form an inflammasome complex. (8) The inflammasome translocates to the cytoplasm and activates Casp-1, (9) which cleavage pro-ILs into ILs. (10) Viral transcription products (EBERs and miR-BARTs), (11) translation products (LMP2A and EBNA1), or other products (ILs, Casp-1, and IFI16) are (12) transported into recipient cells via exosomes. Abbreviations: ICAM-1, intracellular adhesion molecule-1; NMHC IIA, non-muscle myosin heavy chain IIA; IFI16, interferon gamma inducible protein 16; ASC, apoptosis-associated speck-like protein with a caspase recruitment domain; Casp-1, caspase-1; ILs, interleukins; EXO, exosome.
Figure 1. The process of epithelial EBV infection and impacts of exosomes in EBV-associated gastric cancer. (1) EBV itself or EBV-infected B cells cause EBV infection in the gastric epithelial cells. (2) TGF-β released from EBV-infected gastric epithelial cells induces expression of TGF-β receptors on B cells and (3) causes lysis of B cells leading to efficient viral transmission into epithelial cells. (4) EBV DNA transported to the nucleus undergoes (5) circulation and chromatinization. (6) Interferon gamma inducible protein 16 (IFI16) detects EBV DNA, and (7) recruits ASC and Pro-Casp-1 to form an inflammasome complex. (8) The inflammasome translocates to the cytoplasm and activates Casp-1, (9) which cleavage pro-ILs into ILs. (10) Viral transcription products (EBERs and miR-BARTs), (11) translation products (LMP2A and EBNA1), or other products (ILs, Casp-1, and IFI16) are (12) transported into recipient cells via exosomes. Abbreviations: ICAM-1, intracellular adhesion molecule-1; NMHC IIA, non-muscle myosin heavy chain IIA; IFI16, interferon gamma inducible protein 16; ASC, apoptosis-associated speck-like protein with a caspase recruitment domain; Casp-1, caspase-1; ILs, interleukins; EXO, exosome.
Cancers 15 00469 g001
Table 1. EBV genes and their functional roles in EBV-associated gastric cancer.
Table 1. EBV genes and their functional roles in EBV-associated gastric cancer.
GeneRolesRef
EBERConsistent insulin growth factor-1 expression and promotion of cell proliferation[43]
Increases chemoresistance and promotes cell migration[44]
Involved in carcinogenesis by downregulation of E-cadherin expression[45]
Underscores neoplastic transformation by deregulated PAR1b activation[46]
EBNA-1Promotes tumorigenicity, growth ability, immune evasion, and reduce chemosensitivity[47]
Induces impaired responses to DNA damage and promotes cell survival[48]
Maintains latent replication and persistence[49,50]
Leads to modification in alternative splicing profiles[51]
Involved in extensive methylation of the whole genome by upregulating DNMT3a[52]
miR-BARTsInvolved in the initiation and development of EBVaGC[53,54]
miR-BART1-3p suppresses apoptosis and promotes the migration of cancer cells[55]
miR-BART1-3p regulates CXCL10, one of the key elements of EBVaGC[56]
miR-BART1-5p inhibits cell proliferation and migration[57]
miR-BART3-3p promotes tumorigenesis by regulating the senescence pathway[58]
miR-BART4-5p reduces apoptosis[59]
miR-BART5-3p maintains EBV latency and contributes to tumorigenesis[60]
miR-BART5-5p contributes to antiapoptosis, proliferation, invasion, tumor cell migration, and immune escape[61]
miR-BART15-3p increases apoptosis by targeting BRUCE[62]
miR-BART15-3p increases chemosensitivity to 5-FU[63]
miR-BART17-5p promotes migration and anchorage-independent growth[64]
miR-BART9 plays a role during carcinogenesis through EMT[65]
miR-BART20-5p stabilizes latent infection and regulates cell proliferation and apoptosis[66,67]
miR-BART16 facilitates the establishment of latent EBV infection[68]
miR-BART clusters I and II are involved in protection from apoptosis[69]
LMP2AIncreases epithelial cell growth and differentiation with activation of the PI3K-AKT pathway[70,71,72]
Contributes to vasculogenic mimicry formation[73]
Promotes angiogenesis under hypoxia[74]
Contributes to tumorigenesis via phosphorylation of STAT3[75,76]
Inhibits apoptosis with cell cycle arrest[77]
Increases cell invasion and chemoresistance with the IL-6-STAT3 pathway[44]
Elevates cell migration and metastasis by the Notch signaling pathway[78]
Involved in immune evasion by activation of the Sonic Hedgehog pathway[79,80]
Involved in autophagy and EBV replication[81]
Maintains cancer stem cells in EBVaGC with NF-kB pathway[82]
Downregulates cell proliferation[83,84,85]
Downregulates expression of GCNT3 and KLF5, which promotes cell proliferation and migration[84,86,87]
Suppresses aryl hydrocarbon receptor pathway, which promotes cell proliferation and migration[85]
Promotes malignancy through epigenetic modifications by hypermethylation of AQP3 promoter[88]
SNHG8Involved in cell proliferation, colony formation, and cell growth by cell cycle regulation[40]
Table 2. Potential diagnostic and prognostic biomarkers of EBVaGC.
Table 2. Potential diagnostic and prognostic biomarkers of EBVaGC.
Gene/PathwaySpecimenClinical SignificanceRef
EBV DNAsPlasmaHigh in EBVaGC with high sensitivity and high specificity
monitoring after treatment and determination of relapse
[117,118]
miR-BART20-5pFFPEAssociated with poor recurrence-free survival in EBVaGC[119]
Circular RNA LMP2AFFPE, cell lineAssociated with distant metastasis and poor prognosis[74,120]
EBV-copy number/genomeFFPE, cell lineAssociated with PD-L1 expression and poor disease-specific survival in EBVaGC[121]
PI3K/Akt activationCell lineMay induce drug resistance to chemotherapy[70,123]
ATR/MAPK phosphorylationCell lineIncrease susceptibility to olaparib[122]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, B.; Kim, K.-M. Role of Exosomes and Their Potential as Biomarkers in Epstein-Barr Virus-Associated Gastric Cancer. Cancers 2023, 15, 469. https://doi.org/10.3390/cancers15020469

AMA Style

Kim B, Kim K-M. Role of Exosomes and Their Potential as Biomarkers in Epstein-Barr Virus-Associated Gastric Cancer. Cancers. 2023; 15(2):469. https://doi.org/10.3390/cancers15020469

Chicago/Turabian Style

Kim, Binnari, and Kyoung-Mee Kim. 2023. "Role of Exosomes and Their Potential as Biomarkers in Epstein-Barr Virus-Associated Gastric Cancer" Cancers 15, no. 2: 469. https://doi.org/10.3390/cancers15020469

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop