Epstein-Barr virus latent membrane protein 1 induces CD69 expression through activation of nuclear factor-κB

  • Authors:
    • Chie Ishikawa
    • Naoki Mori
  • View Affiliations

  • Published online on: March 28, 2013     https://doi.org/10.3892/ijo.2013.1871
  • Pages: 1786-1792
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Latent membrane protein‑1 (LMP-1) of Epstein‑Barr virus (EBV) promotes tumorigenesis. Here, we report that LMP-1 activates the immunoregulatory molecule CD69 gene transcription through a nuclear factor-κB (NF-κB)‑dependent pathway. CD69 expression was upregulated in LMP-1‑expressing EBV-immortalized human B-cell lines and an EBV-positive Burkitt's lymphoma cell line. LMP-1 expression increased CD69 expression at the transcriptional level. CD69 promoter was regulated by LMP-1 activation of NF-κB via the carboxy-terminal activation region 1 and 2. Promoter deletion analysis indicated that two NF-κB binding sites are necessary for activation of the CD69 promoter. Electrophoretic mobility shift analysis demonstrated that LMP-1 activates both NF-κB binding sites in the CD69 promoter. This is the first report of the regulation of CD69 expression by LMP-1, and this novel finding may, thus, represent an important link between the EBV oncoprotein LMP-1 and its critical role in the development of EBV-associated diseases.

Introduction

Epstein-Barr virus (EBV) is a ubiquitous human γ-herpesvirus and was one of the first human virus linked to cancer (1). EBV can infect, transform and immortalize B-lymphocytes in vitro, giving rise to lymphoblastoid cell lines (LCLs), which display elevated levels of several cellular activation antigens (2). Consistent with this feature, persistent, latent EBV infection is present in several lymphoid malignancies including Burkitt’s lymphoma (BL) (3).

EBV latent membrane protein-1 (LMP-1) is a key effector in EBV-mediated transformation of B cells (4). It consists of 386 amino acids and its carboxy-terminal cytoplasmic domain contains two carboxy-terminal activation regions (CTARs), CTAR-1 and CTAR-2. CTAR-1 and CTAR-2 are known to activate the nuclear factor-κB (NF-κB) signaling pathway (58). NF-κB is inactive in the cytosol because it is bound to IκBα and IκBβ, and becomes active after phosphorylation and subsequent degradation of IκBα and IκBβ (9). The released NF-κB from the complex translocates to the nucleus where it activates a variety of genes (9). The high-molecular weight complex, IκB kinase (IKK), which is composed of two catalytic subunits, IKKα and IKKβ, and a regulatory subunit, IKKγ, phosphorylates IκBs (9). Members of the p38 mitogen-activated protein kinase (MAPK) kinase kinase protein kinase family mediate the physiological activation of IKK (10). These kinases include NF-κB-inducing kinase (NIK) (11) and MAPK/extracellular signal-regulated kinase kinase 1 (12). CTAR-1 binds to tumor necrosis factor receptor-associated factors (TRAFs) (5), whereas CTAR-2 binds to the tumor necrosis factor receptor-associated death domain (TRADD) (6). NF-κB activation by the CTAR-1 and CTAR-2 domains of LMP-1 is likely to be mediated by the binding of TRAFs directly or indirectly to both the CTAR-1 and CTAR-2 domains (58). NF-κB activation by LMP-1 is mediated by NIK or a related MAPK kinase kinase, and subsequent activation of the IKK complex (7,13). NIK is activated by aggregated TRAF2 (13). Thus, binding of LMP-1 to TRAFs and TRADD initiates the formation of a signaling complex that leads to activation of NF-κB and MAPK (58), resulting in upregulation of several genes expression.

The human activation inducer molecule CD69 is a disulfide-linked transmembrane homodimeric glycoprotein (14). CD69 is not detected in peripheral blood lymphocytes, but it is expressed by small subsets of T and B cells in peripheral lymphoid tissues (15,16). CD69 acts as a signal transducer in inflammatory processes. Recent studies have established the role of CD69 as an intrinsic negative modulator of T cell responses (17). In the present report, we show that CD69 is a transcriptional target of LMP-1 activation of NF-κB via CTAR-1 and CTAR-2.

Materials and methods

Cell lines

Raji is a human EBV-positive BL cell line, and LCL-Ao, LCL-Ka and LCL-Ku are EBV-immortalized human B-cell lines generated from peripheral blood mononuclear cells of healthy volunteers. These lymphoid cell lines were cultured in Roswell Park Memorial Institute (RPMI)-1640 medium supplemented with 10 or 20% heat-inactivated fetal bovine serum, 50 U/ml penicillin and 50 μg/ml streptomycin. Human embryonic kidney 293T cells were maintained in Dulbecco’s modified Eagle’s medium supplemented with 10% heat-inactivated fetal bovine serum, 50 U/ml penicillin and 50 μg/ml streptomycin.

Flow cytometry

The expression of CD69 was analyzed by flow cytometry with phycoerythrin (PE)-labeled mouse monoclonal antibody against CD69 (clone TP1.55.3; Beckman Coulter, Fullerton, CA). Analyses were carried out with isotype-matched control antibody. The cells were incubated with the antibody for 30 min, washed with cell WASH (Becton-Dickinson Immunocytometry Systems, San Jose, CA) and then subjected to analysis in the cytometer.

RNA detection

Total RNA was prepared from various cell cultures by TRIzol (Invitrogen, Carlsbad, CA) according to the protocol provided by the manufacturer. First-strand cDNA was synthesized from 1 μg total cellular RNA using a PrimeScript RT-PCR kit (Takara Bio Inc., Otsu, Japan) with random primers. The primers used were 5′-CATAGCTCTCATTGCCTTATCAGT-3′ (forward) and 5′-CCTCTCTACCTGCGTATCGTTT-3′ (reverse) for CD69, 5′-GTGACTGGACTGGAGGAGCC-3′ (forward) and 5′-GAGGGAGTCATCGTGGTGGTG-3′ (reverse) for LMP-1, and 5′-GTGGGGCGCCCCAGGCACCA-3′ (forward) and 5′-CTCCTTAATGTCACGCACGATTTC-3′ (reverse) for β-actin. The length of the semiquantitative reverse transcription-PCR (RT-PCR) for each gene was: 30 cycles for CD69 and LMP-1, and 28 cycles for β-actin. The PCR products were fractionated on 2% agarose gels and visualized by ethidium bromide staining.

Plasmids and transfections

Luciferase assay was performed to confirm that LMP-1 induces the CD69 promoter activation. Approximately 3×105 293T cells per plate were transfected by the calcium phosphate DNA co-precipitation method. All transfections included appropriate reporter and effector plasmids. The expression plasmids pSG5-LMP-1, pSG5-LMP-1Δ187-351, pSG5-LMP-1Δ 349 and pSG5-LMP-1Δ194-386 were kindly provided by Dr Martin Rowe (University of Wales College of Medicine, Cardiff, UK) (18,19). The CD69 promoter-luciferase gene constructs have already been described (20,21). The single and combined internal deletion mutants of NF-κB sites were constructed by deletion of the NF-κB sites of the plasmid pAIM 255-LUC. The IκBαΔN- and IκBβΔN-dominant negative mutants are IκBα and IκBβ deletion mutants lacking the amino-terminal 36 and 23 amino acids, respectively (22,23). The dominant negative mutants of IKKα, IKKα (K44M), IKKβ, IKKβ (K44A), IKKγ, IKKγ (1–305) and NIK, NIK (KK429/430AA) have been described previously (24,25). Plasmids for truncated TRAF2 and TRAF5 proteins retaining only the TRAF domain, ΔTRAF2 and ΔTRAF5, have been described previously (26,27). In all cases, the reference plasmid phRL-TK, which contains the Renilla luciferase gene under the control of the herpes simplex virus thymidine kinase promoter, was cotransfected to correct for transfection efficiency. After 24 h, the transfected cells were collected by centrifugation, washed with phosphate-buffered saline and lysed in reporter lysis buffer (Promega, Madison, WI). Luciferase assays were conducted using the dual luciferase reporter system (Promega), in which the relative luciferase activity was calculated by normalizing transfection efficiency according to the Renilla luciferase activities.

Preparation of nuclear extracts and electrophoretic mobility shift assay (EMSA)

Nuclear proteins were extracted as described by Antalis and Godbolt (28) with some modifications, and NF-κB binding activity to the NF-κB element was examined by EMSA. Briefly, 5 μg of nuclear extracts were preincubated in a binding buffer containing 1 μg polydeoxyinosinic-deoxycytidylic acid (GE Healthcare Biosciences, Buckinghamshire, UK), followed by the addition of 32P-labeled oligonucleotide probes containing the NF-κB element. The mixtures were incubated for 15 min at room temperature. The DNA protein complexes were separated on 4% polyacrylamide gels and visualized by autoradiography. The probes and competitors used were prepared by annealing the sense and antisense synthetic oligonucleotides as follows: the NF-κB element (κB1) of the CD69 gene (5′-GATCCAGACAACAGGGAAAACCCATACTTC-3′); the NF-κB element (κB2) of the CD69 gene (5′-GATCCAGAGTCTGGGAAAATCCCACTTTCC-3′); a typical NF-κB element from the IL-2 receptor α chain (IL-2Rα) gene (5′-GATCCGGCAGGGGAATCTCCCTCTC-3′) and an AP-1 element of the IL-8 gene (5′-GATCGTGATGACTCAGGTT-3′). The above underlined sequences represent the NF-κB and AP-1 binding sites, respectively.

Results

Upregulated CD69 expression in EBV-immortalized human B-cell lines and EBV-positive BL cell line

We examined first whether CD69 upregulation is a general feature of EBV-infected B cells. All EBV-immortalized B-cell lines, LCL-Ao, LCL-Ka and LCL-Ku, and EBV-positive BL cell line, Raji, constitutively expressed CD69 on the cell surface (Fig. 1A). In contrast, CD69 was hardly expressed on normal peripheral blood mononuclear cells (data not shown).

LMP-1 upregulates CD69 mRNA and protein expression

EBV-immortalized B-cell lines and Raji cells constitutively express LMP-1 (29,30). Therefore, we analyzed the induction of CD69 expression by LMP-1. Transient expression assays using a mammalian expression vector for LMP-1 were performed in 293T cells. In 293T cells transfected with empty vector (pSG5), CD69 mRNA was not detectable as determined by RT-PCR analysis (Fig. 1B). In contrast, ectopic expression of LMP-1 induced the expression of CD69 mRNA in 293T cells. Next, we investigated whether LMP-1 also transcribed the endogenous CD69 gene. The expression of CD69 antigen on the cell surface of 293T cells increased after ectopic expression of LMP-1 in 293T cells, and such increase was time-dependent (Fig. 1C).

LMP-1 activates CD69 promoter

To determine whether LMP-1 regulates CD69 promoter activity, transient expression assays using the reporter plasmid pAIM 1.4-LUC and either an expression vector for LMP-1 or empty vector (pSG5) were performed in 293T cells. LMP-1 induced relative levels of CD69 promoter-directed luciferase expression in a dose-dependent manner, suggesting that LMP-1 functionally activates CD69 promoter (Fig. 2A). To map the region in the LMP-1 protein that mediates activation of CD69 promoter, LMP-1 mutants were expressed in 293T cells and their effect on CD69 promoter activity was investigated. The LMP-1 mutants used included LMP-1Δ187-351 (which contains only CTAR-2 in the carboxy-terminus), LMP-1Δ349 (which lacks CTAR-2), and LMP-1Δ194-386 (in which the entire carboxy-terminal cytoplasmic region is deleted) (Fig. 2B, left panel). In cells that expressed LMP-1Δ194-386, CD69 promoter activity was not significantly increased (Fig. 2B, right panel). In contrast, LMP-1Δ187-351 induced 50% of wild-type LMP-1 CD69 promoter activation. Furthermore, LMP-1Δ349 showed substantial impairment of CD69 promoter activation. These results suggest that LMP-1 activates CD69 expression via the cooperative activity of CTAR-1 and CTAR-2 signaling motifs.

LMP-1 activates the CD69 promoter via the NF-κB signaling pathway

CTAR-1 and CTAR-2 are known to activate the NF-κB signaling pathway (58). Based on the discussed background, we tested the ability of the dominant interfering mutants of IκBα, IκBβ, IKKγ, TRF2 and TRAF5, and kinase-deficient mutants of IKKα, IKKβ and NIK to inhibit LMP-1-mediated transactivation of CD69-driven reporter gene activity. Expression of each of these inhibitory mutants inhibited LMP-1-induced activation of CD69 promoter (Fig. 2C). These results demonstrated that the signaling components, TRAFs, NIK and IKKs, which are involved in the activation of NF-κB, are also necessary for LMP-1 transactivation of the CD69 promoter.

LMP-1 activates the CD69 promoter through two NF-κB sites

The CD69 promoter contains three putative binding sites for NF-κB [positions −373 (κB3), −223 (κB2) and −160 (κB1)], a putative binding site for early growth response (EGR) at −69, a composite binding site for Sp1 and EGR at −56, a cyclic adenosine 3′,5′-monophosphate response element-binding protein (CREB) binding site at −46, and two putative AP-1 binding sites at −16 and +1 (Fig. 3A) (20). To study the role of NF-κB sites in the transcriptional induction of CD69 gene by LMP-1, we transfected 293T cells with several 5′-deletion fragments extending from position −1.4 k to −78 (Fig. 3A), and an expression vector for LMP-1. Progressive removal of 5′-sequence up to position −255 did not significantly inhibit the induced promoter activity, suggesting that the 271-bp fragment, spanning positions −255 to +16, contained the LMP-1-responsive elements (Fig. 3A). Further deletion of upstream sequences up to position −170 resulted in significant loss of responsiveness to LMP-1. Additional removal of the 5′-sequences up to position −78 further affected the inducible promoter activity.

Two potential NF-κB-binding sequences were identified at positions −160 (κB1) and −223 (κB2) spanning positions −255 to +16 bp. κB1 and κB2 were identical to those found in the gene promoters of c-myc and IL-6, respectively (31). To test the relative contribution of the NF-κB binding sites to the LMP-1-mediated activation of CD69, plasmids with internal deletion mutants of these sites in the CD69 promoter were transfected (Fig. 3B). Single deletion of the κB1 or κB2 site resulted in reduction of the inducible activity. Double deletions of the κB1 and κB2 sites further reduced Tax-mediated activation of this reporter construct. These results indicate that the two NF-κB binding sites in the CD69 promoter regulate LMP-1-induced upregulation of CD69.

LMP-1 induces NF-κB binding to DNA

Since both NF-κB binding sites of the CD69 promoter are required for LMP-1-mediated CD69 gene transcription, we studied the ability of LMP-1 to activate NF-κB binding to DNA. For this purpose, 293T cells were transfected with control plasmid (empty vector) or LMP-1 expression plasmid for 24 or 48 h, followed by extraction of nuclear proteins. EMSA demonstrated that LMP-1 increased the binding of NF-κB to 32P-labeled oligonucleotides representing the κB1 and κB2 sites (Fig. 4A). The specificity of DNA-protein complex formation was determined by competition studies with unlabeled competitors. As expected, excess of cold CD69 κB1 or κB2 double-stranded oligonucleotide, or consensus NF-κB site from the IL-2Rα promoter, effectively competed with the labeled probes and eliminated the binding of nuclear extracts from 293T cells transfected with LMP-1 expression plasmid (Fig. 4B, lanes 2–5). In contrast, the unlabeled IL-8 AP-1 element did not compete with labeled probes (Fig. 4B, lanes 2 and 6). The nucleoproteins binding to both NF-κB sites were composed of p50, p65 and c-Rel subunits of the NF-κB family, as demonstrated by supershift experiments with specific antibodies (Fig. 4B, lanes 7–9).

Finally, to determine the role of LMP-1 on endogenous NF-κB binding to DNA, we measured NF-κB binding to respective NF-κB elements in the CD69 promoter in LMP-1- and CD69-expressing Raji cells. As expected, protein complexes bound to both κB1 and κB2 sites were detected in nuclear extracts from Raji cells (Fig. 4C, lane 1). The specificity of DNA-protein complexes in these extracts was determined by competition studies using unlabeled competitors. As observed in nuclear extracts from 293T cells transfected with LMP-1 expression plasmid, cold κB1 and κB2 oligonucleotides, and consensus NF-κB site from the IL-2Rα promoter, but not the IL-8 AP-1 element, efficiently competed with labeled probes (Fig. 4C, lanes 1–5). Antibodies against p50, p65, c-Rel and RelB induced a supershift of the DNA-protein complexes (Fig. 4C, lanes 6–8 and 10). Taken together, the results indicate that NF-κB proteins bind to both κB elements of the CD69 promoter in LMP-1- and CD69-expressing Raji cells.

Discussion

The main issue addressed in this study was whether LMP-1 induces CD69 gene transcription and the possible mechanisms underlying this activity. In this study, we showed that EBV LMP-1 activates CD69 gene transcription through an NF-κB-dependent pathway. The activation of CD69 promoter was mediated cooperatively by CTAR-1 and CTAR-2 of LMP-1, as demonstrated by ectopic expression of LMP-1 mutants. Interestingly, previous studies have demonstrated that CTAR-1 and CTAR-2 cooperatively activate the NF-κB signaling pathway (58), and that CD69 promoter harbors three κB consensus sites (κB1-κB3) (20). The results also showed that both κB1 and κB2 domains of the CD69 promoter were absolutely necessary for LMP-1-induced transcription.

The present results indicate that LMP-1 induces CD69 promoter activation through NIK-, IKKα- and IKKβ kinase-dependent pathways. Dominant negative forms of NIK, IKKα, IKKβ- and IKKγ inhibited LMP-1-induced CD69 promoter activation. These results are consistent with previous evidence that NIK and IKKs are implicated in the pathways through which LMP-1 induces NF-κB activation (7,13). LMP-1 CTAR-1 directly recruits TRAF1, 2, 3 and 5 whereas CTAR-2 indirectly recruits TRAF2 and 6 (58). CTAR-1 activates TRAF2-, NIK-and IKKα-mediated NF-κB pathways, and CTAR-2 activates TRAF6-, IKKα-, IKKβ- and IKKγ-mediated NF-κB pathways (7). Overexpression of dominant negative forms of TRAF2 and TRAF5 inhibited LMP-1-mediated CD69 promoter activation, indicating that NF-κB is an important component of LMP-1-mediated CD69 gene induction from TRAFs-interacting sites.

CD69 has been found to be rapidly upregulated on all the leukocyte lineages studied, upon activation with the corresponding stimuli (14). CD69 expression has also been reported in infections (3235). Earlier studies showed that CD69 regulates the immune response by modulating the expression of various cytokines. CD69-deficient mice show increased anti-tumor and autoimmune responses, which are caused at least in part by increased production of proinflammatory cytokines and chemokines (36,37). More recently, tumor-derived CD69+ T cells have been found to induce immune tolerance in the tumor environment (38). CD69 is reported to be a critical negative regulator of immune activation during intracellular bacterial infection (35). LMP-1-mediated CD69 may have an important role in the immune surveillance evasion by EBV-infected cells.

In summary, our experiments indicate that LMP-1 CTAR-1 and -2 can regulate CD69 gene at the transcriptional level via two NF-κB-binding sites in its promoter, and TRAFs, NIK and IKKs are effectors of CD69 promoter activation from LMP-1. Further studies are necessary to resolve the question of the role of CD69 in EBV-associated diseases.

Acknowledgements

We thank Dr Martin Rowe, Dr Francisco Sánchez-Madrid, Dr Dean W. Ballard, Dr Romas Geleziunas, Dr Kuan-Teh Jeang, Dr Marta Muzio and Dr Toshiki Watanabe, for providing the expression vectors for LMP-1 and its mutants; CD69 promoter pXP2 luciferase reporter plasmids; expression vectors for IκBα- and IκBβ-dominant negative mutants; for NIK-, IKKα- and IKKβ-dominant negative mutants; for IKKγ-dominant negative mutant; and for plasmids for truncated TRAF2 and TRAF5. We also thank Dr Zahidunnabi Dewan for providing LCL-Ao, LCL-Ka and LCL-Ku.

References

1. 

Epstein MA, Achong BG and Barr YM: Virus particles in cultured lymphoblasts from Burkitt’s lymphoma. Lancet. 1:702–703. 1964.PubMed/NCBI

2. 

Kieff ED and Rickinson AB: Epstein-Barr virus and its replication. Fields’ virology. Knipe DM and Howley PM: Lippincott Williams & Wilkins; Philadelphia: pp. 2603–2654. 2006

3. 

Taylor GS and Blackbourn DJ: Infectious agents in human cancers: lessons in immunity and immunomodulation from gammaherpesviruses EBV and KSHV. Cancer Lett. 305:263–278. 2011. View Article : Google Scholar : PubMed/NCBI

4. 

Kaye KM, Izumi KM and Kieff E: Epstein-Barr virus latent membrane protein 1 is essential for B-lymphocyte growth transformation. Proc Natl Acad Sci USA. 90:9150–9154. 1993. View Article : Google Scholar : PubMed/NCBI

5. 

Devergne O, Cahir McFarland ED, Mosialos G, Izumi KM, Ware CF and Kieff E: Role of the TRAF binding site and NF-κB activation in Epstein-Barr virus latent membrane protein 1-induced cell gene expression. J Virol. 72:7900–7908. 1998.

6. 

Izumi KM and Kieff ED: The Epstein-Barr virus oncogene product latent membrane protein 1 engages the tumor necrosis factor receptor-associated death domain protein to mediate B lymphocyte growth transformation and activate NF-κB. Proc Natl Acad Sci USA. 94:12592–12597. 1997.PubMed/NCBI

7. 

Soni V, Cahir-McFarland E and Kieff E: LMP1 TRAFficking activates growth and survival pathways. Adv Exp Med Biol. 597:173–187. 2007. View Article : Google Scholar : PubMed/NCBI

8. 

Schultheiss U, Püschner S, Kremmer E, Mak TW, Engelmann H, Hammerschmidt W and Kieser A: TRAF6 is a critical mediator of signal transduction by the viral oncogene latent membrane protein 1. EMBO J. 20:5678–5691. 2001. View Article : Google Scholar : PubMed/NCBI

9. 

Hayden MS and Ghosh S: Shared principles in NF-κB signaling. Cell. 132:344–362. 2008.

10. 

Zandi E and Karin M: Bridging the gap: composition, regulation, and physiological function of the IκB kinase complex. Mol Cell Biol. 19:4547–4551. 1999.PubMed/NCBI

11. 

Woronicz JD, Gao X, Cao Z, Rothe M and Goeddel DV: IκB kinase-β: NF-κB activation and complex formation with IκB kinase-α and NIK. Science. 278:866–869. 1997.

12. 

Lee FS, Peters RT, Dang LC and Maniatis T: MEKK1 activates both IκB kinase α and IκB kinase β. Proc Natl Acad Sci USA. 95:9319–9324. 1998.

13. 

Sylla BS, Hung SC, Davidson DM, Hatzivassiliou E, Malinin NL, Wallach D, Gilmore TD, Kieff E and Mosialos G: Epstein-Barr virus-transforming protein latent infection membrane protein 1 activates transcription factor NF-κB through a pathway that includes the NF-κB-inducing kinase and the IκB kinases IKKα and IKKβ. Proc Natl Acad Sci USA. 95:10106–10111. 1998.PubMed/NCBI

14. 

Sánchez-Mateos P and Sánchez-Madrid F: Structure-function relationship and immunochemical mapping of external and intracellular antigenic sites on the lymphocyte activation inducer molecule, AIM/CD69. Eur J Immunol. 21:2317–2325. 1991.

15. 

Sant’Angelo DB, Lucas B, Waterbury PG, Cohen B, Brabb T, Goverman J, Germain RN and Janeway CA Jr: A molecular map of T cell development. Immunity. 9:179–186. 1998.

16. 

Testi R, D’Ambrosio D, De Maria R and Santoni A: The CD69 receptor: a multipurpose cell-surface trigger for hematopoietic cells. Immunol Today. 15:479–483. 1994. View Article : Google Scholar : PubMed/NCBI

17. 

De la Fuente H, Cibrián D and Sánchez-Madrid F: Immunoregulatory molecules are master regulators of inflammation during the immune response. FEBS Lett. 586:2897–2905. 2012.PubMed/NCBI

18. 

Huen DS, Henderson SA, Croom-Carter D and Rowe M: The Epstein-Barr virus latent membrane protein-1 (LMP1) mediates activation of NF-κB and cell surface phenotype via two effector regions in its carboxy-terminal cytoplasmic domain. Oncogene. 10:549–560. 1995.

19. 

Floettmann JE and Rowe M: Epstein-Barr virus latent membrane protein-1 (LMP1) C-terminus activation region 2 (CTAR2) maps to the far C-terminus and requires oligomerisation for NF-κB activation. Oncogene. 15:1851–1858. 1997.PubMed/NCBI

20. 

López-Cabrera M, Muñoz E, Blázquez MV, Ursa MA, Santis AG and Sánchez-Madrid F: Transcriptional regulation of the gene encoding the human C-type lectin leukocyte receptor AIM/CD69 and functional characterization of its tumor necrosis factor-α-responsive elements. J Biol Chem. 270:21545–21551. 1995.PubMed/NCBI

21. 

Castellanos Mdel C, López-Giral S, López-Cabrera M and de Landázuri MO: Multiple cis-acting elements regulate the expression of the early T cell activation antigen CD69. Eur J Immunol. 32:3108–3117. 2002.PubMed/NCBI

22. 

Brockman JA, Scherer DC, McKinsey TA, Hall SM, Qi X, Lee WY and Ballard DW: Coupling of a signal response domain in IκBα to multiple pathways for NF-κB activation. Mol Cell Biol. 15:2809–2818. 1995.

23. 

McKinsey TA, Brockman JA, Scherer DC, Al-Murrani SW, Green PL and Ballard DW: Inactivation of IκBβ by the Tax protein of human T-cell leukemia virus type 1: a potential mechanism for constitutive induction of NF-κB. Mol Cell Biol. 16:2083–2090. 1996.

24. 

Geleziunas R, Ferrell S, Lin X, Mu Y, Cunningham ET Jr, Grant M, Connelly MA, Hambor JE, Marcu KB and Greene WC: Human T-cell leukemia virus type 1 Tax induction of NF-κB involves activation of the IκB kinase α (IKKα) and IKKβ cellular kinases. Mol Cell Biol. 18:5157–5165. 1998.

25. 

Iha H, Kibler KV, Yedavalli VRK, Peloponese JM, Haller K, Miyazato A, Kasai T and Jeang K-T: Segregation of NF-κB activation through NEMO/IKKγ by Tax and TNFα: implications for stimulus-specific interruption of oncogenic signaling. Oncogene. 22:8912–8923. 2003.

26. 

Muzio M, Ni J, Feng P and Dixit VM: IRAK (Pelle) family member IRAK-2 and MyD88 as proximal mediators of IL-1 signaling. Science. 278:1612–1615. 1997. View Article : Google Scholar

27. 

Aizawa S, Nakano H, Ishida T, Horie R, Nagai M, Ito K, Yagita H, Okumura K, Inoue J and Watanabe T: Tumor necrosis factor receptor associated factor (TRAF) 5 and TRAF2 are involved in CD30-mediated NFκB activation. J Biol Chem. 272:2042–2045. 1997.PubMed/NCBI

28. 

Antalis TM and Godbolt D: Isolation of intact nuclei from hematopoietic cell types. Nucl Acids Res. 19:43011991. View Article : Google Scholar : PubMed/NCBI

29. 

Dewan MZ, Tomita M, Katano H and Yamamoto N, Ahmed S, Yamamoto M, Sata T, Mori N and Yamamoto N: An HIV protease inhibitor, ritonavir targets the nuclear factor-kappaB and inhibits the tumor growth and infiltration of EBV-positive lymphoblastoid B cells. Int J Cancer. 124:622–629. 2009. View Article : Google Scholar : PubMed/NCBI

30. 

Xu Z-G, Iwatsuki K, Oyama N, Ohtsuka M, Satoh M, Kikuchi S, Akiba H and Kaneko F: The latency pattern of Epstein-Barr virus infection and viral IL-10 expression in cutaneous natural killer/T-cell lymphomas. Br J Cancer. 84:920–925. 2001. View Article : Google Scholar : PubMed/NCBI

31. 

Baeuerle PA: The inducible transcription activator NF-κB: regulation by distinct protein subunits. Biochim Biophys Acta. 1072:63–80. 1991.

32. 

Hodge G, Hodge S, Han P and Haslam R: Multiple leucocyte activation markers to detect neonatal infection. Clin Exp Immunol. 135:125–129. 2004. View Article : Google Scholar : PubMed/NCBI

33. 

Böhler T, Walcher J, Hölzl-Wenig G, Schnitzler P, Geiss M, Buchholz B, Linde R, Rütschle H and Debatin K-M: Expression of CD69 on T-cells from HIV-1-infected children and adolescents increases with increasing viral load. Eur J Pediatr. 158:638–644. 1999.PubMed/NCBI

34. 

Iwashiro M, Messer RJ, Peterson KE, Stromnes IM, Sugie T and Hasenkrug KJ: Immunosuppression by CD4+ regulatory T cells induced by chronic retroviral infection. Proc Natl Acad Sci USA. 98:9226–9230. 2001.

35. 

Vega-Ramos J, Alari-Pahissa E, Valle JD, Carrasco-Marín E, Esplugues E, Borràs M, Martínez-A C and Lauzurica P: CD69 limits early inflammatory diseases associated with immune response to Listeria monocytogenes infection. Immunol Cell Biol. 88:707–715. 2010. View Article : Google Scholar : PubMed/NCBI

36. 

Esplugues E, Sancho D, Vega-Ramos J, Martínez C, Syrbe U, Hamann A, Engel P, Sánchez-Madrid F and Lauzurica P: Enhanced antitumor immunity in mice deficient in CD69. J Exp Med. 197:1093–1106. 2003. View Article : Google Scholar : PubMed/NCBI

37. 

Sancho D, Gómez M, Viedma F, Esplugues E, Gordón-Alonso M, García-López MA, de la Fuente H, Martínez-A C, Lauzurica P and Sánchez-Madrid F: CD69 downregulates autoimmune reactivity through active transforming growth factor-β production in collagen-induced arthritis. J Clin Invest. 112:872–882. 2003.PubMed/NCBI

38. 

Zhao Q, Kuang D-M, Wu Y, Xiao X, Li X-F, Li T-J and Zheng L: Activated CD69+ T cells foster immune privilege by regulating IDO expression in tumor-associated macrophages. J Immunol. 188:1117–1124. 2012.

Related Articles

Journal Cover

May 2013
Volume 42 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ishikawa C and Ishikawa C: Epstein-Barr virus latent membrane protein 1 induces CD69 expression through activation of nuclear factor-κB. Int J Oncol 42: 1786-1792, 2013
APA
Ishikawa, C., & Ishikawa, C. (2013). Epstein-Barr virus latent membrane protein 1 induces CD69 expression through activation of nuclear factor-κB. International Journal of Oncology, 42, 1786-1792. https://doi.org/10.3892/ijo.2013.1871
MLA
Ishikawa, C., Mori, N."Epstein-Barr virus latent membrane protein 1 induces CD69 expression through activation of nuclear factor-κB". International Journal of Oncology 42.5 (2013): 1786-1792.
Chicago
Ishikawa, C., Mori, N."Epstein-Barr virus latent membrane protein 1 induces CD69 expression through activation of nuclear factor-κB". International Journal of Oncology 42, no. 5 (2013): 1786-1792. https://doi.org/10.3892/ijo.2013.1871