Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter Open Access June 3, 2017

Role of Epstein-Barr virus in the development of nasopharyngeal carcinoma

  • Hui Zhang , Jing Wang , Dan Yu , Yan Liu EMAIL logo , Kai Xue and Xue Zhao
From the journal Open Medicine

Abstract

Southern China experiences larger extent of total cancer pathologies, of which nasopharyngeal carcinoma has the highest incidence under otorhinolaryngeal malignant carcinomas. Risk factor of nasopharyngeal carcinoma varies from hereditary causes to virus infection, among which Epstein-Barr virus (EBV) infection is the mostly investigated. The study into mechanism of EBV in occurrence, development and prognosis of nasopharyngeal carcinoma has been studied for several decades. The pathophysiology in making of EBV into a cancerogen includes proteins as latent membrane protein 1 (LMPs) and nucleic acids as micro-RNAs. In this paper, we reviewed till date studies focusing on relationship between EBV and nasopharyngeal carcinoma.

1 Introduction

According to recent research, 80% of nasopharyngeal carcinomas (NPC) in China are found in southern geographical areas. The incidence of NPC in male is two to four times higher than that in female. Radiotherapy is often treatment of choice while the prognosis is not satisfying ascribed to relapse and cause early metastasis. Causes of NPC are not explicit, while mainstream opinions are that NPC is closely related with latent Epstein-Barr virus (EBV) infection, hereditary factors and environmental factors are also responsible for many kinds of lymphomas and epithelial tumors and hence can modulate mechanisms affecting carcinogenesis, proliferation, apoptosis, death and migration of cells, epigenetically change lymphocyte-specific processes and induce cell immortalization. Mechanisms underlying the carcinogenic effects of EBV involve LMP1, LMP2, microRNA and other molecules that we will introduce in the following sections [1-3].

2 LMP1

EBV-encoded latent membrane protein 1 (LMP1) is a 66-KD integral membrane protein that is closely associated with poor prognosis of NPC. Therefore, LMP1 is considered behind the fact that EBV happens to modulate most of the cell processes including migration, proliferation, metabolism and tumorigenesis through alternation of various kind of target proteins, RNAs and signaling pathways.

Endothelial cell specific molecule (endocan, or called Esm-1), was found in 52% of NPC specimens. Endocan could stimulate migration and invasion of endothelial cells, and indicates a shorter survival in NPC patients. Endocan can be upregulated by LMP1 through the LMP1-activated NF-κB, MEK-ERK and JNK signaling pathways [4]. The phosphorylation of insulin-like growth factor 1 receptor (IGF1R) can be altered by LMP1, which depends on activation of NF-κB signaling pathway and could be suppressed by IκBα and TRAF6. These contributes to the transformation of epithelial cells induced by LMP1 [5]. Through phosphorylation and degradation of IκBα, LMP1 activates NFκB signaling pathway [6].

Phosphatase and tension homolog (PTEN) is a major tumor suppressor. LMP1 can induce a DNA methylation of PTEN via DNMT3b transcription up-regulated by LMP1-mediated NF-κB. Thus tumor suppressor PTEN is silenced at the cellular and molecular level [7]. Expression of tumor necrosis factor α-induced protein 2 (TNFAIP2) is high in NPC tissues. This over-expression is transcriptionally induced by LMP1 through C-terminal-activating region (CTAR2) domain of TNFAIP2.NF-κB participated in this process through a NF-κB-binding site within the TNFAIP2 promoter. this enhances the expression of TNFAIP2 which further induce cell motility and thus contributes to promoting NPC tumor progression [8]. Glucose transporter-1 (Glut-1) is one of the direct target genes of NF-κB signaling that is down-stream of activation of mTORC1 by LMP1. LMP1-inducedNF-κB activation leads to upregulation of Glut-1 transcription and growth of NPC cells, which results in aerobic glycolysis, cell proliferation and colony formation [9]. Through Toll-like receptor 3 (TLR3), EBERs induce inflammatory response in which macrophages are recruited in NPC cells. EBERs, LMP1 with NF-κB form a positive regulatory loop that amplifies the inflammatory signals, which leads to a favorable microenvironment for solid tumor growth [10].

As a signal transducer and a transcriptional activator of many essential genes, the important roles of STAT3 in tumor generation, progression, metastasis and drug-resistance have been thoroughly investigated. LMP1 was found to be able to cause phosphorylation of STAT3 through activation of Janus kinase 3 (JAK3) and extracellular signal-regulated kinase (ERK) and then stimulated STAT3 nuclear accumulation [11]. Bcl-3 induction was mediated by this activation of STAT3. Through its carboxyl-terminal activation domain 1 (CTAR1), LMP1 activates both STAT3 and EGFR [12]. n NPC cells, level and stability of transcription of the HIF-1α were significantly enhanced by LMP1 via interaction with the ERK1/2 and STAT3 signaling pathways through CTAR1 and CTAR3. ERK1/2/NF-κB pathway recruited by LMP1 CTAR1 also facilitated HIF-1A promoter activity [13]. Through the JNKs/c-Jun signaling pathway, VEGF expression is increased by LMP1 [14].

Survivin is an inhibitor of apoptosis protein which is specifically expressed in tumor tissues and is related with proliferation of tumor cells. Expression of Survivin could be promoted by LMP1 in G0/G1, S and G2/M phase. LMP1 could also trigger accumulation of Survivin and CDK4 in nuclei and thus keep tumor cells from apoptosis. The function of Survivin is known to be closely connected with tumor suppressor gene P53. P53 protein levels were reduced byLMP1 through the increase in the polyubiquitination of p53 in NPC cells [15, 16].

MicroRNAs (miRNAs) are a collection of endogenous non-coding small RNAs found in eukaryocyte that regulate cell behaviors such as proliferation, apoptosis and tumor progression through binding to target RNAs. LMP1 could suppress miR-1 expression, of which K-ras is found to be a novel direct target. By repressing K-ras expression, tumour-suppressive effects of miR-1 was suppressed by LMP1 [17]. MicroRNA-21, a biomarker for chemo-resistance, its expression is triggered by LMP1 via the PI3K/Akt/ FOXO3a pathway, which lead to the expression of PDCD4 and Fas-L, and at last results in chemo-resistance in NPC cells [18]. Aberrant expression of miR-155, which significantly increased in radio-resistant NPC tissues, can also be induced by LMP. Ubiqulin-1 expressionis negatively correlated to MiR-155. The axisof miR-155-UBQLN1 could affected some important genes regulating cell proliferation, cycling, migration and invasion through PI3K/Akt pathway. As well, up-regulation of miR-155 in NPC driven by LMP1 lead to a downregulation of, which results in poor prognosis of NPC patients [19, 20]. Mir-204 inhibited invasion and metastasis of NPC cells partly through targeting cdc42. In NPC cells and tissues, miR-204 is found to be down-regulated, which indicates a more aggressive phenotype of NPC and poor prognostic. By activating Stat-3, LMP-1 suppressed miR-204 expression [21]. LMP1 and transcription factor Twist-1 is also associated with miR-10b that was significantly up-regulated in NPC.MiR-10b is related with young age and advanced clinical stage [22].

The repair of DNA double-strand breaks (DSBs) is repressed by LMP1 through inhibiting phosphorylation and activity of DNA-dependent protein kinase (DNA-PK). LMP1 could also reduce the phosphorylation of AMP-activated protein kinase (AMPK), which is associated with glycolysis and resistance to apoptosis mediated by LMP1. The AMPKα (Thr172) reduction is a predictive factor for poorer clinical outcomes of radiation therapy in NPC patients [23]. The LKB1-AMPK pathway has anti-tumor activity via modulation of energy metabolism. LMP1-mediated AMPK inactivation is related to the proliferation and transformation of epithelial cells, which implicates the LMP1-driven pathogenesis of NPC [24].

LMP1 increases glucose and glutamine uptake in NPC cell, stimulates LDHA activity and production of lactate, while reduces pyruvate kinase activity and pyruvate concentrations. PKM2, LDHA and FGFR1 phosphorylation, as well as PDHK1, FGFR1, c-Myc and HIF-1α expression, are also increased by LMP1 [25].

Rho GTPases, such as Cdc42 and Cdc2, are associated with actin cytoskeleton reorganization, which modulates cell morphology and motility and tumorigenesis. LMP1 can enhance FGD4 activity toward Cdc42, resulting in actin cytoskeleton rearrangement and motility increase of NPC cells [26]. LMP1 can also regulate Op18/stathmin signaling by cdc2 mediation and affect tumor phenotype and metastasis [27].

Mammalian target of rapamycin (mTOR) is a serine/ threonine protein kinase which is involved in guideline of cell proliferation, differentiation and cell cycle. Many aberrant expression of proteins in mTOR signaling pathway are important for tumor occurrence. Through phosphorylation of AKT/mTOR/P70S6K/4EBP1, LMP1 can upregulate the mTOR signaling pathway in NPC cell. Expression of genes in the mTOR pathway such as p-P70S6K, p-4EBP1 are significantly correlated with overall survival of NPC patients [28].

Transcription coactivator TAZ is a member of Hippo-related pathways, which can restrict cell proliferation and induce cell apoptosis. In a recent study, for LMP1-mediated cell proliferation, cancer stem cell-like properties and EMT, TAZ, frequently expressed in LMP1-positive NPC, plays an important role, which provide new insights into oncogenic mechanism of LMP1 [29].

ATOH8 is a transcript factor among the basic helix-loop-helix (bHLH) gene family. LMP1 can impair the occupancy of activated H3K4me3 and enhance the repressive occupancy of H3K27me3 on ATOH8 promoter, which leads to ATOH8 expression inhibition that promote malignant phenotype of NPC [30].

Ezrin, a membrane cross-linker protein, takes parts in signal transduction and phagocytosis of tumor cells and thus implicated in tumor cell metastasis through interaction with cell adhesion molecules. Recent data showed that LMP1-stimulated cell motility and invasion of NPC require the phosphorylation and recruitment of ezrin [31].

Mitogen- and Stress-Activated Kinase 1 (MSK1) is a nuclear kinase that is important for cell proliferation. High level of phosphorylated MSK1 is observed in poorly differentiated NPC tissues. LMP1-promoted cell proliferation is associated with increased MSK1 activity, which may be correlated with Fra-1 and c-Jun induction by it through phosphorylation of histone H3 [32].

Fibronectin is a high molecular weight glycoprotein. As an extracellular matrix protein, it is important in an adhensive growth of cells and is closely related with occurrence, development and prognosis of tumors. Induction of activin A and TGFβ1 and JNK/SAPK signaling are required for LMP1-mediated expression of fibronectin. The expression and activation of the major fibronectin receptor, α5β1 integrin, is also induced by LMP1. Thus, these proteins contribute in the pathogenesis of LMP1-positive NPC by increase the metastatic potential of epithelial cells [33]. Aside of fibronectin, LMP-1 could induce cell surface interactions involving integrin-α5 and N-cadherin as well and promote EMT of NPC [34].

Several stemness-related gene can be up-regulated by LMP1 and lead to increase of the cell number of side population (SP), enhanced self-renewal properties and tumor initiation ability in vivo. Cancer stem cell (CSC) marker CD44 and radio-resistance are also regulated by LMP1, which might be the results of inactivation of DNA damage response (DDR) proteins including ATM, Chk1, Chk2 and p53 in EBV-positive NPC cells.[35] Phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) pathway also plays an important role in the CSC properties induction and maintenance in NPC. LMP1, PI3K/AKT, miR-21 and PTEN could constitute a positive feedback loop that regulates LMP1-induced CSCs in NPC cell.[36]Some sequence variations of LMP1 may lead to a potential escape from host cell immune recognition, protecting latent EBV infection and causing an increase in tumorigenicity [37].

Learning from above, LMP1 is so important to EBV-associated NPC that anti-LMP1 HELA/CAR-T cells, LMP1-targeted DNA enzyme and human antibody Fab against LMP1 conjugated with mitomycin C (MMC) can all control NPC development in vitro and in vivo [38-40].

3 MicroRNAs

MiRNAs are small non-coding RNAs which through negatively regulating gene expression post-transcriptionally, mediate cell proliferation, apoptosis, and carcinogenesis. In NPC infected by EBV, few viral proteins are expressed but high levels of BamHI-A rightward transcripts (BARTs) are found to be anticipating in cell functions. EBV-encoded BART-miRNAs, including long noncoding RNAs (lncRNAs) and BART microRNAs (miRNAs) are closely related to EBV pathogenesis in NPC [41, 42].

MiR-BART3, miR-BART7 and miR-BART13 microRNAs are detected to be at abundant levels and regularly secreted extracellular of NPC cells which may make them new biomarkers for diagnosis and clinical predictors of NPC, and circulating examination shows that miR-BART17-5p can be a potential biomarker of a poor prognosis in post-treatment detections [43, 44].

EBV-miR-BART1 is highly expressed in NPC. Reduction of PTEN directly mediated by EBV-miR-BART1 activates PTEN-dependent pathways, PI3K-Akt, FAK-p130(Cas) and Shc-MAPK/ERK1/2 signaling included. As a result, migration, invasion and metastasis of NPC cells increases, and thus drive EMT [45]. EBV-miR-BART1 could also up- and down-modulate a number of metabolism-associated genes, such as PSAT1 and PHGDH [46].

BART promoters can be activated and the expression of BARTs can be modulated by NF-κB in EBV-infected NPC cells. NF-κB activity is correlated with expression of BART miRNAs and lnc RNAs in EBV-infected epithelial cells [47].

EBV-miR-BART7-3p is highly expressed in NPC cells. Through targeting PTEN and modulating PI3K/Akt/GSK-3β signaling, EBV-miR-BART7-3penhances cell migration/ invasion, metastasis and EMT, leading to gain of mesenchymal features and loss of epithelial markers in NPC cells. That makes it correlated positively with node metastasis and clinical stage of NPC [48].

Over-expression of EBV-miR-BART10-3p is found in clinical samples which is correlated with poor prognosis. EBV-miR-BART10-3p directly modulates BTRC gene that encodes beta-transducin repeat containing E3 ubiquitin protein ligase (βTrCP). Through targeting BTRC and regulating the expression of the β-catenin and Snail down-stream substrates, EBV-miR-BART10-3p promote the invasion and migration of NPC cells and lead to EMT [49].

Forkhead box P1 (FOXP1) plays a key role in monocyte to macrophage differentiation. EBV-miR-BART11 promotes inflammation-induced NPC carcinogenesis by directly targeting FOXP1 gene and inhibiting TAM differentiation mediated by FOXP1, and induce inflammatory cytokines secretion into the tumor microenvironment [50].

4 Other molecules

EBV-encoded Latent Membrane Protein 2A (LMP2A) is regularly expressed in NPC. Recently studies indicate that-LMP2A expression interferes with Syk tyrosine kinase and integrin α6β4 interaction by competitive binding to Syk, which is associated with migration and invasive property of LMP2A-expressing NPC [51].

Epstein-Barr virus-encoded latent membrane protein 2A (LMP2A) is an oncoprotein of EB virus and a wellknown NPC activator which could increase tumor invasion through promotion of the epithelial-mesenchymal transition (EMT) of NPC. Overexpression of metastatic tumor antigen 1 (MTA1) significantly correlated with tumor metastasis via the Wnt1 pathway and β-catenin activation. A molecular connection between LMP2 and MTA1 has been established. LMP2A reinforces EMT by induce the expression of MTA1 via activation of the mTOR pathway and 4EBP1-eIF4E axis in NPC [52].

EBNA1 protein is a nuclear protein encoded by EB virus that is highly expressed in NPC tissues, and its expression was associated with transcription of EB virus and NPC lymph node metastasis. Expression of microRNA 200a (miR-200a) and miR-200b can be inhibited by over-expressed EBNA1 that is mediated by transforming growth factor-β1. Expression of target genes of these microRNAs, zinc finger E-box binding homeobox 1 (ZEB1) and ZEB2, are up-regulated, which in turn affect NPC cell morphology and the expression of EMT markers [53].

Peptidyl-prolyl-cis-trans isomerase NIMA-interacting 1 (PIN1) is found to be consistently expressed in almost all EBV-associated NPC cells. It is a vital regulator in cell survival and apoptosis through isomerizing specific phosphorylated amino acid residues. Suppression of PIN1 can lead to inhibition of cyclin D1 expression and activation of caspase-3 in NPC cells and restrain tumor growth. At the same time, PIN1 can regulate proliferation, colony formation and anchorage-independent growth of NPC cell [54].

In NPC cells, the EBV immediate-early protein BZLF1 plays a key role in transformation of EBV infection from latent to lytic forms, and the later form is implicated in human carcinogenesis. BZLF1 functions to bind with several DNA damage response (DDR) proteins and thus impair DNA damage repair and abrogate G2/M checkpoint, which induced genomic instability. In summary, BZLF1 contributes to the carcinogenesis of EBV-associated epithelial malignancies by induction of mis-localization of important DDR proteins, and BZLF1 may be the connection of lytic EBV infection with impaired DNA damage repair [55].

5 Conclusions

EBV plays an essential role in the development of NPC and targeting EBV may a great help in treatment of NPC. And that claims deep insight of underlying mechanisms of NPC derived from EBV infection. Associations of LMP1, EBNA and microRNAs with NPC cell behaviors and important signaling pathways have been elucidated. But targeted therapy has not been exploited, which makes further investigation still in need.

  1. Conflict of interest No authors report any conflict of interest.

References

[1] Myers JS. Review complementary and integrative interventions for cancer-related cognitive changes. Asia-Pacific journal of oncology nursing. 2015;2(4):215-22610.4103/2347-5625.162825Search in Google Scholar PubMed PubMed Central

[2] Zaghloul MS, Eldebawy E, Ahmed S, Ammar H, Khalib E, Abdelrahman H, Zekri W, Elzomor H, Taha H, Elnashar A. Does primary tumor volume predict the outcome of pediatric nasopharyngeal carcinoma?: A prospective single-arm study using neoadjuvant chemotherapy and concomitant chemotherapy with intensity modulated radiotherapy. Asia-Pacific Journal of Clinical Oncology. 2016;12(2):810.1111/ajco.12460Search in Google Scholar PubMed

[3] Hsu N-Y, Lee H, Yen Y, Cheng Y-W. Human papillomavirus and non-small cell lung cancer. Thoracic Cancer. 2013;4(4):345-35310.1111/1759-7714.12030Search in Google Scholar PubMed

[4] Yu PH, Chou SF, Chen CL, Hung H, Lai CY, Yang PM, et al. Upregulation of endocan by Epstein-Barr virus latent membrane protein 1 and its clinical significance in nasopharyngeal carcinoma. PloS one. 2013;8(12):e8225410.1371/journal.pone.0082254Search in Google Scholar PubMed PubMed Central

[5] Tworkoski K, Raab-Traub N. LMP1 promotes expression of insulin-like growth factor 1 (IGF1) to selectively activate IGF1 receptor and drive cell proliferation. Journal of virology. 2015;89(5):2590-260210.1128/JVI.02921-14Search in Google Scholar PubMed PubMed Central

[6] Yin L, Liao W, Deng X, Tang M, Gu H, Li X, et al. LMP1 activates NF-kappa B via degradation of I kappa B alpha in nasopharyngeal carcinoma cells. Chinese medical journal. 2001;114(7):718-722Search in Google Scholar

[7] Peng H, Chen Y, Gong P, Cai L, Lyu X, Jiang Q, et al. Higher methylation intensity induced by EBV LMP1 via NF-kappaB/DNMT3b signaling contributes to silencing of PTEN gene. Oncotarget. 2016;7(26):40025-4003710.18632/oncotarget.9474Search in Google Scholar PubMed PubMed Central

[8] Chen CC, Liu HP, Chao M, Liang Y, Tsang NM, Huang HY, et al. NF-kappaB-mediated transcriptional upregulation of TNFAIP2 by the Epstein-Barr virus oncoprotein, LMP1, promotes cell motility in nasopharyngeal carcinoma. Oncogene. 2014;33(28):3648-365910.1038/onc.2013.345Search in Google Scholar PubMed

[9] Zhang J, Jia L, Lin W, Yip YL, Lo KW, Lau VM, et al. Epstein-Barr Virus encoded Latent Membrane Protein-1 upregulates glucose transporter-1 transcription via the mTORC1/NF-kappaB signaling pathways. Journal of virology. 201710.1128/JVI.02168-16Search in Google Scholar PubMed PubMed Central

[10] Li Z, Duan Y, Cheng S, Chen Y, Hu Y, Zhang L, et al. EBV-encoded RNA via TLR3 induces inflammation in nasopharyngeal carcinoma. Oncotarget. 2015;6(27):24291-2430310.18632/oncotarget.4552Search in Google Scholar PubMed PubMed Central

[11] Liu YP, Tan YN, Wang ZL, Zeng L, Lu ZX, Li LL, et al. Phosphorylation and nuclear translocation of STAT3 regulated by the Epstein-Barr virus latent membrane protein 1 in nasopharyngeal carcinoma. International journal of molecular medicine. 2008;21(2):153-16210.3892/ijmm.21.2.153Search in Google Scholar

[12] Kung CP, Meckes DG, Jr., Raab-Traub N. Epstein-Barr virus LMP1 activates EGFR, STAT3, and ERK through effects on PKCdelta. Journal of virology. 2011;85(9):4399-440810.1128/JVI.01703-10Search in Google Scholar PubMed PubMed Central

[13] Sung WW, Chu YC, Chen PR, Liao MH, Lee JW. Positive regulation of HIF-1A expression by EBV oncoprotein LMP1 in nasopharyngeal carcinoma cells. Cancer letters. 2016;382(1):21-3110.1016/j.canlet.2016.08.021Search in Google Scholar PubMed

[14] Yang L, Liu L, Xu Z, Liao W, Feng D, Dong X, et al. EBV-LMP1 targeted DNAzyme enhances radiosensitivity by inhibiting tumor angiogenesis via the JNKs/HIF-1 pathway in nasopharyngeal carcinoma. Oncotarget. 2015;6(8):5804-581710.18632/oncotarget.3331Search in Google Scholar PubMed PubMed Central

[15] Guo L, Tang M, Yang L, Xiao L, Bode AM, Li L, et al. Epstein-Barr virus oncoprotein LMP1 mediates survivin upregulation by p53 contributing to G1/S cell cycle progression in nasopharyngeal carcinoma. International journal of molecular medicine. 2012;29(4):574-58010.3892/ijmm.2012.889Search in Google Scholar PubMed PubMed Central

[16] Ai MD, Li LL, Zhao XR, Wu Y, Gong JP, Cao Y. Regulation of survivin and CDK4 by Epstein-Barr virus encoded latent membrane protein 1 in nasopharyngeal carcinoma cell lines. Cell research. 2005;15(10):777-78410.1038/sj.cr.7290347Search in Google Scholar PubMed

[17] Chen X, Shi J, Zhong J, Huang Z, Luo X, Huang Y, et al. miR-1, regulated by LMP1, suppresses tumour growth and metastasis by targeting K-ras in nasopharyngeal carcinoma. International journal of experimental pathology. 2015;96(6):427-43210.1111/iep.12162Search in Google Scholar PubMed PubMed Central

[18] Yang GD, Huang TJ, Peng LX, Yang CF, Liu RY, Huang HB, et al. Epstein-Barr Virus_Encoded LMP1 upregulates microRNA-21 to promote the resistance of nasopharyngeal carcinoma cells to cisplatin-induced Apoptosis by suppressing PDCD4 and Fas-L. PloS one. 2013;8(10):e7835510.1371/journal.pone.0078355Search in Google Scholar PubMed PubMed Central

[19] Yang F, Liu Q, Hu CM. Epstein-Barr virus-encoded LMP1 increases miR-155 expression, which promotes radioresistance of nasopharyngeal carcinoma via suppressing UBQLN1. European review for medical and pharmacological sciences. 2015;19(23):4507-4515Search in Google Scholar

[20] Du ZM, Hu LF, Wang HY, Yan LX, Zeng YX, Shao JY, et al. Upregulation of MiR-155 in nasopharyngeal carcinoma is partly driven by LMP1 and LMP2A and downregulates a negative prognostic marker JMJD1A. PloS one. 2011;6(4):e1913710.1371/journal.pone.0019137Search in Google Scholar PubMed PubMed Central

[21] Ma L, Deng X, Wu M, Zhang G, Huang J. Down-regulation of miRNA-204 by LMP-1 enhances CDC42 activity and facilitates invasion of EBV-associated nasopharyngeal carcinoma cells. FEBS letters. 2014;588(9):1562-157010.1016/j.febslet.2014.02.039Search in Google Scholar PubMed

[22] Allaya N, Khabir A, Sallemi-Boudawara T, Sellami N, Daoud J, Ghorbel A, et al. Over-expression of miR-10b in NPC patients: correlation with LMP1 and Twist1. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. 2015;36(5):3807-381410.1007/s13277-014-3022-6Search in Google Scholar PubMed

[23] Lu J, Tang M, Li H, Xu Z, Weng X, Li J, et al. EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma. Cancer letters. 2016;380(1):191-20010.1016/j.canlet.2016.05.032Search in Google Scholar PubMed

[24] Lo AK, Lo KW, Ko CW, Young LS, Dawson CW. Inhibition of the LKB1-AMPK pathway by the Epstein-Barr virus-encoded LMP1 promotes proliferation and transformation of human nasopharyngeal epithelial cells. The Journal of pathology. 2013;230(3):336-34610.1002/path.4201Search in Google Scholar PubMed

[25] Lo AK, Dawson CW, Young LS, Ko CW, Hau PM, Lo KW. Activation of the FGFR1 signalling pathway by the Epstein-Barr virus-encoded LMP1 promotes aerobic glycolysis and transformation of human nasopharyngeal epithelial cells. The Journal of pathology. 2015;237(2):238-24810.1002/path.4575Search in Google Scholar PubMed

[26] Liu HP, Chen CC, Wu CC, Huang YC, Liu SC, Liang Y, et al. Epstein-Barr virus-encoded LMP1 interacts with FGD4 to activate Cdc42 and thereby promote migration of nasopharyngeal carcinoma cells. PLoS pathogens. 2012;8(5):e100269010.1371/journal.ppat.1002690Search in Google Scholar PubMed PubMed Central

[27] Lin X, Liu S, Luo X, Ma X, Guo L, Li L, et al. EBV-encoded LMP1 regulates Op18/stathmin signaling pathway by cdc2 mediation in nasopharyngeal carcinoma cells. International journal of cancer. 2009;124(5):1020-102710.1002/ijc.23767Search in Google Scholar PubMed

[28] Chen J, Hu CF, Hou JH, Shao Q, Yan LX, Zhu XF, et al. Epstein-Barr virus encoded latent membrane protein 1 regulates mTOR signaling pathway genes which predict poor prognosis of nasopharyngeal carcinoma. Journal of translational medicine. 2010;8:3010.1186/1479-5876-8-30Search in Google Scholar PubMed PubMed Central

[29] He J, Tang F, Liu L, Chen L, Li J, Ou D, et al. Positive regulation of TAZ expression by EBV-LMP1 contributes to cell proliferation and epithelial-mesenchymal transition in nasopharyngeal carcinoma. Oncotarget. 201610.18632/oncotarget.13775Search in Google Scholar PubMed PubMed Central

[30] Wang Z, Xie J, Yan M, Wang J, Wang X, Zhang J, et al. Downregulation of ATOH8 induced by EBV-encoded LMP1 contributes to the malignant phenotype of nasopharyngeal carcinoma. Oncotarget. 2016;7(18):26765-2677910.18632/oncotarget.8503Search in Google Scholar PubMed PubMed Central

[31] Endo K, Kondo S, Shackleford J, Horikawa T, Kitagawa N, Yoshizaki T, et al. Phosphorylated ezrin is associated with EBV latent membrane protein 1 in nasopharyngeal carcinoma and induces cell migration. Oncogene. 2009;28(14):1725-173510.1038/onc.2009.20Search in Google Scholar PubMed PubMed Central

[32] Li B, Wan Z, Huang G, Huang Z, Zhang X, Liao D, et al. Mitogen- and stress-activated Kinase 1 mediates Epstein-Barr virus latent membrane protein 1-promoted cell transformation in nasopharyngeal carcinoma through its induction of Fra-1 and c-Jun genes. BMC cancer. 2015;15:39010.1186/s12885-015-1398-3Search in Google Scholar PubMed PubMed Central

[33] Morris MA, Dawson CW, Laverick L, Davis AM, Dudman JP, Raveenthiraraj S, et al. The Epstein-Barr virus encoded LMP1 oncoprotein modulates cell adhesion via regulation of activin A/TGFbeta and beta1 integrin signalling. Scientific reports. 2016;6:1953310.1038/srep19533Search in Google Scholar PubMed PubMed Central

[34] Wasil LR, Shair KH. Epstein-Barr virus LMP1 induces focal adhesions and epithelial cell migration through effects on integrin-alpha5 and N-cadherin. Oncogenesis. 2015;4:e17110.1038/oncsis.2015.31Search in Google Scholar PubMed PubMed Central

[35] Yang CF, Peng LX, Huang TJ, Yang GD, Chu QQ, Liang YY, et al. Cancer stem-like cell characteristics induced by EB virus-encoded LMP1 contribute to radioresistance in nasopharyngeal carcinoma by suppressing the p53-mediated apoptosis pathway. Cancer letters. 2014;344(2):260-27110.1016/j.canlet.2013.11.006Search in Google Scholar PubMed

[36] Yang CF, Yang GD, Huang TJ, Li R, Chu QQ, Xu L, et al. EB-virus latent membrane protein 1 potentiates the stemness of nasopharyngeal carcinoma via preferential activation of PI3K/AKT pathway by a positive feedback loop. Oncogene. 2016;35(26):3419-343110.1038/onc.2015.402Search in Google Scholar PubMed

[37] Tang YL, Lu JH, Cao L, Wu MH, Peng SP, Zhou HD, et al. Genetic variations of EBV-LMP1 from nasopharyngeal carcinoma biopsies: potential loss of T cell epitopes. Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas. 2008;41(2):110-11610.1590/S0100-879X2008000200006Search in Google Scholar

[38] Tang X, Zhou Y, Li W, Tang Q, Chen R, Zhu J, et al. T cells expressing a LMP1-specific chimeric antigen receptor mediate antitumor effects against LMP1-positive nasopharyngeal carcinoma cells in vitro and in vivo. Journal of biomedical research. 2014;28(6):468-475Search in Google Scholar

[39] Ke X, Yang YC, Hong SL. EBV-LMP1-targeted DNAzyme restrains nasopharyngeal carcinoma growth in a mouse C666-1 xenograft model. Medical oncology. 2011;28 Suppl 1:S326-3210.1007/s12032-010-9681-2Search in Google Scholar PubMed

[40] Chen R, Zhang D, Mao Y, Zhu J, Ming H, Wen J, et al. A human Fab-based immunoconjugate specific for the LMP1 extracellular domain inhibits nasopharyngeal carcinoma growth in vitro and in vivo. Molecular cancer therapeutics. 2012;11(3):594-60310.1158/1535-7163.MCT-11-0725Search in Google Scholar PubMed

[41] Zeng Z, Huang H, Huang L, Sun M, Yan Q, Song Y, et al. Regulation network and expression profiles of Epstein-Barr virus-encoded microRNAs and their potential target host genes in nasopharyngeal carcinomas. Science China Life sciences. 2014;57(3):315-32610.1007/s11427-013-4577-ySearch in Google Scholar PubMed

[42] Szeto CY, Lin CH, Choi SC, Yip TT, Ngan RK, Tsao GS, et al. Integrated mRNA and microRNA transcriptome sequencing characterizes sequence variants and mRNA-microRNA regulatory network in nasopharyngeal carcinoma model systems. FEBS open bio. 2014;4:128-14010.1016/j.fob.2014.01.004Search in Google Scholar PubMed PubMed Central

[43] Zhang G, Zong J, Lin S, Verhoeven RJ, Tong S, Chen Y, et al. Circulating Epstein-Barr virus microRNAs miR-BART7 and miR-BART13 as biomarkers for nasopharyngeal carcinoma diagnosis and treatment. International journal of cancer. 2015;136(5):E301-31210.1002/ijc.29206Search in Google Scholar PubMed

[44] Hirai N, Wakisaka N, Kondo S, Aga M, Moriyama-Kita M, Ueno T, et al. Potential Interest in Circulating miR-BART17-5p As a Post-Treatment Biomarker for Prediction of Recurrence in Epstein-Barr Virus-Related Nasopharyngeal Carcinoma. PloS one. 2016;11(9):e016360910.1371/journal.pone.0163609Search in Google Scholar PubMed PubMed Central

[45] Cai L, Ye Y, Jiang Q, Chen Y, Lyu X, Li J, et al. Epstein-Barr virus-encoded microRNA BART1 induces tumour metastasis by regulating PTEN-dependent pathways in nasopharyngeal carcinoma. Nature communications. 2015;6:735310.1038/ncomms8353Search in Google Scholar PubMed PubMed Central

[46] Ye Y, Zhou Y, Zhang L, Chen Y, Lyu X, Cai L, et al. EBV-miR-BART1 is involved in regulating metabolism-associated genes in nasopharyngeal carcinoma. Biochemical and biophysical research communications. 2013;436(1):19-2410.1016/j.bbrc.2013.05.008Search in Google Scholar PubMed

[47] Verhoeven RJ, Tong S, Zhang G, Zong J, Chen Y, Jin DY, et al. NF-kappaB Signaling Regulates Expression of Epstein-Barr Virus BART MicroRNAs and Long Noncoding RNAs in Nasopharyngeal Carcinoma. Journal of virology. 2016;90(14):6475-648810.1128/JVI.00613-16Search in Google Scholar PubMed PubMed Central

[48] Cai LM, Lyu XM, Luo WR, Cui XF, Ye YF, Yuan CC, et al. EBV-miR-BART7-3p promotes the EMT and metastasis of nasopharyngeal carcinoma cells by suppressing the tumor suppressor PTEN. Oncogene. 2015;34(17):2156-216610.1038/onc.2014.341Search in Google Scholar PubMed

[49] Yan Q, Zeng Z, Gong Z, Zhang W, Li X, He B, et al. EBV-miR-BART10-3p facilitates epithelial-mesenchymal transition and promotes metastasis of nasopharyngeal carcinoma by targeting BTRC. Oncotarget. 2015;6(39):41766-4178210.18632/oncotarget.6155Search in Google Scholar PubMed PubMed Central

[50] Song Y, Li X, Zeng Z, Li Q, Gong Z, Liao Q, et al. Epstein-Barr virus encoded miR-BART11 promotes inflammation-induced carcinogenesis by targeting FOXP1. Oncotarget. 2016;7(24):36783-3679910.18632/oncotarget.9170Search in Google Scholar PubMed PubMed Central

[51] Zhou X, Matskova L, Rathje LS, Xiao X, Gish G, Werner M, et al. SYK interaction with ITGbeta4 suppressed by Epstein-Barr virus LMP2A modulates migration and invasion of nasopharyngeal carcinoma cells. Oncogene. 2015;34(34):4491-449910.1038/onc.2014.380Search in Google Scholar PubMed

[52] Lin Z, Wan X, Jiang R, Deng L, Gao Y, Tang J, et al. Epstein-Barr virus-encoded latent membrane protein 2A promotes the epithelial-mesenchymal transition in nasopharyngeal carcinoma via metastatic tumor antigen 1 and mechanistic target of rapamycin signaling induction. Journal of virology. 2014;88(20):11872-1188510.1128/JVI.01867-14Search in Google Scholar PubMed PubMed Central

[53] Wang L, Tian WD, Xu X, Nie B, Lu J, Liu X, et al. Epstein-Barr virus nuclear antigen 1 (EBNA1) protein induction of epithelial-mesenchymal transition in nasopharyngeal carcinoma cells. Cancer. 2014;120(3):363-37210.1002/cncr.28418Search in Google Scholar PubMed

[54] Xu M, Cheung CC, Chow C, Lun SW, Cheung ST, Lo KW. Overexpression of PIN1 Enhances Cancer Growth and Aggressiveness with Cyclin D1 Induction in EBV-Associated Nasopharyngeal Carcinoma. PloS one. 2016;11(6):e015683310.1371/journal.pone.0156833Search in Google Scholar PubMed PubMed Central

[55] Yang J, Deng W, Hau PM, Liu J, Lau VM, Cheung AL, et al. Epstein-Barr virus BZLF1 protein impairs accumulation of host DNA damage proteins at damage sites in response to DNA damage. Laboratory investigation; a journal of technical methods and pathology. 2015;95(8):937-95010.1038/labinvest.2015.69Search in Google Scholar PubMed

Received: 2017-2-20
Accepted: 2017-3-14
Published Online: 2017-6-3

© 2017 Hui Zhang et al.

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 3.0 License.

Downloaded on 23.4.2024 from https://www.degruyter.com/document/doi/10.1515/med-2017-0025/html
Scroll to top button