The Epstein–Barr virus (EBV)-encoded latent membrane protein-1 induces NF-κB activity by targeting IκBα. To understand the role of NF-κB activation in EBV-related oncogenesis, we have subcloned mutated IκBα32/36A cDNA into a pHEBo vector containing doxycycline regulatory sequences and stably transfected this construct into a lymphoblastoid cell line. Two tightly regulated clones were obtained in which IκBα32/36A was inducible in a doxycycline dose-dependent manner. Levels of inducible IκBα32/36A peaked at day 2. Inhibition of NF-κB activity was closely correlated with levels of inducible IκBα32/36A. Levels of 3 well-known NF-κB-dependent genes, CD54, p105, and endogenous IκBα, were decreased when IκBα32/36A was induced, and the growth of IκBα32/36A-induced EBV-infected cells was slightly reduced. Loss of NF-κB activity was associated with decreased Bcl-2 protein levels. Finally, the induction of apoptosis was strongly increased in IκBα32/36A-overexpressing cells. Together these results show that it is possible to control IκBα32/36A levels, ie, NF-κB activity, in EBV-infected B-lymphocytes using a doxycycline-inducible vector. Moreover, our results indicate that NF-κB can protect EBV-infected cells from apoptosis by Bcl-2. Finally, our results suggest that a cellular model with doxycycline-inducible IκBα32/36A may be useful in the identification of genuine NF-κB target genes in EBV-infected B cells.

Epstein–Barr virus (EBV)-related B-cell transformation involves cell reprogramming through the modulation of critical cellular transcription factors. Such dysregulation may be the result of direct interactions between viral proteins and cellular transcription factors and from a rerouting of defined cellular signal transduction pathways. For example, EBNA2 protein interacts directly with RBP-Jκ, an inhibitory transcription factor, converting it to a positive transcription regulator. Another well-documented example is the activation of both the AP-1 and the NF-κB transcription factors by LMP-1, which mimics a constitutively active receptor of the tumor necrosis factor (TNF) receptor family (see Mosialos1 and Manet et al2 for review).

In mammalian cells, nuclear NF-κB activity corresponds to heterodimers and homodimers between the 5 Rel/NF-κB proteins, including p50/p105 (NF-κB1), p65 (RelA), p52/p100 (NF-κB2), c-Rel, and RelB. These proteins share a common Rel homology domain, involved in both dimerization and DNA binding to the consensus κB site. NF-κB activation has been implicated in the positive regulation of a variety of genes of the immune response, such as i-NOS, IL-1, IL-2, IL-6, IL-8, tumor necrosis factor-α (TNF-α), major histocompatibility complex (MHC) class 1 and 2, CD25, CD54, and CD80, A20 protein, the proto-oncogene c-myc, and the Rel/NF-κB proteins itself, including NF-κB1, NF-κB2, c-Rel, and the inhibitor IκBα.3 Moreover, even though NF-κB has been associated with the induction of apoptosis in some cellular systems, there is increasing evidence for a role of NF-κB activation in the protection of cells against apoptosis.3,4 

Rel/NF-κB complexes containing RelA or c-Rel are usually trapped within the cytoplasm of most cell types by interaction with inhibitory proteins called IκBα, IκBβ, and IκBε. Activation of NF-κB depends on the phosphorylation and degradation of IκBs, leading to the accessibility of nuclear transport signals and nuclear translocation of the RelA- and c-Rel-containing dimers. Various cell activation pathways such as the TNF-α, IL-1, PKC, and lipopolysaccharide signal transduction pathways converge at the IκB phosphorylation step through the activation of a specific, multisubunit complex called the IκB kinase (IKK) that contains at least 3 polypeptides directly involved in IκB phosphorylation: IKKα, IKKβ,58 and NEMO.9 IKKα and IKKβ are able to phosphorylate IκBα and IκBβ specifically on serine residues 32/36 and 19/23 respectively, whereas the NEMO polypeptide is thought to be involved in signal integration from a variety of signaling pathways that converge at IκB phosphorylation and NF-κB activation.

That constitutive NF-κB activation can contribute directly to cell transformation has been recently highlighted in the Tax model. Indeed, transfection of NEMO into mutant cells unable to activate NF-κB is associated with both the restoration of NF-κB activation and the transformation capacity of Tax.9,10 Various studies suggest a role of Rel/NF-κB in oncogenesis. For example, v-Rel, a virally truncated and mutated version of c-Rel, is responsible for fatal hematologic diseases in chicken and turkey, and transgenic mice expressing v-Rel have rapid T-cell lymphoblastic disorders.1113 In humans, the chromosomal translocation t(10;14)(q24;q32) affects p52 and represents a recurrent chromosomal rearrangement found in certain lymphoid malignancies1416 of B-cell origin. Amplification of the REL locus has been reported to be a feature of primary extranodal lymphoma.17 

LMP-1, one of the major transforming proteins of EBV, contributes to enhanced NF-κB activation in EBV-infected cells.18 It is a transmembrane protein consisting of a short amino terminal cytoplasmic region, 6 transmembrane domains, and 200 cytoplasmic carboxyterminal amino acids. Deletion experiments revealed that the carboxyterminal cytoplasmic tail carries two domains, CTAR1 and CTAR2, collectively responsible for LMP-1 tumorigenesis and the activation of both NF-κB19,20 and JNK kinase.21 CTAR1 is responsible for approximately 30% and CTAR2 for approximately 70% of LMP1-associated NF-κB activation. The 6 transmembrane domains are required for LMP-1 aggregation and the consequent signal transduction that, through the aggregation of TRADD and TRAF molecules,22,23 allows LMP-1 to mimic a constitutively active receptor of the TNF receptor family. The CTAR1 and CTAR2 domains of LMP-1 contain binding sites for TRAF1 and TRADD molecules, respectively.24,25 Both adaptor proteins can themselves bind to TRAF2, which leads to NF-κB activation through the recruitment of the NF-κB-inducing kinase (NIK) and to subsequent IKK activation, which leads in turn to the phosphorylation and degradation of IκBs.26 

The aim of this work was to design a cellular model that would allow us functionally to analyze the role of NF-κB and to identify putative NF-κB target genes in EBV-infected lymphocytes. For this purpose, we chose to inhibit Rel/NF-κB complexes specifically by stable transfection of the cDNA encoding for IκBα mutated on serine 32 and 36 (IκBα32/36A).27 Furthermore, to avoid selection bias for those clones able to escape the potentially deleterious effects of IκBα overexpression, dominant-negative IκBα32/36A was expressed in a conditional fashion from a doxycycline-inducible promoter.28 To prevent vector integration into the genomic DNA, which is difficult to obtain in lymphoblastoid cells and may give rise to position effects, we used the pHEBo backbone containing the EBV OriP sequences. The pHEBO construct allows the vector to replicate episomally in the presence of EBV-encoded nuclear protein-1 (EBNA1).29,30 Our results showed that it was possible to obtain cells in which IκBα32/36A is inducible after doxycycline treatment in a dose-responsive manner. Levels of IκBα32/36A induction were tightly correlated with those of NF-κB inhibition. In addition, we demonstrated that the inhibition of NF-κB was correlated with a decrease in CD54, p105/NF-κB1, and endogenous IκBα expression, implicating these genes as potentially NF-κB-regulated in EBV-infected cells. Finally, we showed that the loss of NF-κB activity in EBV-infected lymphocytes was associated with a significant decrease of Bcl-2 protein expression and with a dramatic increase in their sensitivity to apoptosis.

Plasmid constructs

MS4A was derived by cloning the reverse tetracyclin repressor-transactivator protein (rtTA28) and a tetracycline regulatable promoter31 onto the pHEBo vector.29 To achieve tight regulation, the cytomegalovirus (CMV) promoter was replaced by the EBV–LMP2A promoter in which the endogenous EBNA2 response element was substituted by a heptamerized tet operator sequence. The IκBα32/36A cDNA was excised from pCMV–IκBα32/36A27 by HindIII and XbaI digestion and subcloned into the MS4A vector at the Sfi1 sites (MS4A–IκBα32/36A; see Figure 1). The 0.4SK-luc plasmid containing the IκBα promoter with 3 κB sites upstream of the luciferase gene was a kind gift of A. Israël (Institut Pasteur, Paris, France).32 

Figure 1.

Schematic diagram of the MS4A-IκBα32/36A vector. The different cassettes are indicated in grey boxes: tetO7, tetracycline-responsive elements; IκBα32/36A, c-DNA encoding for IκBα32/36A super-repressor; rtTA, cassette encoding for the doxycycline-responsive factor; EBV–OriP, EBV-OriP sequences; Hygro, cassette encoding for hygromycin resistance.

Figure 1.

Schematic diagram of the MS4A-IκBα32/36A vector. The different cassettes are indicated in grey boxes: tetO7, tetracycline-responsive elements; IκBα32/36A, c-DNA encoding for IκBα32/36A super-repressor; rtTA, cassette encoding for the doxycycline-responsive factor; EBV–OriP, EBV-OriP sequences; Hygro, cassette encoding for hygromycin resistance.

Close modal

Protein extracts and electromobility shift assay

Nuclear and cytoplasmic extracts were performed in parallel as described.33 Gel shift experiments were performed with 20 μg nuclear proteins as described34 with the PRE oligonucleotide containing the 2-κB sites of the human immunodeficiency virus enhancer.35 

Western blot

Our standard gel conditions consisted of 10% SDS–PAGE without stacking gel as described.36 Where indicated, a stacking gel (4% polyacrylamide, 125 mmol/L glycine, pH 6.8) was used according to standard procedures.37 Thirty micrograms protein was fractionated and electrotransferred onto a polyvinylidene difluoride membrane (Millipore, Bedford, MA) 0.8 mA/cm2 with the Hoefer semidry apparatus (Pharmacia Biotech, Orsay, France). Antibodies used for Western blot analysis were MAD 10B for IκBα38 (generous gift of R. Hay, University of St Andrew, St Andrew, UK) diluted at 1/500, monoclonal antibody 124 for Bcl-2 (Dako, Trappes, France) diluted at 1/100, rabbit immune serum no. 1140 diluted at 1/1500 for p105 (generous gift of N. Rice, NCI, Frederick). Detection of the antigen–antibody complexes was performed using a horseradish peroxidase-coupled antimouse or antirabbit antibody (Biorad, Ivry/Seine, France) diluted at 1/10 000, followed by chemiluminescent visualization (Biolabs, Hitchin, UK).

Cell culture and cell transfection

One of our lymphoblastoid cell lines, named PRI, was cultured continuously in RPMI 1640 with standard concentrations of l-glutamine and antibiotics and 10% fetal calf serum. Transfected cells were cultured in the presence of hygromycin (Calbiochem, Meudon, France) at 300 μg/mL and 20% fetal calf serum. Twenty million cells were electroporated with 20 μg MS4A–IκBα32/36A at 250 V and 960 μF. After 4 days, hygromycin was added at 50 μg/mL. Hygromycin concentration was progressively increased up to 300 μg/mL during the first 2 weeks of selection. After 4 weeks of culture in the presence of hygromycin, transfected cells were plated at a limiting dilution on 96-well plates. Screening of clones was based on the inducibility of IκBα expression after doxycycline treatment. To induce IκB overexpression with doxycycline, cells were washed once in RPMI and resuspended in standard medium without hygromycin. Doxycycline (purchased from AP-HP, Paris, France and stored at −20°C at 1 mg/mL in water) was then added at concentrations ranging from 0.01 μg/mL to 8 μg/mL (usually 2 μg/mL). Western blot analysis of IκBα expression was performed 48 hours after doxycycline treatment. Transient transfections were conducted in triplicate with 20 μg 0.4SK-luc plasmid for 10 million cells. Cells were washed once in RPMI, resuspended in standard medium, and electroporated at 250 V and 960 μF. Then the cells were resuspended in standard medium at 106 cells/mL, left 1 hour at 37°C, washed once, and resuspended in standard medium without hygromycin and with or without doxycycline at 2 μg/mL. Luciferase assays were performed after 2 days (Promega, Madison, WI), according the manufacturer’s instructions.

Immunolabelings and flow cytometry analysis

Antibodies tested are listed in Table 1. The expression of cell-surface molecules was assessed by direct and indirect immunofluorescence staining according standard protocols, using a Coulter XL flow cytometer (Coulter, Margency, France).

Table 1.

Antibodies used

SpecificityCloneDilutionSupplierFluorochrome
CD10 J5 1/20 Coulter* FITC 
CD19 B4:89D 1/25 Coulter PE 
CD20 BLy.1 1/20 Dako FITC 
CD21 B120 1/20 Coulter PE 
CD23 MHM6 1/20 Dako FITC 
CD25 1HT44H3 1/20 Coulter FITC 
CD30 HRS4 1/30 Immunotech None 
CD38 T16 1/30 Becton-Dickinson PE 
CD39 AC2 1/20 Immunotech None 
CD44 J173 1/20 Immunotech None 
CD48 J4.57 1/30 Immunotech None 
CD54 84H10 1/10 Immunotech FITC 
CD58 AICD58 1/30 Immunotech None 
CD71 37L 1/20 Coulter FITC 
CD80 MAB1.4 1/20 Immunotech None 
CD95 CH-11 1/5 Immunotech None 
IgM AF6 1/20 Dako FITC 
HLAI B9.12.1 1/50 Immunotech None 
HLAII B8.12.2 1/50 Immunotech None 
F(ab′) Polyclonal 1/20 Dako FITC 
F(ab′) Polyclonal 1/20 Dako FITC 
SpecificityCloneDilutionSupplierFluorochrome
CD10 J5 1/20 Coulter* FITC 
CD19 B4:89D 1/25 Coulter PE 
CD20 BLy.1 1/20 Dako FITC 
CD21 B120 1/20 Coulter PE 
CD23 MHM6 1/20 Dako FITC 
CD25 1HT44H3 1/20 Coulter FITC 
CD30 HRS4 1/30 Immunotech None 
CD38 T16 1/30 Becton-Dickinson PE 
CD39 AC2 1/20 Immunotech None 
CD44 J173 1/20 Immunotech None 
CD48 J4.57 1/30 Immunotech None 
CD54 84H10 1/10 Immunotech FITC 
CD58 AICD58 1/30 Immunotech None 
CD71 37L 1/20 Coulter FITC 
CD80 MAB1.4 1/20 Immunotech None 
CD95 CH-11 1/5 Immunotech None 
IgM AF6 1/20 Dako FITC 
HLAI B9.12.1 1/50 Immunotech None 
HLAII B8.12.2 1/50 Immunotech None 
F(ab′) Polyclonal 1/20 Dako FITC 
F(ab′) Polyclonal 1/20 Dako FITC 

FITC, fluorescein isothiocyanate; PE, phycoerythrin.

*

Coulter–Immunotech, Margency, France.

Dako, Trappes, France.

Becton–Dickinson, Erembodegem–Aalst, Belgium.

Cell-cycle analysis and apoptosis detection

Cell-cycle analysis and detection of apoptosis were performed by flow cytometry as described39 on the basis of BrdU incorporation into DNA and propidium iodide staining. Fluorescence analysis of chromatin condensation and fragmentation was conducted after cytocentrifugation of 50 000 cells and DNA staining with the Hoechst 33258 diluted at 20 μg/mL in phosphate-buffered saline. Staining of cells with fluorescein isothiocyanate-conjugated Annexin V (Pharmingen, San Diego, CA) was performed according to the manufacturer’s protocol.

Characteristics of IκBα32/36A induction by doxycycline in lymphoblastoid cells transfected with the MSA4-IκBα32/36A vector

The MS4A-IκBα32/36A vector was generated by subcloning the IκBα cDNA mutated at positions encoding for the two serine 32 and 36 residues (IκBα32/36A)27 into the MS4A vector, whose transcriptional activity is positively regulated by the addition of doxycycline. The MS4A construct consists of a pHEBo backbone29 (with its EBV–OriP sequences) into which have been subcloned both a promoter carrying the tetO7 tetracycline responsive element and the cDNA encoding for rtTA protein28 (Figure 1).

After transfection of cells with the MS4A-IκBα32/36A vector, cells were grown in the presence of hygromycin and cloned by limiting dilution. Twenty-four clones were screened by Western blotting for IκBα32/36A induction after 48 hours of doxycycline treatment at 2 μg/mL. Two clones were found to be IκBα32/36A inducible. Then the NF-κB activity of IκBα32/36A-inducible clones was assessed by both EMSA and luciferase assay after the transfection of 0.4SK-luc plasmid, which harbors the promoter of IκBα with its 3 κB sites.32 A typical result is illustrated in Figure 2, which shows that a significant induction of IκBα32/36A was correlated with a dramatic decrease of NF-κB activity in DNA binding and transcriptional activity. We next set up a dose–response experiment (Figure 3). Results demonstrated that IκBα32/36A was inducible by doxycycline in a dose-dependent manner and that the decrease of NF-κB binding activity was tightly correlated with the levels of IκBα32/36A induction. In the following experiment, induction of IκBα32/36A was performed at 2 μg/mL doxycycline because doxycycline concentrations greater than 5 μg/mL had been reported to be toxic. SDS–PAGE without a stacking gel36 did not allow the separation of endogenous IκBα from IκBα32/36A (Figures 2, 3). Endogenous IκBα could, however, be distinguished from the transfected gene product on the basis of its higher electrophoretic mobility if a stacking gel was used.27 A comparison of both gel conditions is depicted in Figure 4. Our results showed that there was no detectable leakage of the tet07 promoter (Figure 4). Furthermore, levels of endogenous IκBα decreased when IκBα32/36A expression was induced by doxycycline (Figure 4), suggesting that endogenous IκBα is a genuine target gene of NF-κB in EBV-infected cells.

Figure 2.

Characterization of the IκBα32/36A inducibility after doxycycline treatment. (A) Western blot detection of IκBα protein in MS4A-IκBα32/36A-transfected cells with (+) or without (−) doxycycline at 2 μg/mL. (B) NF-κB binding activity assessed by EMSA in MS4A-IκBα32/36A-transfected cells with (+) or without (−) doxycycline at 2 μg/mL. (C) Schematic representation of the transcriptional regulatory element of the IκBα promoter with its 3 κB sites in front of the luciferase gene (construct 0.4SK-luc). (D) Relative NF-κB transcriptional activity in MS4A-IκBα32/36A-transfected cells with (+) or without (−) doxycycline at 2 μg/mL. Presented results correspond to the mean of 3 transfection experiments with the 0.4SK-luc construct. Each transfection experiment was conducted in triplicate.

Figure 2.

Characterization of the IκBα32/36A inducibility after doxycycline treatment. (A) Western blot detection of IκBα protein in MS4A-IκBα32/36A-transfected cells with (+) or without (−) doxycycline at 2 μg/mL. (B) NF-κB binding activity assessed by EMSA in MS4A-IκBα32/36A-transfected cells with (+) or without (−) doxycycline at 2 μg/mL. (C) Schematic representation of the transcriptional regulatory element of the IκBα promoter with its 3 κB sites in front of the luciferase gene (construct 0.4SK-luc). (D) Relative NF-κB transcriptional activity in MS4A-IκBα32/36A-transfected cells with (+) or without (−) doxycycline at 2 μg/mL. Presented results correspond to the mean of 3 transfection experiments with the 0.4SK-luc construct. Each transfection experiment was conducted in triplicate.

Close modal
Figure 3.

Dose-responsive inducibility of IκBα32/36A by doxycycline. MS4A-IκBα32/36A-transfected cells were treated for 48 hours with various concentrations of doxycycline as indicated. Upper panel: Western blot detection of IκBα. Lower panel: NF-κB-binding activity assessed by EMSA.

Figure 3.

Dose-responsive inducibility of IκBα32/36A by doxycycline. MS4A-IκBα32/36A-transfected cells were treated for 48 hours with various concentrations of doxycycline as indicated. Upper panel: Western blot detection of IκBα. Lower panel: NF-κB-binding activity assessed by EMSA.

Close modal
Figure 4.

Gel electrophoresis with or without stacking gel followed by Western blot analysis of IκBα32/36A. MS4A-IκBα32/36A-transfected cells were (lanes 1 and 3) or were not (lanes 2 and 4) treated with doxycycline for 48 hours. SDS–PAGE and Western blot detection of IκBα was performed without (lanes 1 and 2) or with (lanes 3 and 4) stacking gel.

Figure 4.

Gel electrophoresis with or without stacking gel followed by Western blot analysis of IκBα32/36A. MS4A-IκBα32/36A-transfected cells were (lanes 1 and 3) or were not (lanes 2 and 4) treated with doxycycline for 48 hours. SDS–PAGE and Western blot detection of IκBα was performed without (lanes 1 and 2) or with (lanes 3 and 4) stacking gel.

Close modal

Finally, we established the kinetics of IκBα32/36A induction. As shown in Figure 5, IκBα32/36A induction was highest after 48 hours of doxycycline treatment at 2 μg/mL. Levels of IκBα32/36A slowly decreased at day 4 and day 7. Kinetics of the inhibition of NF-κB binding activity strictly correlated with the kinetics of IκBα32/36A induction. Therefore, phenotypic analysis of doxycycline-treated cells was systematically performed at day 2.

Figure 5.

Kinetics of IκBα32/36A inducibility after doxycycline treatment. Cells were (+) or were not (−) treated with doxycycline at 2 μg/mL. Times of protein extraction are indicated at the top of the figure (D0: protein extraction was performed immediately after doxycycline treatment; D1, D2, D4, and D7: protein extraction was performed immediately after 1, 2, 4, and 7 days of doxycycline treatment, respectively). Upper panel: Western blot detection of IκBα after SDS–PAGE with stacking gel; mutated and endogenous IκBα are indicated by the arrow. Lower panel: NF-κB-binding activity assessed by EMSA.

Figure 5.

Kinetics of IκBα32/36A inducibility after doxycycline treatment. Cells were (+) or were not (−) treated with doxycycline at 2 μg/mL. Times of protein extraction are indicated at the top of the figure (D0: protein extraction was performed immediately after doxycycline treatment; D1, D2, D4, and D7: protein extraction was performed immediately after 1, 2, 4, and 7 days of doxycycline treatment, respectively). Upper panel: Western blot detection of IκBα after SDS–PAGE with stacking gel; mutated and endogenous IκBα are indicated by the arrow. Lower panel: NF-κB-binding activity assessed by EMSA.

Close modal

CD54, p105, and IκBα are putative target genes of NF-κB in EBV infected B cells

Loss of NF-κB activity in the presence of IκBα32/36A was associated with down-regulation of the transcriptional activity of the IκBα promoter (Figure 2D) and resulted in a significant decrease of endogenous IκBα levels (Figures 4, 5). This strongly implicates IκBα to be a genuine target gene of NF-κB in EBV-infected cells. To further evaluate the function of the induced IκBα32/36A protein, we analyzed the expression of some genes that may be positively regulated by NF-κB. Expression of CD54 and p105 was decreased in these cells after 48 hours of doxycycline treatment at 2 μg/mL (Figure 6). These results suggested that the CD54 and p105 genes may also be genuine target genes of NF-κB in EBV-immortalized lymphoblastoid cells.

Figure 6.

Down-regulation of CD54 and p105 expression after induction of IκBα32/36A. Levels of CD54 membrane expression were assessed by flow cytometry on cells treated (+ DoxyC) or not (− DoxyC) with doxycycline at 2 μg/mL for 48 hours (A). Levels of p105 expression were assessed by Western blot on cells treated (+) or not (−) with doxycycline at 2 μg/mL for 48 hours (B).

Figure 6.

Down-regulation of CD54 and p105 expression after induction of IκBα32/36A. Levels of CD54 membrane expression were assessed by flow cytometry on cells treated (+ DoxyC) or not (− DoxyC) with doxycycline at 2 μg/mL for 48 hours (A). Levels of p105 expression were assessed by Western blot on cells treated (+) or not (−) with doxycycline at 2 μg/mL for 48 hours (B).

Close modal

Functional consequences of the IκBα32/36A induction in lymphoblastoid cells

Because NF-κB has been reported to protect cells from apoptosis,4042 and because LMP-1 transfection has been reported to enhance Bcl-2 expression in B cells,43,44 we examined the relationship between the inducible inhibition of NF-κB and any changes in cell proliferation sensitivity to apoptosis. Proliferation of doxycycline-treated cells was decreased slightly in terms of the growth kinetics of cells in culture and the fraction of cells in S-phase, as estimated by flow cytometry after BrdU incorporation (not shown).

Spontaneous apoptosis did not increase significantly in doxycycline-treated cells. However, treatment of cells with daunorubicine, a DNA-intercalating chemotoxic drug that induces apoptosis, clearly showed that IκBα32/36A overexpression (ie, the loss of NF-κB activity) can sensitize EBV-infected B cells to apoptosis, as revealed by chromatin condensation and DNA fragmentation (Figures 7A, 7B), sub-G1 peak induction (Figure 7C), and annexin-V cell-surface fixation (Figure 7D). Finally, we found that the loss of NF-κB binding activity was correlated with a dramatic decrease of Bcl-2 expression within 48 hours in EBV-infected B cells (Figure 7E).

Figure 7.

Induction of apoptosis and expression of Bcl-2 in MS4A-IκBα32/36A-transfected cells. (A) Analysis of nuclear condensation after Hoechst 33258 staining. Cells were (+ DoxyC) or were not (− DoxyC) pretreated with doxycycline at 2 μg/mL for 24 hours. Daunorubicine (Dauno) was added for 18 hours at the concentrations indicated. (B) Dose response to apoptosis induction by daunorubicine in cells pretreated (+ DoxyC) or not (− DoxyC) with doxycycline at 2 μg/mL for 24 hours. Percentages of apoptotic cells were assessed by counting cells with apoptotic nuclei under fluorescence microscope after Hoechst 33258 staining. (C) Flow cytometry analysis of DNA content after propidium iodide staining of the DNA of cells pretreated (+ DoxyC) or not (− DoxyC) with doxycycline at 2 μg/mL for 24 hours and then treated with daunorubicine at 0.2 μmol/L for 18 hours. (D) Annexin V staining of cells pretreated (+ DoxyC) or not (− DoxyC) with doxycycline at 2 μg/mL for 24 hours and then treated with daunorubicine at 0.2 μmol/L for 18 hours. (E) Bcl-2 expression of cells treated (+) or not (−) with doxycycline at 2 μg/mL for 48 hours was assessed by Western blot.

Figure 7.

Induction of apoptosis and expression of Bcl-2 in MS4A-IκBα32/36A-transfected cells. (A) Analysis of nuclear condensation after Hoechst 33258 staining. Cells were (+ DoxyC) or were not (− DoxyC) pretreated with doxycycline at 2 μg/mL for 24 hours. Daunorubicine (Dauno) was added for 18 hours at the concentrations indicated. (B) Dose response to apoptosis induction by daunorubicine in cells pretreated (+ DoxyC) or not (− DoxyC) with doxycycline at 2 μg/mL for 24 hours. Percentages of apoptotic cells were assessed by counting cells with apoptotic nuclei under fluorescence microscope after Hoechst 33258 staining. (C) Flow cytometry analysis of DNA content after propidium iodide staining of the DNA of cells pretreated (+ DoxyC) or not (− DoxyC) with doxycycline at 2 μg/mL for 24 hours and then treated with daunorubicine at 0.2 μmol/L for 18 hours. (D) Annexin V staining of cells pretreated (+ DoxyC) or not (− DoxyC) with doxycycline at 2 μg/mL for 24 hours and then treated with daunorubicine at 0.2 μmol/L for 18 hours. (E) Bcl-2 expression of cells treated (+) or not (−) with doxycycline at 2 μg/mL for 48 hours was assessed by Western blot.

Close modal

The relationship between LMP-1 and NF-κB activation has been extensively studied, and every step between the autoaggregation of LMP-1 and the activation of the IKK has been documented. LMP-1, acting through TRAF molecules, activates several signal transduction pathways including the NF-κB,45 JNK,21 and ras-MAPK46 activation cascades. Therefore, to address the question of the specific role of NF-κB in EBV-infected B lymphocytes, our strategy was to specifically inhibit NF-κB in EBV-infected B cells.

Using the doxycycline-inducible system, we obtained EBV-immortalized B lymphocytes in which it was possible to inhibit NF-κB. To our knowledge, this is the first model that allows one to study the role of NF-κB in EBV-infected B lymphocytes. Establishment of stably transfected cells is often a critical step in the analysis of the function of a particular protein. In current stable transfection protocols, a major bias is introduced by the fact that selection takes place while the transfected gene of interest is expressed. If that particular gene is toxic or induces cell-cycle arrest, establishment of transfected clones is either impossible or will lead to the selection of clones which, through mutation, have escaped the effect of the protein to be studied. To circumvent this problem, various groups have worked on and developed inducible vectors. Fusion of the protein of interest to the hormone binding domain of the estrogen or glucocorticoid receptor has provided a powerful tool with which to study the induction of proliferation, differentiation, and apoptosis.4749 However, in this system, interference of estrogen with NF-κB cannot be excluded. Indeed, a physical association between the activated glucocorticoid receptors and p65 has been reported,50 and estrogen molecules may interact directly with glucocorticoid receptors.51 However, the tetracycline/doxycycline inducible system originally designed by Bujard and collaborators31,52 has proven itself useful for the analysis of the function of proteins whose continuous expression may be deleterious for the cell or may prevent selection in vitro.53 The system described by Gossen and Bujard31 and Gossen et al52 consists of two plasmids: one encodes the tetracycline repressor-VP16 or the reversed tetracycline repressor-VP16 transactivator fusion protein; the other plasmid encodes the gene of interest under the control of a tetracycline-regulatable promoter. The major difficulties of this system are that cells must be stably transfected twice and that the level of expression is highly variable, depending on the site of vector integration into the genomic DNA—a phenomenon known as position effect.

To circumvent these problems, we cloned both the doxycycline-dependent transcription factor rtTA and the tetracycline-regulatable sequences onto the same derived episomal vector (Schuhmacher, manuscript in preparation). pHEBo vectors can be easily transfected into EBV-infected B lymphocytes and maintained episomally. They do not exhibit position effects and, therefore, show little variation in the expression of the transfected gene among different, stably transfected cells.54,55 We have cloned the mutated IκBα32/36A cDNA into this vector and have established stably transfected EBV-immortalized cells that indeed express the mutated IκBα32/36A cDNA in a doxycycline dosage-dependent, inducible fashion. Furthermore IκBα32/36A was functional in terms of the inhibition of NF-κB binding, NF-κB transcriptional activity, and NF-κB-dependent gene expression such as that of endogenous IκBα, p105, and CD54. We obtained a tight correlation between doxycycline concentrations in the medium, levels of IκBα32/36A protein induction, and levels of NF-κB binding inhibition. These results indicated that IκBα32/36A protein levels directly correlated with the level of active rtTA molecules.

Conditional gene expression systems have also proven useful in the analysis of the role of LMP-1 in EBV-driven proliferation. A conditional LMP-1 gene has been introduced into a mini-EBV background56 haboring a tetR–KRAB chimeric repressor cassette and used to immortalize primary B cells.57 Inducible loss of LMP-1 expression was associated with the proliferative arrest of these cells. Reinduction of LMP-1 expression was associated with the reentry of the cells into the cell cycle and the reactivation of the JNK1 and NF-κB signal transduction pathways. These results do not clarify to which extent either JNK1 or NF-κB activation contributes to cellular proliferation. In our cellular model, the inducible loss of NF-κB was associated with only minor effects on the cell cycle in terms of DNA synthesis and cell growth in culture (not shown). These data could appear to suggest that continuous LMP-1-induced NF-κB activation was not required for proliferation and that proliferation would be caused by the activation of a different LMP-1-mediated signal transduction pathway. However, this interpretation is not conclusive because analysis of the kinetics of NF-κB inhibition has revealed a gradual decrease in the level of mutated IκBα32/36A protein over time. This decrease in protein level was associated with a concomitant, gradual increase of NF-κB DNA binding activity to considerable, albeit lower, levels than in the absence of doxycycline. An autoregulatory loop between NF-κB and endogenous IκBα is well established32 but was not expected for the mutated IκBα32/36A in EBV-infected cells. Several possibilities may account for this phenomenon. For example, levels of rtTA–VP16 fusion protein may be down-regulated after IκBα32/36A overexpression. In fact, a search of cryptic κB sites in the MS4A-IκBα32/36A vector revealed the presence of 3 κB consensus sequences (GGGRNNYYCC) within the CMV promoter of the rtTA gene. On the other hand, the accumulation of IκBα32/36A-mutated protein may allow its phosphorylation on tyrosine 4258 or on its caboxyterminal end,59 promoting its proteolytic degradation. This suggests that over a longer period of time, a new equilibrium is reached in the transfected cells between the inhibition of NF-κB by the mutated IκBα32/36A and the activation of NF-κB by LMP-1, resulting in significant and stable levels of NF-κB activation, though at a lower level than in absence of doxycycline. A relationship between NF-κB activation and cyclin D1 expression was reported recently.60 Therefore, our results do not answer whether EBV-immortalized B cells can in fact proliferate in the absence of NF-κB over a longer period of time. Furthermore, our results do not exclude a role for NF-κB in the initial steps of EBV immortalization of primary B cells. Indeed, it is noteworthy that acetylsalicylic acid, which inhibits NF-κB, can block EBV-induced B-cell proliferation in vitro.61 

A series of recent publications points to a relationship between NF-κB activity and the protection of cells from apoptosis.4042,62,63 These results have been obtained in various cell types, including EBV-negative B-cells and EBV-infected lymphoblastoid cells.6365 Here we show that the induction of IκBα32/36A protein is associated with the sensitization of cells to apoptosis in EBV-infected B-cells. Furthermore, we show that the loss of NF-κB is correlated with a significant decrease of the anti-apoptotic protein Bcl-2. Induction of Bcl-2 by EBV in B cells has already been demonstrated.43,44 Transfection of these cells by LMP-1 is associated with an increase in NF-κB activity and the subsequent overexpression of Bcl-2.44 A correlation between LMP-1 and Bcl-2 expression has been noted in AIDS-related primary brain lymphomas.66 Antisense oligonucleotides against LMP-1 suppress Bcl-2 expression in EBV-infected B cells.67 Bcl-2 induction by LMP-1 may be a feature exclusive to B cells. Rowe et al44 have shown that the transfection of LMP-1 into non-B-cell lines is associated with NF-κB activation and CD54 up-regulation without any effect on Bcl-2. Furthermore, even in B cells, Bcl-2 up-regulation by LMP-1 does not seem to be mediated directly by NF-κB because after the transfection of LMP-1, NF-κB activation and CD54 occurred simultaneously whereas Bcl-2 induction was delayed.44 Therefore, to date, a direct causal relationship between NF-κB activation and Bcl-2 up-regulation by LMP-1 has not been determined. Our results clearly show that the loss of NF-κB is associated with a down-regulation of Bcl-2. This suggests that Rel/NF-κB complexes participate in the positive control of Bcl-2 gene expression in EBV-infected B cells, most likely through intermediate target gene expression.

It is noteworthy that protection against apoptosis through NF-κB activation does not seem to be restricted to a particular cell type. A recent report62 has shown that NF-κB’s anti-apoptotic effect is mediated by the up-regulation of TRAF1, TRAF2, c-IAP1, and c-IAP2 molecules in the HT1080 fibrosarcoma cell line, resulting in the inhibition of caspase-8 activation. No up-regulation of Bcl-2 by NF-κB activation was noted in this non-B-cell line. Caspase-8 activation and Bcl-2 down-regulation are initial steps in the cascade of events leading to programmed cell death. The caspase-8 cascade is activated after the aggregation of death-domain proteins at the membrane in response to external stimuli (see Vaux and Korsmeyer68 for review). It would be interesting to verify whether EBV-mediated NF-κB activation leads to the control of TRAF and c-IAP gene expression in our cellular model. Bcl-2 down-regulation seems to be provoked by endogenous cell damage, such as DNA strand breaks after x-ray irradiation. Apoptosis induced by x-ray irradiation is mediated by p53 (see Levine69 for review). P53 exerts both positive transcriptional control on the Bax gene70 and negative control of Bcl-2 gene transcription.71 LMP-1 is responsible for the up-regulation of the A20 protein, a 790-amino acid protein with a unique zinc finger motif.18 A20 gene expression is up-regulated by NF-κB in response to TNF-α and protects cells against the cytotoxic effect of TNF-α.72 LMP-1-mediated protection of cells from apoptosis occurs through A20 overexpression, which in turn inhibits p53.73 Therefore, it can be hypothesized that the loss of NF-κB in EBV-infected cells could be responsible for the down-regulation of A20 gene transcription, thus allowing p53 to inhibit Bcl-2 gene expression and to activate the transcription of Bax. This in turn results in the down-regulation of Bcl-2 gene expression and a sensitization of cells to apoptosis.

One of the next tasks to further our understanding of the role of NF-κB in EBV-related lymphomagenesis will be the identification of NF-κB target genes. MS4A-IκBα32/36A-transfected cells may allow the identification of such genes in EBV-infected B cells. Using bidimensional gel electrophoresis, we are performing a comparative analysis of MS4A–IκBα32/36A-transfected lymphoblas-toid cells either overexpressing or not expressing IκBα32/36A. Initial results show that at least 15 cytosolic polypeptides are down-regulated when NF-κB is inhibited. Thus, MS4A–IκBα32/36A-transfected B-lymphoblastoid cells may be a useful and valuable cellular model to help us in our search for genuine NF-κB target genes in the context of EBV-transformed B lymphocytes.

We thank Hermann Bujard and Manfred Gossen for the gift of plasmids and for sharing their results before publication. We also thank R. Hay (University of St. Andrews, UK) and N. Rice (NCI, Frederick) for the kind gift of monoclonal antibodies and immune sera. We thank Alain Israël (Institut Pasteur, Paris, France) for the kind gift of the 0.4SK-luc plasmid and Charles Hall for critically reading the manuscript.

Supported by a EU grant (Pathogenesis of EBV-related lymphoproliferative disorders in immunocompromised patients contract CHRX-CT94-0651) to M.R. and G.W.B. Partially supported by ANRS (contrat 98017), Association pour la Recherche sur le Cancer, Villejuif, France; contrat 9348, Comité Départemental de la Ligue contre le Cancer; BMBF (01 GE 9609/3); Die Deutsche Forschungsgemeinschaft; and Fonds der Chemischen Industrie. J.F. was supported by a grant from the Ligue Départementale Contre le Cancer (Seine Saint Denis).

1.
Mosialos
G
The role of Rel/NF-kappa B proteins in viral oncogenesis and the regulation of viral transcription
Semin Cancer Biol
1997
8
121
2.
Manet
E
Bourillot
PY
Waltzer
L
Sergeant
A
EBV genes and B cell proliferation
Crit Rev Oncol Hematol
1998
28
129
3.
Grilli
M
Chiu
JJ
Lenardo
MJ
NF-kappa B and Rel: participants in a multiform transcriptional regulatory system
Int Rev Cytol
1993
143
1
4.
Whiteside
ST
Israel
A
I kappa B proteins: structure, function and regulation
Semin Cancer Biol
1997
8
75
5.
DiDonato
JA
Hayakawa
M
Rothwarf
DM
Zandi
E
Karin
M
A cytokine-responsive IkappaB kinase that activates the transcription factor NF-kappaB
Nature
1997
388
548
6.
Woronicz
JD
Gao
X
Cao
Z
Rothe
M
Goeddel
DV
IkappaB kinase-beta: NF-kappaB activation and complex formation with IkappaB kinase-alpha and NIK
Science
1997
278
866
7.
Mercurio
F
Zhu
H
Murray
BW
et al
IKK-1 and IKK-2: cytokine-activated IkappaB kinases essential for NF-kappaB activation
Science
1997
278
860
8.
Zandi
E
Rothwarf
DM
Delhase
M
Hayakawa
M
Karin
M
The IkappaB kinase complex (IKK) contains two kinase subunits, IKKalpha and IKKbeta, necessary for IkappaB phosphorylation and NF-kappaB activation
Cell
1997
91
243
9.
Yamaoka
S
Courtois
G
Bessia
C
et al
Complementation cloning of NEMO, a component of the IkappaB kinase complex essential for NF-kappaB activation
Cell
1998
93
1231
10.
Yamaoka
S
Inoue
H
Sakurai
M
et al
Constitutive activation of NF-kappa B is essential for transformation of rat fibroblasts by the human T-cell leukemia virus type I Tax protein
EMBO J
1996
15
873
11.
Simek
SL
Stephens
RM
Rice
NR
Localization of the v-rel protein in reticuloendotheliosis virus strain T-transformed lymphoid cells
J Virol
1986
59
120
12.
Stephens
RM
Rice
NR
Hiebsch
RR
Bose
HR
Jr
Gilden
RV
Nucleotide sequence of v-rel: the oncogene of reticuloendotheliosis virus
Proc Natl Acad Sci U S A
1983
80
6229
13.
Carrasco
D
Rizzo
CA
Dorfman
K
Bravo
R
The v-rel oncogene promotes malignant T-cell leukemia/lymphoma in transgenic mice
EMBO J
1996
15
3640
14.
Neri
A
Chang
CC
Lombardi
L
et al
B cell lymphoma-associated chromosomal translocation involves candidate oncogene lyt-10, homologous to NF-kappa B p50
Cell
1991
67
1075
15.
Migliazza
A
Lombardi
L
Rocchi
M
et al
Heterogeneous chromosomal aberrations generate 3′ truncations of the NFKB2/lyt-10 gene in lymphoid malignancies
Blood
1994
84
3850
16.
Garatti
SA
Roscetti
E
Trecca
D
Fracchiolla
NS
Neri
A
Berti
E
bcl-1, bcl-2, p53, c-myc, and lyt-10 analysis in cutaneous lymphomas
Recent Results Cancer Res
1995
139
249
17.
Houldsworth
J
Mathew
S
Rao
PH
et al
REL proto-oncogene is frequently amplified in extra-nodal diffuse large cell lymphoma
Blood
1996
87
25
18.
Laherty
CD
Hu
HM
Opipari
AW
Wang
F
Dixit
VM
The Epstein-Barr virus LMP1 gene product induces A20 zinc finger protein expression by activating nuclear factor kappa B
J Biol Chem
1992
267
24,157
19.
Huen
DS
Henderson
SA
Croom-Carter
D
Rowe
M
The Epstein-Barr virus latent membrane protein-1 (LMP1) mediates activation of NF-kappa B and cell surface phenotype via two effector regions in its carboxy-terminal cytoplasmic domain
Oncogene
1995
10
549
20.
Mitchell
T
Sugden
B
Stimulation of NF-kappa B-mediated transcription by mutant derivatives of the latent membrane protein of Epstein-Barr virus
J Virol
1995
69
2968
21.
Kieser
A
Kilger
E
Gires
O
Ueffing
M
Kolch
W
Hammerschmidt
W
Epstein-Barr virus latent membrane protein-1 triggers AP-1 activity via the c-Jun N-terminal kinase cascade
EMBO J
1997
16
6478
22.
Gires
O
Zimber-Strobl
U
Gonnella
R
et al
Latent membrane protein 1 of Epstein-Barr virus mimics a constitutively active receptor molecule
EMBO J
1997
16
6131
23.
Hatzivassiliou
E
Miller
WE
Raab-Traub
N
Kieff
E
Mosialos
G
A fusion of the EBV latent membrane protein-1 (LMP1) transmembrane domains to the CD40 cytoplasmic domain is similar to LMP1 in constitutive activation of epidermal growth factor receptor expression, nuclear factor-kappa B, and stress-activated protein kinase
J Immunol
1998
160
1116
24.
Devergne
O
Hatzivassiliou
E
Izumi
KM
et al
Association of TRAF1, TRAF2, and TRAF3 with an Epstein-Barr virus LMP1 domain important for B-lymphocyte transformation: role in NF-kappaB activation
Mol Cell Biol
1996
16
7098
25.
Izumi
KM
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-kappaB
Proc Natl Acad Sci U S A
1997
94
12,592
26.
Sylla
BS
Hung
SC
Davidson
DM
et al
Epstein-Barr virus-transforming protein latent infection membrane protein 1 activates transcription factor NF-kappaB through a pathway that includes the NF-kappaB-inducing kinase and the IkappaB kinases IKKalpha and IKKbeta
Proc Natl Acad Sci U S A
1998
95
10,106
27.
Traenckner
EB
Pahl
HL
Henkel
T
Schmidt
KN
Wilk
S
Baeuerle
PA
Phosphorylation of human I kappa B-alpha on serines 32 and 36 controls I kappa B-alpha proteolysis and NF-kappa B activation in response to diverse stimuli
EMBO J
1995
14
2876
28.
Gossen
M
Freundlieb
S
Bender
G
Muller
G
Hillen
W
Bujard
H
Transcriptional activation by tetracyclines in mammalian cells
Science
1995
268
1766
29.
Sugden
B
Marsh
K
Yates
J
A vector that replicates as a plasmid and can be efficiently selected in B-lymphoblasts transformed by Epstein-Barr virus
Mol Cell Biol
1985
5
410
30.
Yates
JL
Warren
N
Sugden
B
Stable replication of plasmids derived from Epstein-Barr virus in various mammalian cells
Nature
1985
313
812
31.
Gossen
M
Bujard
H
Tight control of gene expression in mammalian cells by tetracycline-responsive promoters
Proc Natl Acad Sci U S A
1992
89
5547
32.
Le Bail
O
Schmidt-Ullrich
R
Israel
A
Promoter analysis of the gene encoding the I kappa B-alpha/MAD3 inhibitor of NF-kappa B: positive regulation by members of the rel/NF-kappa B family
EMBO J
1993
12
5043
33.
Feuillard
J
Gouy
H
Bismuth
G
Lee
LM
Debre
P
Korner
M
NF-kappa B activation by tumor necrosis factor alpha in the Jurkat T cell line is independent of protein kinase A, protein kinase C, and Ca(2+)-regulated kinases
Cytokine
1991
3
257
34.
Feuillard
J
Dargemont
C
Ferreira
V
et al
Nuclear Rel-A and c-Rel protein complexes are differentially distributed within human thymocytes
J Immunol
1997
158
2585
35.
Wu
F
Garcia
J
Mitsuyasu
R
Gaynor
R
Alterations in binding characteristics of the human immunodeficiency virus enhancer factor
J Virol
1988
62
218
36.
Porzio
MA
Pearson
AM
Improved resolution of myofibrillar proteins with sodium dodecyl sulfate–polyacrylamide gel electrophoresis
Biochim Biophys Acta
1977
490
27
37.
Cooper
T
Electrophoresis
Ed
TC
The Tools of Biochemistry
New York
John Wiley & Sons
1977
194
38.
Jaffray
E
Wood
KM
Hay
RT
Domain organization of I kappa B alpha and sites of interaction with NF-kappa B p65
Mol Cell Biol
1995
15
2166
39.
Lowe
SW
Ruley
HE
Jacks
T
Housman
DE
p53-dependent apoptosis modulates the cytotoxicity of anticancer agents
Cell
1993
74
957
40.
Beg
AA
Baltimore
D
An essential role for NF-kappaB in preventing TNF-alpha-induced cell death
Science
1996
274
782
41.
Wang
CY
Mayo
MW
Baldwin
AS
Jr
TNF- and cancer therapy-induced apoptosis: potentiation by inhibition of NF-kappaB
Science
1996
274
784
42.
Van Antwerp
DJ
Martin
SJ
Kafri
T
Green
DR
Verma
IM
Suppression of TNF-alpha-induced apoptosis by NF-kappaB
Science
1996
274
787
43.
Henderson
S
Rowe
M
Gregory
C
et al
Induction of bcl-2 expression by Epstein-Barr virus latent membrane protein 1 protects infected B cells from programmed cell death
Cell
1991
65
1107
44.
Rowe
M
Peng-Pilon
M
Huen
DS
et al
Upregulation of bcl-2 by the Epstein-Barr virus latent membrane protein LMP1: a B-cell-specific response that is delayed relative to NF-kappa B activation and to induction of cell surface markers
J Virol
1994
68
5602
45.
Mosialos
G
Birkenbach
M
Yalamanchili
R
VanArsdale
T
Ware
C
Kieff
E
The Epstein-Barr virus transforming protein LMP1 engages signaling proteins for the tumor necrosis factor receptor family
Cell
1995
80
389
46.
Roberts
ML
Cooper
NR
Activation of a ras-MAPK-dependent pathway by Epstein-Barr virus latent membrane protein 1 is essential for cellular transformation
Virology
1998
240
93
47.
Picard
D
Salser
SJ
Yamamoto
KR
A movable and regulable inactivation function within the steroid binding domain of the glucocorticoid receptor
Cell
1988
54
1073
48.
Eilers
M
Picard
D
Yamamoto
KR
Bishop
JM
Chimaeras of myc oncoprotein and steroid receptors cause hormone-dependent transformation of cells
Nature
1989
340
66
49.
Kempkes
B
Spitkovsky
D
Jansen-Durr
P
et al
B-cell proliferation and induction of early G1-regulating proteins by Epstein-Barr virus mutants conditional for EBNA2
EMBO J
1995
14
88
50.
Scheinman
RI
Gualberto
A
Jewell
CM
Cidlowski
JA
Baldwin
AS
Jr
Characterization of mechanisms involved in transrepression of NF-kappa B by activated glucocorticoid receptors
Mol Cell Biol
1995
15
943
51.
Shyamala
G
Glucocorticoid receptors in mouse mammary tumors: specific binding to nuclear components
Biochemistry
1975
14
437
52.
Gossen
M
Bonin
AL
Bujard
H
Control of gene activity in higher eukaryotic cells by prokaryotic regulatory elements
Trends Biochem Sci
1993
18
471
53.
Resnitzky
D
Gossen
M
Bujard
H
Reed
SI
Acceleration of the G1/S phase transition by expression of cyclins D1 and E with an inducible system
Mol Cell Biol
1994
14
1669
54.
Polack
A
Feederle
R
Klobeck
G
Hortnagel
K
Regulatory elements in the immunoglobulin kappa locus induce c-myc activation and the promoter shift in Burkitt’s lymphoma cells
EMBO J
1993
12
3913
55.
Polack
A
Hortnagel
K
Pajic
A
et al
c-myc activation renders proliferation of Epstein-Barr virus (Ebv)-transformed cells independent of Ebv nuclear antigen 2 and latent membrane protein 1
Proc Natl Acad Sci U S A
1996
93
10,411
56.
Kempkes
B
Pich
D
Zeidler
R
Sugden
B
Hammerschmidt
W
Immortalization of human B lymphocytes by a plasmid containing 71 kilobase pairs of Epstein-Barr virus DNA
J Virol
1995
69
231
57.
Kilger
E
Kieser
A
Baumann
M
Hammerschmidt
W
Epstein-Barr virus-mediated B-cell proliferation is dependent upon latent membrane protein 1, which simulates an activated CD40 receptor
EMBO J
1998
17
1700
58.
Imbert
V
Rupec
RA
Livolsi
A
et al
Tyrosine phosphorylation of I kappa B-alpha activates NF-kappa B without proteolytic degradation of I kappa B-alpha
Cell
1996
86
787
59.
Schwarz
EM
Van Antwerp
D
Verma
IM
Constitutive phosphorylation of IkappaBalpha by casein kinase II occurs preferentially at serine 293: requirement for degradation of free IkappaBalpha
Mol Cell Biol
1996
16
3554
60.
Hinz
M
Krappmann
D
Eichten
A
Heder
A
Scheidereit
C
Strauss
M
NF-kappaB function in growth control: regulation of cyclin D1 expression and G0/G1-to-S-phase transition
Mol Cell Biol
1999
19
2690
61.
Sugano
N
Chen
W
Roberts
ML
Cooper
NR
Epstein-Barr virus binding to CD21 activates the initial viral promoter via NF-kappaB induction
J Exp Med
1997
186
731
62.
Wang
CY
Mayo
MW
Korneluk
RG
Goeddel
DV
Baldwin
AS
Jr
NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation
Science
1998
281
1680
63.
Jeremias
I
Kupatt
C
Baumann
B
Herr
I
Wirth
T
Debatin
KM
Inhibition of nuclear factor kappaB activation attenuates apoptosis resistance in lymphoid cells
Blood
1998
91
4624
64.
Wu
M
Lee
H
Bellas
RE
et al
Inhibition of NF-kappaB/Rel induces apoptosis of murine B cells
EMBO J
1996
15
4682
65.
Asso-Bonnet
M
Feuillard
J
Ferreira
V
et al
Relationship between IkappaBalpha constitutive expression, TNFalpha synthesis, and apoptosis in EBV-infected lymphoblastoid cells
Oncogene
1998
17
1607
66.
Camilleri-Broet
S
Davi
F
Feuillard
J
et al
High expression of latent membrane protein 1 of Epstein-Barr virus and BCL-2 oncoprotein in acquired immunodeficiency syndrome-related primary brain lymphomas
Blood
1995
86
432
67.
Kenney
JL
Guinness
ME
Curiel
T
Lacy
J
Anti-sense to the epstein-barr virus (EBV)-encoded latent membrane protein 1 (LMP-1) suppresses LMP-1 and bcl-2 expression and promotes apoptosis in EBV-immortalized B cells
Blood
1998
92
1721
68.
Vaux
D
Korsmeyer
S
Cell death in development
Cell
1999
96
245
69.
Levine
AJ
p53, the cellular gatekeeper for growth and division
Cell
1997
88
323
70.
Miyashita
T
Reed
JC
Tumor suppressor p53 is a direct transcriptional activator of the human bax gene
Cell
1995
80
293
71.
Miyashita
T
Harigai
M
Hanada
M
Reed
JC
Identification of a p53-dependent negative response element in the bcl-2 gene
Cancer Res
1994
54
3131
72.
Krikos
A
Laherty
CD
Dixit
VM
Transcriptional activation of the tumor necrosis factor alpha-inducible zinc finger protein, A20, is mediated by kappa B elements
J Biol Chem.
1992
267
17,971
73.
Fries
KL
Miller
WE
Raab-Traub
N
Epstein-Barr virus latent membrane protein 1 blocks p53-mediated apoptosis through the induction of the A20 gene
J Virol
1996
70
8653

Author notes

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

Sign in via your Institution