Role of Exosomes in Epstein-Barr Virus-Associated Gastric Cancer: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: ,

Exosomes are considered to be involved in the pathogenesis of various diseases, including cancer, viral infections, and autoimmune diseases. 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. 

  • exosome
  • EBV
  • gastric cancer
  • biomarker
  • immunotherapy

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].

2. 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 [22], suggesting that clonal growth of EBV-infected gastric epithelial cells results in carcinoma rather than augmenting EBV infection of gastric epithelial cells per se [23][24]. 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) [22]. 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 [25][26][27]. Moreover, the knockdown of SNHG8 inhibits cell proliferation and colony formation, arrests the G0/G1 phase in vitro, and suppresses tumor growth in vivo [27]. 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 [28][29]. 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.
Table 1. EBV genes and their functional roles in EBV-associated gastric cancer.

3. 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 [76][77][78]. A previous study characterized exosomes released from EBV-infected cells, including latency I and III cell lines [79]. 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 [79]. Further, EBV infection alters exosome contents [80]. 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 [80]. The expression level of miR-155 in recipient epithelial cells was also increased in the presence of EBV [81].
These genetic materials are incorporated into cells via exosomes, suggesting that they might function through exosomal transfer [82][83]. Furthermore, accumulating evidence suggests that this exosomal transfer affects the phenotype of recipient cells and influences the microenvironment surrounding the infected cells [79][82]. 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 [84]; furthermore, exosomes derived from EBV-positive gastric cancer cell lines suppress dendritic cell maturation [80]. 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 [82]. 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 [85]. They can further create an immunosuppressive microenvironment that influences the T-cell immune response [85]. IDO1, an effective immunosuppressive enzyme, is upregulated in EBVaGC, indicating the possibility of an immune evasion strategy [86]. 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 [87]. 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 [87]. Excessive stimulation and uncontrolled proliferation of EBV-infected B cells have been suggested to induce T-cell exhaustion, allowing escape from T-cell surveillance [88]. Furthermore, exosomal IL-1β may promote EBV persistence owing to its inability to recruit neutrophils [87]. In summary, EBV may trigger inflammasome activity through exosomal transfer in the pathogenesis of EBVaGC.

4. 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][89]. 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 [90]. 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 [90]. In latent AGS cells, host cell transcription analysis by RNA-seq revealed that many downregulated genes were BART miRNA targets [89]; this was also observed when a full-length cDNA clone of one of the BART isoforms was artificially expressed in EBV-negative AGS cells [89]. 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 [46]. 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 [91][92], 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 [93], 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 [88]. 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 [94], 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 [95]. 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 [96]. Further, LMP2A in exosomes can be used for diagnosing NPC with high accuracy [97]. 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 [98]. 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.

This entry is adapted from the peer-reviewed paper 10.3390/cancers15020469

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.
  2. Zhang, L.; Yu, D. Exosomes in cancer development, metastasis, and immunity. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 455–468.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  14. Zhao, M.; Nanbo, A.; Sun, L.; Lin, Z. Extracellular Vesicles in Epstein-Barr Virus’ Life Cycle and Pathogenesis. Microorganisms 2019, 7, 48.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  22. Iizasa, H.; Nanbo, A.; Nishikawa, J.; Jinushi, M.; Yoshiyama, H. Epstein-Barr Virus (EBV)-associated gastric carcinoma. Viruses 2012, 4, 3420–3439.
  23. 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.
  24. 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.
  25. 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. . Zhonghua Bing Li Xue Za Zhi 2017, 46, 84–87.
  26. 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.
  27. 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.
  28. 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.
  29. 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.
  30. 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.
  31. 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.
  32. 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.
  33. 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.
  34. 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.
  35. 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.
  36. Leight, E.R.; Sugden, B. EBNA-1: A protein pivotal to latent infection by Epstein-Barr virus. Rev. Med. Virol. 2000, 10, 83–100.
  37. 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.
  38. 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.
  39. 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.
  40. 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.
  41. 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.
  42. 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.
  43. 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.
  44. 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.
  45. 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.
  46. 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.
  47. 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.
  48. 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.
  49. 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.
  50. 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.
  51. 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.
  52. 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.
  53. 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.
  54. 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.
  55. 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.
  56. 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.
  57. Chen, J. Roles of the PI3K/Akt pathway in Epstein-Barr virus-induced cancers and therapeutic implications. World J. Virol. 2012, 1, 154–161.
  58. 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.
  59. 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.
  60. 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.
  61. 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.
  62. 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.
  63. 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.
  64. 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.
  65. 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.
  66. 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.
  67. 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.
  68. 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.
  69. 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.
  70. 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.
  71. 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.
  72. 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.
  73. 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.
  74. 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.
  75. 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.
  76. 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.
  77. 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.
  78. 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.
  79. 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.
  80. 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.
  81. 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.
  82. Iwakiri, D. Multifunctional non-coding Epstein-Barr virus encoded RNAs (EBERs) contribute to viral pathogenesis. Virus Res. 2016, 212, 30–38.
  83. 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.
  84. 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.
  85. 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.
  86. 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.
  87. 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.
  88. 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.
  89. 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.
  90. 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.
  91. 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.
  92. 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.
  93. 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.
  94. Raab-Traub, N. Nasopharyngeal Carcinoma: An Evolving Role for the Epstein-Barr Virus. Curr. Top. Microbiol. Immunol. 2015, 390, 339–363.
  95. Ikeda, M.; Longnecker, R. Cholesterol is critical for Epstein-Barr virus latent membrane protein 2A trafficking and protein stability. Virology 2007, 360, 461–468.
  96. 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.
  97. 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.
  98. 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.
More
This entry is offline, you can click here to edit this entry!
Video Production Service