Open Access

Epstein‑Barr virus‑encoded microRNA BART7 downregulates major histocompatibility complex class I chain‑related peptide A and reduces the cytotoxicity of natural killer cells to nasopharyngeal carcinoma

  • Authors:
    • Thian‑Sze Wong
    • Siqi Chen
    • Min‑Juan Zhang
    • Jimmy Yu‑Wai Chan
    • Wei Gao
  • View Affiliations

  • Published online on: June 28, 2018     https://doi.org/10.3892/ol.2018.9041
  • Pages: 2887-2892
  • Copyright: © Wong et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Evasion from natural killer (NK) cell surveillance enables cancer to proliferate and spread at the early stages. NK cells mediate specific cytolysis by activation of the triggering receptors on their cell surface, of which the communication between natural killer group 2, member D (NKG2D) and major histocompatibility complex class I chain‑related peptide A (MICA) is a key regulatory axis. It has been indicated that cancer cells can reduce the surface expression of MICA, which thereby reduces the cytotoxicity of NK cells. In nasopharyngeal carcinoma (NPC), however, the underlying mechanism remains unclear. The present study indicated that MICA expression in NPC was regulated by TGFβ1. Furthermore, the human MICA gene was demonstrated to contain the c‑Myc binding site in the promoter region. Notably, the results suggested that TGFβ1 upregulated MICA expression by promoting c‑Myc expression. Additionally, the findings demosntrated that TGFβ1 expression in NPC was negatively controlled by Epstein‑Barr virus‑encoded microRNA BART7 (ebv‑miR‑BART7). In ebv‑miR‑BART7‑expressing NPC, the TGFβ1/c‑Myc/MICA regulatory axis was significantly inhibited. Notably, functional analysis indicated that NPC cells expressing ebv‑miR‑BART7 were less sensitive to the cytolysis mediated by NK cells. In conclusion, the present results revealed that ebv‑miR‑BART7‑expressing NPC may impair NK cells recognition and activity, which suggests that targeting ebv‑miR‑BART7 may be a useful therapeutic strategy in NPC immunotherapy.

Introduction

Nasopharyngeal carcinoma (NPC) is a unique head and neck cancer as the pathogenesis of cancer is closely linked with Epstein-Barr virus (EBV). Guangdong province of southern China has the highest NPC incidence and EBV-associated NPC is the major histological type (1). EBV infection could be found in the normal individuals. In immunocompetent EBV carrier, EBV is in the latency 0 which does not trigger innate immune response (2). When the virus enters lytic phases, many viral gene products could interfere with the innate and adaptive response which protects the cells from detection and elimination by the immune effectors (3). EBV in NPC is in latency II stages. The characteristic viral gene products in EBV latency II can exert immunomodulatory functions and contribute to the immunoevasion to the host.

Natural killer (NK) cell is a cytotoxic effector in human innate system with the capability to kill tumor cells. Evasion from NK cell surveillance enables cancer to proliferate and spread at early stage (4). NK cells mediate target specific cytolysis by activation of the triggering receptors on their cell surface. Of which, the communication between immunoreceptor natural killer group 2, member D (NKG2D) and major histocompatibility complex class I chain-related peptide A (MICA) is a key regulatory axis. NKG2D is a c-type lectin-like receptor expressed on the NK cell surface (5). MICA is the NKG2D ligand expressed on transformed cells surface. NKG2D is an activating receptor of NK cells. Binding of MICA will trigger the activating signals and antitumor response of NK cells (6).

In NPC, it has been shown that the cytotoxic activity of NK cells in NPC patients were reduced in comparison with the normal individuals (7). NK cells can recognize cancer with high surface MICA expression. However, the cancer cells can suppress MICA expression which reduces their susceptibility to NK cell cytolysis. For NPC, the underlying mechanism remains unclear. MICA polymorphism (MICA-A9 and A5.1) is associated with the NPC risk in the Southern China male NPC patients (8,9). In liver cancer, it has been shown that MICA expression is regulated by microRNA (10). MicroRNA is short non-coding RNA which functions as post-transcriptional regulators to specific gene expression. In view of the close association between EBV and NPC, we speculate that EBV-encoded microRNA has a regulatory role on MICA expression in NPC. It has been shown that MICA expression could be upregulated by transforming growth factor β 1 (TGFβ1) (11). As our previous findings indicate that the EBV-encoded microRNA BART7 (ebv-miR-BART7) is a functional TGFβ1 suppressor (12), we hypothesized that NPC expressing ebv-miR-BART7 has lower MICA expression and has reduced sensitivity to NK cell cytolysis.

Materials and methods

Cell lines

NPC cell line HK1 was maintained in PRMI-1640 medium supplemented with 10% fetal bovine serum, 200 Unit/ml penicillin G sodium, 20 µg/ml streptomycin sulfate, and 0.5 µg/ml amphotericin B (all Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA). NK92MI cells were incubated in Alpha Minimum Essential medium supplemented with 12.5% fetal bovine serum, 12.5% horse serum (both Gibco; Thermo Fisher Scientific, Inc.), 0.2 mM inositol, 0.1 mM 2-mercaptoethanol, 0.02 mM folic acid (all Sigma, St. Louis, MO, USA), 200 Unit/ml penicillin G sodium, 20 µg/ml streptomycin sulfate, and 0.5 µg/ml amphotericin B (all Gibco; Thermo Fisher Scientific, Inc.).

Recombinant TGFβ1 treatment

HK1 cells were treated by recombinant TGFβ1 (Thermo Fisher Scientific, Inc.) as previously described (12). HK1 was incubated with 0.5 ng/ml recombinant TGFβ1 for 72 h.

RNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted with TRIzol (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. RNA was converted to cDNA using High Capacity cDNA Reverse Transcription kit (Thermo Fisher Scientific, Inc.) following the supplier's guideline. RT-qPCR was carried out using FastStart Universal Probe Master on a LightCycler® 480 (both Roche Applied Science, Penzberg, Germany). The primers and probes were designed from Universal ProbeLibrary Assay Design Center (http://www.roche-applied-science.com/). Primers and probes were as follows: MICA-forward, 5′-GGCATCTTCCCTTTTGCAC-3′; MICA-reverse, 5′-GGACAGCACCGTGAGGTTAT-3′; MICA-probe, cat. no. 24; c-Myc-forward, 5′-GCTGCTTAGACGCTGGATTT-3′; c-Myc-reverse, 5′-TAACGTTGAGGGGCATCG-3′; c-Myc-probe, cat. no. 66; TGFβ1-forward, 5′-ACTACTACGCCAAGGAGGTCAC-3′; TGFβ1-reverse, 5′-TGCTTGAACTTGTCATAGATTTCG-3′; TGFβ1-probe, cat. no. 27. qPCR reaction was carried out at 95°C for 10 min followed by 45 cycles of 95°C for 15 sec and 60°C for 1 min. Expression levels were calculated using the comparative threshold cycle method (2−ΔΔCT).

BART7 mimic transfection

HK1 cells were transfected with 3 nM ebv-miR-BART7 mimic or negative control siRNA using Hiperfect transfection reagent (all Qiagen Inc., Valencia, CA, USA). After 72 h, the expression levels of TGFβ1, MICA and c-Myc were determined.

Immunocytochemistry (ICC)

HK1 cells were seed on chamber slides and treated with recombinant TGFβ1 or transfected with ebv-miR-BART7 mimic. NPC cells were washed with PBS and fixed with 4% paraformaldehyde. Then, NPC cells were incubated with anti-MICA antibodies (Abcam, Cambridge, UK), followed by the addition of CF488A Secondary Antibody Conjugates (Biotium, Inc., Freemont, CA, USA). Blue-fluorescent 4′,6-Diamidino-2-Phenylindole (DAPI; Thermo Fisher Scientific, Inc.) was used to label nucleus.

Chromatin immunoprecipitation (ChIP) assay

ChIP assay was performed using Magna ChIP A Chromatin Immunoprecipitation kit (EMD Millipore Billerica, MA, USA) following the manufacturer's protocol. In brief, HK1 cells were fixed with 1% paraformaldehyde and were lysed by cell lysis buffer. Then, cells were sonicated to shear cross-linked DNA to approximately 200–1,000 base pairs. The cell lysates were incubated overnight at 4°C with protein A magnetic beads and anti-c-Myc antibodies or normal mouse IgG. Protein/DNA complex were eluted from the beads and free DNA was obtained by reverse cross-links of protein/DNA complex. The precipitated MICA promoter DNA was detected by RT-PCR. The primers spanning the c-Myc binding site of MICA promoter were as follows: Forward, 5′-GGTGGGATAGGGTGAGGAGA-3′; reverse, 5′-CCCCATCTGCTGAATGTCAC-3′. The RT-PCR products were analyzed by QIAxcel Advanced system (Qiagen Inc.), a capillary electrophoresis device.

NK cell cytotoxicity test

NK cell cytotoxicity test was performed using the xCELLigence system (13). HK1 cells were transfected with ebv-miR-BART7 mimic or negative control siRNA followed by seeded on the E16 plate. Then, NK92MI cells were added to NPC cells and survival of NPC cells was continuously monitored. The percentage of cytotoxicity was calculated from the formula: Percentage of cytotoxicity=[(cell index no effector-cell index effector)/cell index no effector] x100%.

Statistical analysis

All results were shown as mean ± SD from 3 independent experiments. Student's t test was used to evaluate the differences between control and experimental groups. P<0.05 was considered statistically significant.

Results

TGFβ1 was implicated in MICA expression in NPC

To study whether TGFβ1 regulated MICA expression, NPC cells were first treated with recombinant TGFβ1. RT-qPCR analysis showed that MICA expression was transcriptionally un-regulated in HK1 cells (Fig. 1A). Consistently, subsequent immunostaining indicated that surface expression of MICA in HK1 were remarkably increased in response to TGFβ1 treatment (Fig. 1B).

C-Myc bound to the promoter region of MICA

To investigate the regulatory mechanism of MICA transcription, we performed promoter analysis and identified potential c-Myc binding site (CATGTG) in the promoter region of MICA gene (Fig. 2A). As shown in the ChIP assay, c-Myc protein could bind to the predicted c-Myc binding site located upstream of the transcription start site of MICA gene (Fig. 2B).

TGFβ1 activated c-Myc expression and the TGFβ1/c-Myc regulatory axis in NPC was inhibited by ebv-miR-BART7

To examine whether TGFβ1 could regulate c-Myc at transcriptional level, we examined the change of c-Myc expression in the TGFβ1-treated NPC cell lines. As shown in Fig. 3A, c-Myc expression was significantly increased in HK1 cells after treatment with recombinant TGFβ1. The expression of TGFβ1 was increased in HK1 cells treated with recombinant TGFβ1 (Fig. 3B). Recombinant TGFβ1 treatment did not affect the expression level of ebv-miR-BART7 (Fig. 3C). We have previously shown that ebv-mir-BART7 expression had a significant negative regulatory function on TGFβ1 expression in NPC (12). Thus, we anticipated that ebv-miR-BART7 expression in the NPC cells will also suppress c-Myc expression. To confirm our hypothesis, we transfected the NPC cell line with synthetic ebv-miR-BART7 microRNA and measured the change of TGFβ1/c-Myc expression. The expression of ebv-miR-BART7 was significantly increased in HK1 cells transfected with BART7 mimic, indicating the successful overexpression of ebv-miR-BART7 (Fig. 3D). NPC cells expressing ebv-miR-BART7 showed significant reduction of TGFβ1/c-Myc as compared with the mock transfectant (Fig. 3E and F).

MICA expression was remarkably reduced in ebv-miR-BART7-expressing NPC

To confirm the importance of ebv-miR-BART7 on MICA expression, changes of MICA expression in ebv-miR-BART7-expressing NPC cell line were performed. As shown in Fig. 4, ebv-miR-BART7 had a significant suppressive effect on MICA mRNA and protein expression in the NPC cell line (Fig. 4).

Ebv-miR-BART7-expressing NPC cells exhibited reduced sensitivity to NK cell cytolysis

To explore whether ebv-miR-BART7 expression will reduce the sensitivity of NPC cells to NK cell lysis, ebv-miR-BART7-expressing NPC cells and the mock transfectants were co-cultured with NK cell line NK92MI. When NPC cells reached their logarithmic growth phase, NK92MI cells were added to NPC cells at effector to target (E:T) ratio of 5:1. For HK1 cells, a significant lysis of target cells was observed at 20 h after the addition of NK92MI cells (Fig. 5). Ebv-miR-BART7-expressing HK1 cells were less lysed by NK92MI cells in comparison with control cells (Fig. 5), indicating that forced expression of ebv-miR-BART7 reduced the sensitivity of NPC cells to NK92MI cells.

Discussion

Reduced expression of major histocompatibility complex MHC class I protein on the cell surface is associated with the oncogenic transformation (14). MICA belongs to the MHC class I family and impairment of MHC class I expression is common in viral infected cancers. MICA can specifically bind to surface NKG2D on different immune cells such as NK cells and CD8αβ T cells and activates their immune functions (15,16). Therefore, suppressing MICA expression could prevent the viral infected cancer cells from immune attack.

TGFβ singling is important for maintaining the epithelial homeostasis and immunity (17). Disruption of TGFβ signaling is implicated in the pathogenesis of head and neck cancers (18). Recently, it was reported that TGFβ1 could inhibit mTOR activity of NK cells, leading to reduced metabolic activity and impaired cytotoxic activity of NK cells (19). TGFβ1 also reduced the expression of NK cell surface receptors including CD122 and IL-15RB, which might confer decreased metabolic activity (19). To identify the soluble factors released by B-acute lymphoblastic leukaemia (ALL) blasts that could impair function of NK cells, Rouce et al compared the levels of known soluble factors upregulated in other cancers between ALL blasts cultured with NK cells and healthy donor B cells cultured with NK cells (20). They found that TGFβ1 was the most notably increased soluble factor. Moreover, TGFβ1 suppressed the expression of activating receptors including NKG2D, NKp30 and NKp46 and enhanced the expression of inhibitory receptor NKG2A of NK cells, resulting in ALL-mediated NK cell dysfunction (20). We speculate that TGFβ can interrupt the interaction of NKG2D (on NK cell surface) and MICA in cancer patients in view of the fact that elevated TGFβ1 and reduced NKG2D expression on the NK cell surface is accompanying with cancer patients (21). It has been shown that TGFβ1 can suppress NKG2D expression on NK cells (22). Whether TGFβ1 has similar suppressing effects on MICA expression in NPC remains unclear. Promoter analysis indicated that MICA contains the binding site for the known TGFβ1-regulated transcriptional factor c-Myc (23). We first examined the effects of TGFβ1 on MICA expression in NPC. Our results demonstrated that TGFβ1 is a potent activator of MICA in NPC. Thus, suppressing TGFβ1 expression in NPC might provide protection against and evasion from NK cells. It has been reported that c-Myc could affect the response of tumor cells to the killing effect of NK cells. In tumor cell, c-Myc enhanced the expression of B7-H6 which was the surface expressed ligand for NKp30, an activating receptor of NK cells. Suppression of c-Myc impaired the cytotoxic activity of NK cells activated by NKp30 (24). Likewise, we found that c-Myc could enhanced the expression of MICA, a ligand for another activating receptor NKG2D of NK cells.

The expression of MICA was much more affected by BART7 mimic in comparison with c-Myc and TGFβ1. This observation might be due to the presence of other mechanisms responsible for the regulatory role of ebv-miR-BART7 on MICA. Therefore, ebv-miR-BART7 modulated MICA expression partially through TGFβ1/c-Myc.

EBV infection is commonly found in the undifferentiated NPC. The viral gene products could be detected in the tumor cells with potent oncogenic functions in NPC development (25). At present, how EBV confers protection to NPC to escape from the attack of NK cells remains to be clarified. However, it has been shown that EBV can express different viral proteins to suppress the immune attack from other immune cells such as cytotoxic T cells. EBV-infected cells in the lytic phase could express BNLF2a to reduce transporter associated with antigen processing (TAP) function and expression of human histocompatibility leukocyte antigen (HLA) class I (26). EBV-expressed BGLF5 (EBV alkaline exonuclease) can impair T-cell recognition by interfering HLA class II immune responses (27).

Recent study has shown that EBV-encoded microRNA can inhibit recognition and immune attack of EBV-specific CD8+ T cells by targeting peptide transporter subunit TAP2 (28). EBV-encoded microRNA could also modulate CD4+ T cells response by targeting cytokine and MHC class II family expression (28). Comparatively, how does the EBV-infected cells evade from the immune attack of NK cells is less well understood. We have previously shown that ebv-miR-BART7 can suppress TGFβ1 in the NPC cells (12). Based on the new observation that TGFβ1 can un-regulate MICA expression in NPC, we speculate that ebv-miR-BART7 could play a part in the NPC immunity by weakening the interaction between NPC and NK cells. The NPC cell lines employed in the current study does not harbor EBV. Transfection of synthetic ebv-miR-BART7 results in a significant decrease in c-Myc and MICA indicating that EBV-associated NPC might escape from the surveillance of NK cells. This functional role is confirmed in the NPC/NK cells co-cultures and the results demonstrate the significance of ebv-miR-BART7 in NPC.

In conclusion, our results show that ebv-miR-BART7 can reduce the susceptibility of NPC cells to NK cell lysis by reducing expression of MICA. As the activating role of MICA is well-studied and confirmed, we speculate that targeting the ebv-miR-BART7 expression in the NPC tissues can increase MICA and thereby increase the susceptibility of NPC to NK cells. Further studies are warranted to confirm the potential role of ebv-miR-BART7 in NPC immunotherapy.

Acknowledgements

Not applicable.

Funding

The present study is supported by the Health and Medical Research Fund from Food and Health Bureau, Hong Kong SAR (grant no. 01121626), Seed Funding of Basic Research from The University of Hong Kong, and S. K. Yee Medical Foundation Grant.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

TSW, WG and JYWC set up the hypothesis and designed the experiments. SC and MJZ performed the experiments and acquired the data. TSW, WG and JYWC analyzed and interpreted the data. TSW and WG wrote the manuscript. All authors read and approve the final manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Tsao SW, Yip YL, Tsang CM, Pang PS, Lau VM, Zhang G and Lo KW: Etiological factors of nasopharyngeal carcinoma. Oral Oncol. 50:330–338. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Young LS and Rickinson AB: Epstein-barr virus: 40 years on. Nat Rev Cancer. 4:757–768. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Ressing ME, van Gent M, Gram AM, Hooykaas MJ, Piersma SJ and Wiertz EJ: Immune evasion by epstein-barr virus. Curr Top Microbiol Immunol. 391:355–381. 2015.PubMed/NCBI

4 

Trinchieri G: Biology of natural killer cells. Adv Immunol. 47:187–376. 1989. View Article : Google Scholar : PubMed/NCBI

5 

Guillerey C, Huntington ND and Smyth MJ: Targeting natural killer cells in cancer immunotherapy. Nat Immunol. 17:1025–1036. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Jinushi M, Takehara T, Tatsumi T, Hiramatsu N, Sakamori R, Yamaguchi S and Hayashi N: Impairment of natural killer cell and dendritic cell functions by the soluble form of MHC class I-related chain A in advanced human hepatocellular carcinomas. J Hepatol. 43:1013–1020. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Lynn TC, Wang JH, Yang CS and Tu SM: Natural killer cell activity in patients with nasopharyngeal carcinoma. Zhonghua Min Guo Wei Sheng Wu Ji Mian Yi Xue Za Zhi. 19:177–182. 1986.(In Chinese). PubMed/NCBI

8 

Tian W, Luo QZ, Li LX, Jin HK, Wang F, Guo SS and Cao Y: Polymorphism of short tandem repeat of exon 5 of MHC class-I chain related gene A and association with nasopharyngeal carcinoma in a southern Chinese population. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 22:309–312. 2005.(In Chinese). PubMed/NCBI

9 

Tian W, Zeng XM, Li LX, Jin HK, Luo QZ, Wang F, Guo SS and Cao Y: Gender-specific associations between MICA-STR and nasopharyngeal carcinoma in a southern Chinese Han population. Immunogenetics. 58:113–121. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Kishikawa T, Otsuka M, Yoshikawa T, Ohno M, Takata A, Shibata C, Kondo Y, Akanuma M, Yoshida H and Koike K: Regulation of the expression of the liver cancer susceptibility gene MICA by microRNAs. Sci Rep. 3:27392013. View Article : Google Scholar : PubMed/NCBI

11 

Song H, Kim Y, Park G, Kim YS, Kim S, Lee HK, Chung WY, Park SJ, Han SY, Cho D and Hur D: Transforming growth factor-β1 regulates human renal proximal tubular epithelial cell susceptibility to natural killer cells via modulation of the NKG2 ligands. Int J Mol Med. 36:1180–1188. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Gao W, Li ZH, Chen S, Chan JY, Yin M, Zhang MJ and Wong TS: Epstein-Barr virus encoded microRNA BART7 regulates radiation sensitivity of nasopharyngeal carcinoma. Oncotarget. 8:20297–20308. 2017.PubMed/NCBI

13 

Moodley K, Angel CE, Glass M and Graham ES: Real-time profiling of NK cell killing of human astrocytes using xCELLigence technology. J Neurosci Methods. 200:173–180. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Seliger B, Harders C, Lohmann S, Momburg F, Urlinger S, Tampé R and Huber C: Down-regulation of the MHC class I antigen-processing machinery after oncogenic transformation of murine fibroblasts. Eur J Immunol. 28:122–133. 1998. View Article : Google Scholar : PubMed/NCBI

15 

Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL and Spies T: Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science. 285:727–729. 1999. View Article : Google Scholar : PubMed/NCBI

16 

Groh V, Rhinehart R, Randolph-Habecker J, Topp MS, Riddell SR and Spies T: Costimulation of CD8alphabeta T cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat immunol. 2:255–260. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Li MO, Wan YY, Sanjabi S, Robertson AK and Flavell RA: Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol. 24:99–146. 2006. View Article : Google Scholar : PubMed/NCBI

18 

White RA, Malkoski SP and Wang XJ: TGFβ signaling in head and neck squamous cell carcinoma. Oncogene. 29:5437–5446. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Viel S, Marçais A, Guimaraes FS, Loftus R, Rabilloud J, Grau M, Degouve S, Djebali S, Sanlaville A, Charrier E, et al: TGF-β inhibits the activation and functions of NK cells by repressing the mTOR pathway. Sci Signal. 9:ra192016. View Article : Google Scholar : PubMed/NCBI

20 

Rouce RH, Shaim H, Sekine T, Weber G, Ballard B, Ku S, Barese C, Murali V, Wu MF, Liu H, et al: The TGF-β/SMAD pathway is an important mechanism for NK cell immune evasion in childhood B-acute lymphoblastic leukemia. Leukaemia. 30:800–811. 2016. View Article : Google Scholar

21 

Crane CA, Han SJ, Barry JJ, Ahn BJ, Lanier LL and Parsa AT: TGF-beta downregulates the activating receptor NKG2D on NK cells and CD8+ T cells in glioma patients. Neuro Oncol. 12:7–13. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Lee JC, Lee KM, Kim DW and Heo DS: Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. J Immunol. 172:7335–7340. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Zhu X, Ozturk F, Liu C, Oakley GG and Nawshad A: Transforming growth factor-β activates c-Myc to promote palatal growth. J Cell Biochem. 113:3069–3085. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Textor S, Bossler F, Henrich KO, Gartlgruber M, Pollmann J, Fiegler N, Arnold A, Westermann F, Waldburger N, Breuhahn K, et al: The proto-oncogene Myc drives expression of the NK cell-activating NKp30 ligand B7-H6 in tumor cells. Oncoimmunology. 5:e11166742016. View Article : Google Scholar : PubMed/NCBI

25 

Young LS and Dawson CW: Epstein-Barr virus and nasopharyngeal carcinoma. Chin J Cancer. 33:581–590. 2014.PubMed/NCBI

26 

Hislop AD, Ressing ME, van Leeuwen D, Pudney VA, Horst D, Koppers-Lalic D, Croft NP, Neefjes JJ, Rickinson AB and Wiertz EJ: A CD8+ T cell immune evasion protein specific to Epstein-Barr virus and its close relatives in Old World primates. J Exp Med. 204:1863–1873. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Zuo J, Thomas W, van Leeuwen D, Middeldorp JM, Wiertz EJ, Ressing ME and Rowe M: The DNase of gammaherpesviruses impairs recognition by virus-specific CD8+ T cells through an additional host shutoff function. J Virol. 82:2385–2393. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Albanese M, Tagawa T, Bouvet M, Maliqi L, Lutter D, Hoser J, Hastreiter M, Hayes M, Sugden B, Martin L, et al: Epstein-Barr virus microRNAs reduce immune surveillance by virus-specific CD8+ T cells. Proc Natl Acad Sci USA. 113:E6467–E6475. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 16 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wong TS, Chen S, Zhang MJ, Chan JY and Gao W: Epstein‑Barr virus‑encoded microRNA BART7 downregulates major histocompatibility complex class I chain‑related peptide A and reduces the cytotoxicity of natural killer cells to nasopharyngeal carcinoma. Oncol Lett 16: 2887-2892, 2018
APA
Wong, T., Chen, S., Zhang, M., Chan, J.Y., & Gao, W. (2018). Epstein‑Barr virus‑encoded microRNA BART7 downregulates major histocompatibility complex class I chain‑related peptide A and reduces the cytotoxicity of natural killer cells to nasopharyngeal carcinoma. Oncology Letters, 16, 2887-2892. https://doi.org/10.3892/ol.2018.9041
MLA
Wong, T., Chen, S., Zhang, M., Chan, J. Y., Gao, W."Epstein‑Barr virus‑encoded microRNA BART7 downregulates major histocompatibility complex class I chain‑related peptide A and reduces the cytotoxicity of natural killer cells to nasopharyngeal carcinoma". Oncology Letters 16.3 (2018): 2887-2892.
Chicago
Wong, T., Chen, S., Zhang, M., Chan, J. Y., Gao, W."Epstein‑Barr virus‑encoded microRNA BART7 downregulates major histocompatibility complex class I chain‑related peptide A and reduces the cytotoxicity of natural killer cells to nasopharyngeal carcinoma". Oncology Letters 16, no. 3 (2018): 2887-2892. https://doi.org/10.3892/ol.2018.9041