Interaction between viral proteins and tumor suppressor p53 is a common mechanism of viral pathogenesis. The Epstein-Barr virus (EBV) BZLF-1 ORF-encoded ZEBRA protein (also denoted EB1, Z, Zta) binds to p53 in vitro and has been associated with the altered transcription of p53-regulated genes in B lymphocytes and epithelial cells. In this work, Jurkat T-lymphoblastoid cells that express ZEBRA were characterized by the use of transiently transfected p53 and p53 reporter genes. Stable expression of ZEBRA was associated with the activation of p53-dependent transcription and increased p53 dependent apoptotic cell death. In Jurkat cell lines, stably expressed ZEBRA protein was apparently localized to the cell cytoplasm, in contrast to the typical nuclear localization of this protein in other cell types. Previous studies have suggested that EBV infection of T lymphocytes may contribute to the malignant transformation of T cells and the increased replication of human immunodeficiency virus. Our observations suggest a mechanism through which ZEBRA protein expressed in human T lymphocytes could alter T-cell proliferation and apoptosis during EBV infection.

The p53 tumor suppressor serves as a checkpoint for DNA and cellular damage in many cell types.1-3 p53 is a transcription factor4-6 that directly activates other genes such as the cyclin-dependent kinase inhibitor p21.7,8Levels of p53 protein are regulated in part by binding between p53 and the mdm2 gene product that targets p53 for ubiquitin-dependent degradation.9 p53-dependent transcription is controlled by phosphorylation-regulated conformational changes in the protein.10 Altered p53 activation that results from hereditary conditions such as ataxia–telangiectasia11 or that exists in mice lacking p53 expression12 is associated with increased malignancy. In lymphocytes, p53 may play a particularly important role as a checkpoint for DNA recombination errors because these cells undergo site-specific recombination during generation of the T- and B-cell repertoire.

Oncogenic viruses often encode proteins that are associated with altered p53 function and altered cellular apoptosis.13,14The p53 tumor suppressor interacts with proteins expressed by common human viruses, including adenovirus,15,16papillomavirus,17-19 human T-cell lymphoma virus,20 human immunodeficiency virus (HIV),21-25and herpes viruses including cytomegalovirus,26human herpes virus 6,27 and Epstein-Barr virus (EBV).28-30 Interactions between p53 and viral proteins have been suggested to regulate viral replication.13,14Activation or altered regulation of pro-apoptotic proteins, such as p53, by viral gene products may result in selection for cells lacking functional p53 or p53-regulated gene products. For example, mutated transcriptionally inactive p53 is usually present in EBV-associated Burkitt lymphoma.31 

ZEBRA protein (also termed Z, Zta, and EB1) encoded by the EBV–BZLF-1 open reading frame binds physically to p53.28 Binding between ZEBRA and p53 occurs in part through sequences in the carboxyl-terminus dimerization domain of the ZEBRA protein and the carboxyl terminus of p53. ZEBRA is a site-specific transcription factor required for activation of the viral lytic cycle in lymphocytes32 that can bind and activate the transcription of AP-1 sites in the host genome and viral sequences resembling AP-1 sites.33,34 ZEBRA also binds to other endogenous transcription factors, including NF-κB p65,35 retinoic acid receptors RAR and RXR,36 the TATA binding protein TFIID,37 and the C/EBP transcription factor.38In EBV genome-positive B-lymphoblastoid cells, the coexpression of p53 and ZEBRA leads to a decrease in the transcription of genes by p53.28 In epithelial cell lines, the coexpression of p53 and ZEBRA leads to an increase in the transcription of genes by p53.29 Discordance between the consequences of ZEBRA expression in these 2 experimental systems has been attributed to differences in metabolism or activation of p53.29 Other gene products encoded by EBV—the LMP-1 and EBNA-2 proteins expressed during viral latency—also increase p53 expression in transformed primary B lymphocytes.30 

Previously, we showed that the cell cycle-regulated expression of p53 is evident during the activation of primary peripheral human T cells.39 Detection of EBV genome and gene expression in T-cell lymphoma40-42 suggests that T cells are targets of EBV infection. In contrast to most EBV genome-positive T-cell tumors that express latency-associated gene products,41,43 lytic gene products including ZEBRA are expressed in human thymocytes.43 We suggested a model of EBV infection of T cells in which the infection of primary T cells with EBV leads to the abortive transcription of EBV lytic gene products that can rarely lead to malignant transformation and stable expression of latent gene products.44 A clinical syndrome has been described in which chronic, active EBV infection, including expression of the ZEBRA protein in T cells, was associated with malignant T-cell lymphoma.42 Increased lytic replication of EBV in patients with immunodeficiency caused by HIV45 and congenital immunodeficiency syndromes or chronic fatigue syndrome46 could also lead interaction between ZEBRA and p53 in T cells. Therefore, we characterized the effects of ZEBRA expression on p53-dependent transcription in Jurkat T-lymphoblastoid cells that can be infected with EBV in vitro, through a receptor similar, but not identical, to the CD21 EBV receptor present on B lymphocytes and thymocytes.47 

Jurkat T-lymphoblastoid cell lines stably expressing ZEBRA protein were established. Both wild-type and mutated p53 were introduced into Jurkat cells using nontoxic lipid reagents. Transcriptional activity of p53 was monitored with reporter genes, including synthetic reporter genes regulated by the p53 consensus binding site5,6 and the more complex endogenous p21 promoter.7 We found that levels of p53 protein detected by Western blotting were increased in Jurkat cells stably expressing ZEBRA and that the transcription of p53 reporter genes was markedly increased. Populations of Jurkat cells expressing ZEBRA transfected with p53 also exhibited increased apoptotic cell death that was p53-dependent. Mechanisms through which interaction with the ZEBRA protein might increase p53 stability and activation are discussed, such as alterations in the ubiquitin-dependent pathways that degrade p539,48,49 and the activation of p53 through regulatory sequences in the p53 carboxyl-terminus.10 We hypothesize that the activation of p53-dependent transcription in EBV-infected T lymphocytes may inactivate T cells required for the control of EBV infection50 as a mechanism of viral pathogenesis.

Plasmids

pSV2-neo-WZhet ZEBRA expression plasmid and control plasmid pSV2-neo32 were obtained from Dr G. Miller (Yale University, New Haven, CT). Plasmids encoding wild-type p53 (pC53-SN3, denoted p53W), DNA binding mutant p53 (pC53-SCX3, denoted p53N), and reporter genes pG13PYluc, pMG15, and p21 WWP/luc7 were obtained from Dr B. Vogelstein (Johns Hopkins University, Baltimore, MD). A plasmid encoding carboxyl terminus deleted p53 (pCEP4-353, denoted p53C) was obtained from Dr J. Pietenpol (Tulane University, Nashville, TN). pRL-SV40 was obtained from Promega Biological (Madison, WI). Plasmids were prepared using the Qiagen method (Qiagen, Chatsworth, CA).

Cell culture and cell lines

Jurkat (JEG-1) cells were obtained from the ATCC (Rockville, MD). Cells were cultured in RPMI medium supplemented with 10% fetal calf serum, penicillin (100 U/mL), and L-glutamine (2 mmol/L). Stably transformed Jurkat cell lines were obtained by the transfection of cells with pSV2-neo-WZhet ZEBRA expression plasmid and control plasmid pSV2-neo by the use of Cell–Fectin (Gibco/BRL, Gaithersburg, MD). Cells were cloned in soft agar using selection for G418 resistance (100 μg/mL) and grown in 200 μg/mL G418. Two independently derived cell lines expressing similar levels of ZEBRA protein, as determined by Western blot analysis and denoted the Z1 and ZA cell lines, were used for experiments, with similar results noted between the 2 cell lines in all transcription experiments. The Z1 cell line was used for immunoprecipitation and p53 expression studies. The ZA cell line was used for apoptosis experiments and for transfection studies (data not shown). The parental strain of Jurkat cells used in these studies lacked the expressed, endogenous p53 detectable by Western blotting and exhibited minimal transcription of p53-responsive reporter genes in the absence of transiently transfected p53 expression vectors (data not shown).

Western blot analysis

Whole-cell lysates were generated by the lysis of cells in a hypertonic RIPA buffer containing 25 mmol/L Tris pH 7.5, 2% NP-40, 0.2% sodium dodecyl sulfate (SDS), 150 mmol/L NaCl, 0.5% sodium deoxycholate (SDC), and 10% vol/vol glycerol and protease inhibitors aprotinin (20 μg/mL), leupeptin (10 μg/mL), and phenymethylsulfonyl fluoride (1 mmol/L). For immunoprecipitation, Z+ and Z− cells were lysed under nondenaturing conditions in a buffer containing 0.15% NP-40, 20 mmol/L HEPES pH 7.5, 70 mmol/L KCl, 2 mmol/L MgCl2, and 1 mmol/L dithiothreitol and protease inhibitors. Lysates were bound to anti-EBV human sera (1/200 dilution of human anti-serum) and precipitated with washed protein G plus/A Sephadex beads (Oncogene Science, Santa Barbara, CA). Lysates of cytoplasmic protein were generated by the lysis of cells in a hypotonic buffer containing 25 mmol/L Tris pH 7.5, 2% NP-40, 0.2% SDS, 50 mmol/L NaCl, and 0.5% SDC. Nuclear extracts were prepared as described.51 After separation of proteins by 12% SDS–PAGE and transfer to nitrocellulose as described previously,43Western blots were bound to antibody (1/1000 dilution of primary antibody OT20A, 1/10,000 dilution of secondary goat antimouse antiserum [Amersham, Arlington Heights, IL]). Western blots were developed using the Renaissance system (NEN, Boston, MA). Anti-ZEBRA murine monoclonal antibody OT20A was obtained from Dr J. Middledorp (Organon–Teknika, The Netherlands) and reactivity of OT20A to ZEBRA protein was confirmed using Akata B-lymphoblastoid cells induced into the lytic cycle by the ligation of surface IgG (data not shown). Anti-p53 (whole p53 protein) rabbit antiserum was obtained from Santa Cruz Biologicals (Santa Cruz, CA).

Cell morphology/immunofluorescence studies

For a demonstration of the altered Z+ cell morphology Z+ (Z1) and Z− cells were photographed 24 hours after plating in standard polystyrene plastic ware (Falcon; Becton Dickinson, Franklin Lakes, NJ). Cells were plated in fresh media and were not otherwise stimulated. Under these conditions, both Z+ cell lines (Z1 and ZA) had a similar adherent phenotype as shown, whereas Z− cells and parental Jurkat cells had a nonadherent phenotype typical of Jurkat cells (ZA and parental Jurkat cell data not shown). For immunofluorescence studies, Z+ and Z− cells were bound to poly-D-lysine–coated coverslips as described previously for T lymphocytes.52 Cells were incubated with rabbit polyclonal antisera (ZEBRA rabbit antisera generated against bacterially produced whole ZEBRA protein/TrpE fusion protein; obtained from Dr G. Miller) at a 1/1000 dilution. Cells were then washed and incubated with biotinylated donkey antirabbit secondary antibody (Jackson Research, South Park, PA), washed, and incubated with streptavidin Cy3 (Jackson Research) used at a 1/180 dilution. Cells were photographed using a Nikon Diaphot 60× oil immersion lens. Data were collected using IP Lab Spectrum software (Signal Analytics, Vienna, VA).

Transfection of cells

Reporter plasmids and p53 expression plasmids (concentration as indicated in the text) were transiently transfected into 2 × 106 logarithmically growing Z+ Jurkat cells stably expressing ZEBRA and Z-control cells with Superfectin (Qiagen) following the recommended procedure for the transfection of nonadherent cells. After transfection, cells were cultured for 36 hours. In experiments shown, cells were also transfected with a plasmid expressing renilla luciferase to control for cell viability and transformation efficiency. Similar results were obtained without the cotransfection of pRL-SV40 and with luciferase activity normalized to micrograms of total protein, determined by the Bradford protein assay (Bio-Rad, Hercules, CA). Cells were irradiated using a hand-held UVB source (Ultraviolet Products, San Gabriel, CA) 3 cm above the washed cells suspended in 1 mm phosphate-buffered saline in a culture dish. For transient transfection of the Jurkat cells shown, a 2-stage transient transfection was used in which ZEBRA expression plasmid and p53 reporter genes were transfected 24 hours before the transfection of p53 expression plasmid to reduce the potential suppression of ZEBRA expression by the coexpression of p53.

Luciferase assay

Luciferase activity was determined either using the firefly luciferase or Stop-and-Glo assay systems (Promega Biologicals) and an Analytical Luminescence Laboratory (San Diego, CA) luminometer. Results of at least 3 separate experiments were used to generate each data point in luciferase assay experiments. Mean luciferase activity and standard error were determined, as shown graphically and analyzed using the JMP Statistical Discovery Software Version 3.1 (SAS Institute, Cary, NC). Student t test was used for comparison of experiments, and significance (P < .05) was determined by the JMP program.

Detection of apoptosis in Jurkat cells

Exponentially growing Z+ and Z− cells 2 × 106 were transiently transfected with 1 μg wild-type p53 as described above. Cells were stained with propidium iodide and annexin–fluorescein isothiocyanate (FITC) of apoptotic cell death using Apo-alert (Clontech, Palo Alto, CA) as instructed by the manufacturer. Apoptotic cells were quantitated in 3 independent experiments for each data point shown by fluorescence-activated cell sorting (FACS) of 2000 cells using a Coulter (Hialeah, FL) Epics XL. In some experiments 0.5 μg of a plasmid expressing green fluorescent protein (Green Lantern; New England Biolabs, Boston, MA) was transfected into Z+ and Z− cells to determine transfection efficiency, and approximately 10% of transfected cells expressed glial fibrillary protein under conditions used in these studies (data not shown).

Characterization of Jurkat cells stably expressing ZEBRA protein

Jurkat T-lymphoblastoid cell lines stably transfected with the ZEBRA expression plasmid pSV2-neo-WZhet were established by selection for growth in G418 selection medium. pSV2-neo-WZhet has previously been shown to express a functional 43-kd variant of the ZEBRA protein that is sufficient to activate the EBV lytic cycle.32 Jurkat cells transfected with pSV2-neo-WZhet (denoted Z+ cells) grown in G418 expressed a 43-kd protein detected by ZEBRA-specific monoclonal antibody OT20A (Figure 1). This 43-kd putative ZEBRA protein was not detected in Jurkat cells stably transfected with control plasmid pSV2-neo (control cells denoted Z− cells; Figure 1A).

Fig. 1.

Characterization of ZEBRA protein expression in Jurkat cell lines.

(A) A novel 43-kd protein (denoted ZEBRA) was detected by monoclonal antibody OT20A only in cells transfected with pSV2-neo-WZhet expression plasmid but not control cells transfected with plasmid pSV2-neo. Whole-cell lysates were generated in hypertonic RIPA buffer from COS cells and 2 independently derived Jurkat cell lines transfected with pSV2-neo-WZhet (Z+) and a Jurkat cell line expressing control plasmid (Z−). (B) A novel 43-kd protein was detected in proteins precipitated from Z+ cells but not Z− cells with polyclonal human antiserum (Figure 1A) and probed with OT20A. (C) Z+ and Z− cells were analyzed by immunofluorescence with rabbit polyclonal serum raised against purified ZEBRA protein. Staining was evident primarily in the cytoplasm of Z+ but not Z− cells. Z− and Z+ cells were analyzed by immunofluorescence with a rabbit polyclonal serum raised against purified ZEBRA protein. Staining was evident in the cytoplasm of Z+ but not Z− cells. (D) A novel 43-kd protein was detected with monoclonal antibody OT20A in proteins extracted from Z+ cells but not Z− cells in hypotonic RIPA buffer. Hypotonic lysis buffer was used to extract cytoplasmic proteins selectively from Z+ and Z− cells. Transient transfection of plasmid pC53-SCX3 encoding DNA binding mutant p53 denoted p53N, plasmid pC53-SN3 encoding wild-type p53 denoted p53W, and pCEP4-353 encoding carboxyl terminus deleted p53 denoted p53C did not alter expression of putative 43-kd ZEBRA protein. (E) Z+ Jurkat cells demonstrated increased adherence to plastic ware and an ameboid morphology in comparison to Z− cells grown under identical conditions.

Fig. 1.

Characterization of ZEBRA protein expression in Jurkat cell lines.

(A) A novel 43-kd protein (denoted ZEBRA) was detected by monoclonal antibody OT20A only in cells transfected with pSV2-neo-WZhet expression plasmid but not control cells transfected with plasmid pSV2-neo. Whole-cell lysates were generated in hypertonic RIPA buffer from COS cells and 2 independently derived Jurkat cell lines transfected with pSV2-neo-WZhet (Z+) and a Jurkat cell line expressing control plasmid (Z−). (B) A novel 43-kd protein was detected in proteins precipitated from Z+ cells but not Z− cells with polyclonal human antiserum (Figure 1A) and probed with OT20A. (C) Z+ and Z− cells were analyzed by immunofluorescence with rabbit polyclonal serum raised against purified ZEBRA protein. Staining was evident primarily in the cytoplasm of Z+ but not Z− cells. Z− and Z+ cells were analyzed by immunofluorescence with a rabbit polyclonal serum raised against purified ZEBRA protein. Staining was evident in the cytoplasm of Z+ but not Z− cells. (D) A novel 43-kd protein was detected with monoclonal antibody OT20A in proteins extracted from Z+ cells but not Z− cells in hypotonic RIPA buffer. Hypotonic lysis buffer was used to extract cytoplasmic proteins selectively from Z+ and Z− cells. Transient transfection of plasmid pC53-SCX3 encoding DNA binding mutant p53 denoted p53N, plasmid pC53-SN3 encoding wild-type p53 denoted p53W, and pCEP4-353 encoding carboxyl terminus deleted p53 denoted p53C did not alter expression of putative 43-kd ZEBRA protein. (E) Z+ Jurkat cells demonstrated increased adherence to plastic ware and an ameboid morphology in comparison to Z− cells grown under identical conditions.

Close modal

Levels of putative ZEBRA protein in several independent Jurkat cell lines were uniformly lower than in nonlymphoid cell lines, such as COS cells transfected with pSV2-neo-WZhet (Figure 1A). To confirm that the 43-kd protein was in fact ZEBRA rather than a cross-reactive cellular protein, we also demonstrated that the putative ZEBRA protein was highly enriched among the proteins precipitated from Z+ cells by human serum reactive against EBV lytic gene products (Figure 1B). Precipitated 43-kd protein appeared as a closely spaced dimer, consistent with the appearance of WZhet ZEBRA expressed in B lymphocytes33 and in EBV-infected primary human thymocytes.43 In these coprecipitation experiments, components of NF-κB, including p65, were also selectively enriched by coprecipitation with anti-ZEBRA antisera, consistent with the previous observation that ZEBRA and p65 form a complex when coexpressed in lymphoid cells.35 ZEBRA protein was also detected in Z+, but not in Z−, cells using rabbit polyclonal antisera generated against purified bacterially expressed ZEBRA protein in immunofluorescence studies (Figure 1C).

In immunofluorescence studies ZEBRA protein was detected largely in the cytoplasm of Z+ but not Z− cells (Figure 1C). A cytoplasmic location for the protein was also suggested by fractionation experiments in which ZEBRA protein was detected in lysates from Z+ cells but not Z− cells generated in a hypotonic lysis buffer (Figure 1D). ZEBRA protein was not detected in the nucleus of Jurkat cells by Western blotting of nuclear extracts (data not shown). These observations suggested that when it was expressed stably in Jurkat cells, ZEBRA protein was localized to the cell cytoplasm, where it maintained its ability to associate with other cytoplasmic proteins. The cytoplasmic localization of ZEBRA protein was in contrast to the nuclear localization of the protein in EBV-positive Akata B-lymphoblastoid cells determined using similar fractionation and staining techniques (data not shown). Jurkat cells stably transfected with pSV2-neo-Wzhet also exhibited a distinct cytoplasmic cellular phenotype of increased adherence to plastic ware and an ameboid morphology not present in control cells transfected with pSV2-neo grown under identical conditions (Figure 1E).

Expression of transiently transfected p53 protein in Jurkat cells stably expressing ZEBRA protein

Because of the previously described effects of ZEBRA protein on p53-dependent gene transcription,28-30 we determined the effects of ZEBRA expression on p53 stability and p53-dependent gene transcription in Z+ cells in comparison to control Z− Jurkat cells. Endogenous p53 protein was not detected in the parental Jurkat cells used to generate Z+ and Z− cell lines (data not shown). To introduce p53 expression, Z+ and Z− cells were transiently transfected with 1 μg of expression vectors encoding wild-type p53 (pC53-SN3, denoted p53W), p53 amino acids 1 to 353 (pCEP4-353, deleted from p53 C-terminal regulatory sequences; denoted p53C), and a DNA binding null mutant form of p53 (pC53-SCX3, denoted p53N). Expression of cytoplasmic ZEBRA protein did not vary significantly with transient transfection of p53 expression vectors (Figure 1D). With the transient transfection of p53 expression vectors, p53 protein encoded by wild-type p53 expression plasmid and the pCEP4-353 C-terminal deleted protein was detected in proteins extracted from the nucleus of Z+ cells in hypertonic buffer (Figure 2A). Expression of p53C was markedly less than the expression of p53W and was barely detectable by Western blotting. Remarkably, transiently expressed p53W or other p53 proteins was not detected in the nuclear proteins of Z− cells under these conditions (Figure 2A). Transiently expressed p53 proteins were not detected in cytoplasmic proteins extracted from Z+ or Z− cells in hypotonic lysis buffer, with the possible exception of p53N, which was evident as a very faint band in the cytoplasm of both Z+ and Z− cells (Figure 2B).

Fig. 2.

Characterization of p53 protein expression in Jurkat cell lines expressing ZEBRA protein (Z+) and control cells (Z−).

B-lymphoblastoid Namalwa cells (NA) used as a positive control for detection of p53 protein. (A) p53 protein was detected by Western blotting using a polyclonal p53-specific anti-sera in nuclear protein extracts prepared from Z+ cells transiently transfected with p53W and p53C expression plasmids, but not in nuclear extracts from Z+ cells transfected with p53N expression plasmid or in nuclear extracts from Z− cells transfected with any p53 expression vectors. Levels of carboxyl terminus deleted p53 protein (53C) were significantly less than wild-type protein (53W) in Z+ cells, and the expected location of p53C protein is indicated (*). (B) p53 protein was not detected by Western blotting using a polyclonal p53-specific anti-sera in cytoplasmic-protein enriched hypotonic lysates of Z+ cells in the presence of transfected p53 expression plasmids with the possible exception of a small amount of p53N detected in both Z+ and Z− cell lysates (**). Hypotonic lysates used in this figure contained detectable ZEBRA protein (Figure 1D).

Fig. 2.

Characterization of p53 protein expression in Jurkat cell lines expressing ZEBRA protein (Z+) and control cells (Z−).

B-lymphoblastoid Namalwa cells (NA) used as a positive control for detection of p53 protein. (A) p53 protein was detected by Western blotting using a polyclonal p53-specific anti-sera in nuclear protein extracts prepared from Z+ cells transiently transfected with p53W and p53C expression plasmids, but not in nuclear extracts from Z+ cells transfected with p53N expression plasmid or in nuclear extracts from Z− cells transfected with any p53 expression vectors. Levels of carboxyl terminus deleted p53 protein (53C) were significantly less than wild-type protein (53W) in Z+ cells, and the expected location of p53C protein is indicated (*). (B) p53 protein was not detected by Western blotting using a polyclonal p53-specific anti-sera in cytoplasmic-protein enriched hypotonic lysates of Z+ cells in the presence of transfected p53 expression plasmids with the possible exception of a small amount of p53N detected in both Z+ and Z− cell lysates (**). Hypotonic lysates used in this figure contained detectable ZEBRA protein (Figure 1D).

Close modal

The ability of pC53-SCX3 to express a stable transcriptionally inactive p53 protein was confirmed in a human neuroblastoma cell line (data not shown). Mutant, transcriptionally inactive p53 alleles, such as the protein encoded by pC53-SCX3, were stabilized, and protein levels protein were increased in many cell types,4-6 possibly accounting for the small amount of p53N protein transiently expressed in the cytoplasm of Z+ and Z− cells (Figure 2B). Less carboxyl terminus-deleted p53 protein was evident in Z+ cells than in wild-type p53, though the deletion of the carboxyl terminus of p53 has been reported10 to result in increased p53-transcriptional activity in some cell types. The ability of pCEP4-353 used in these studies to direct p53-mediated transcription of reporter genes approximately 2 times higher than wild-type p53 per microgram of transfected plasmid in EBV-positive B-lymphoblastoid cell lines was also confirmed (data not shown). These observations demonstrated that stable ZEBRA expression in Jurkat cells was associated with increased levels of some nuclear p53 proteins, including wild-type transcriptionally active p53, but not with increased expression of a transcriptionally inactive mutant p53 protein.

Activation of a synthetic p53 reporter gene in Jurkat cells stably expressing ZEBRA protein

In the absence of activation, the p53 protein is normally rapidly degraded by a ubiquitin-dependent pathway.9,48,49 Binding between p53 and viral proteins in human cells can either stabilize or destabilize p53 through interference with p53 metabolism. In many instances, p53, stabilized by viral proteins, is not transcriptionally active.13 The transcriptional activity of transiently transfected p53 was further characterized in Jurkat cells stably expressing ZEBRA using a reporter gene pG13PYluc. pG13PYluc is a synthetic reporter gene in which 13 copies of an active p53 response element are fused to a polyoma virus promoter.7 Production of luciferase by this reporter gene has previously been shown to be a sensitive and specific measure of p53-dependent transcriptional activity.8 Two hundred nanograms of pG13PYluc was introduced transiently with 500 ng of p53 expression vectors into Z+ and Z− Jurkat cells (Figure 3). As in Western blotting studies (Figure 2), a nontoxic transfection reagent (Superfectin; Qiagen) was used to introduce DNA into cells. Cells were not activated in these experiments by radiation or other stimuli, and viability of cells was similar to that of control cells cultured without transfection.

Fig. 3.

p53-dependent transcription determined by luciferase activity of reporter plasmid pG13PYluc.

(A) Jurkat cells stably expressing ZEBRA (Z+) and control cells (Z−) were transiently transfected with 200 ng of reporter plasmids (pG13PYluc or pGM15PYluc) and with 500 ng of p53 expression plasmids for either p53W or p53C. Significant transcription activation was detected with pG13PYluc reporter, but not with mutated pGM15PYluc reporter in Z+ cells with cotransfection of 53W (*P < .05) or 53C (**P < .05). In the absence of cotransfected p53, transcription of p53 reporter genes was extremely low in both Z+ and Z− cells. (B) Parental Jurkat cells were transiently transfected with ZEBRA (500 ng) and p53 expression plasmids (500 ng) with reporter plasmids (200 ng). Transcription of pG13PYluc with cotransfection of p53W was slightly increased by transient cotransfection of pSV2-neo-WZhet ZEBRA expression plasmid (Z+) compared to transient transfection of control plasmid pSV2-neo (Z−), but this increase was not significant (P > .05) in pooled experimental data. Transcription of mutated pGM15PYluc reporter was not activated by transient transfection of either p53 or ZEBRA.

Fig. 3.

p53-dependent transcription determined by luciferase activity of reporter plasmid pG13PYluc.

(A) Jurkat cells stably expressing ZEBRA (Z+) and control cells (Z−) were transiently transfected with 200 ng of reporter plasmids (pG13PYluc or pGM15PYluc) and with 500 ng of p53 expression plasmids for either p53W or p53C. Significant transcription activation was detected with pG13PYluc reporter, but not with mutated pGM15PYluc reporter in Z+ cells with cotransfection of 53W (*P < .05) or 53C (**P < .05). In the absence of cotransfected p53, transcription of p53 reporter genes was extremely low in both Z+ and Z− cells. (B) Parental Jurkat cells were transiently transfected with ZEBRA (500 ng) and p53 expression plasmids (500 ng) with reporter plasmids (200 ng). Transcription of pG13PYluc with cotransfection of p53W was slightly increased by transient cotransfection of pSV2-neo-WZhet ZEBRA expression plasmid (Z+) compared to transient transfection of control plasmid pSV2-neo (Z−), but this increase was not significant (P > .05) in pooled experimental data. Transcription of mutated pGM15PYluc reporter was not activated by transient transfection of either p53 or ZEBRA.

Close modal

As shown in Figure 3A, markedly increased levels of p53-dependent transcription were evident in Jurkat cells stably expressing ZEBRA transfected with a plasmid expressing wild-type p53 (pC53-SN3, denoted p53W). Transfection of a plasmid expressing p53 lacking regulatory sites in the carboxyl terminus of the p53 protein (pCEP4-353, denoted p53C) was also associated with the activation of p53-dependent transcription in cells expressing ZEBRA, though the activation of p53-dependent transcription was less than that with transfection of a plasmid expressing wild-type p53. Levels of pG13PYluc transcription, when quantitated by luciferase assay (Figure 3A), corresponded to levels of p53 protein detected by Western blotting (Figure 2A) in Jurkat cells. Levels of pG13PYluc transcription, with transfection of a transcriptionally inactive p53 (p53N) or with a control plasmid not encoding p53, were similar to basal levels of pG13PYluc transcription in the absence of any p53 expression plasmid transfection (data not shown). Transcription of pG13PYluc by wild-type and carboxyl terminus-deleted p53 was eliminated by a subtle mutation7 in the p53 consensus site (plasmid MG15PYLuc) and thus was specific for the transcriptionally active p53 protein and for the p53 consensus binding site in pG13PYluc (Figure 3A). Transfection of more p53 plasmid into Z+ cells resulted in nonlinear increases in p53-dependent transcription (data not shown), suggesting the quantity of expressed p53 was a limiting factor in p53 expression.

In these experiments, 5 ng pRL/SV40 expressing Renilla luciferase (pRL/SV40), under the control of an SV40 promoter and enhancer, was introduced into cells as a control for transfection efficiency and cell viability. Renilla luciferase transcription was similar in the presence of wild-type p53 in Z+ and Z− cells, and thus differences in transfection efficiency or viability between Z+ and Z− cells could not account for the activation of p53-dependent transcription associated with ZEBRA protein expression. Similar results were found in 2 independently established Z+ Jurkat cell lines and did not differ in the presence or absence of pRL/SV40.

To determine whether transient expression of ZEBRA could reproduce the activation of p53-dependent transcription evident in Jurkat cells stably expressing ZEBRA, the parental Jurkat cell line used to generate cells stably expressing ZEBRA was transiently transfected with ZEBRA expression plasmid pSV2-neo-WZhet, p53W expression plasmid, and p53 reporter genes (Figure 3B). Expression of pG13PYluc was compared between cells transfected with pSV2-neo-WZhet or control plasmid pSV2-neo. In the parental Jurkat cell line, levels of p53-dependent transcription of pG13PYluc were low. Transient coexpression of wild-type ZEBRA and wild-type p53 in Jurkat cells resulted in a trend toward increased transcription of p53 reporter gene pG13PYluc, but this trend was not significant because of greater interexperimental variability with cotransfected ZEBRA expression plasmid and low levels of luciferase activity. These results suggested that either prolonged or high levels of ZEBRA protein were required to activate p53-dependent transcription in Jurkat cells or that the expression of ZEBRA selected for a specific phenotype in Jurkat cells was not present in the parental cells.

Activation of a p21 promoter reporter gene in Jurkat cells stably expressing ZEBRA protein

A luciferase reporter gene regulated by the p21 promoter (plasmid WWP-Luc) was previously used21 to characterize the activation of a physiologic target of the p53 protein in epithelial cells. The p21 promoter is a physiologic target of p53 activation, and it was used in addition to the synthetic reporter, pG13Pyluc, to determine the effects of stable ZEBRA expression in Jurkat cells. Transcription of the p21 promoter was activated by cotransfected wild-type p53 (pC53-SN3, denoted p53W) in Jurkat cells stably expressing ZEBRA but not in Z− cells (Figure4A). In these experiments, synergy was evident between activation of the p53-responsive p21 promoter in Z+ cells and other stimuli, such as ultraviolet irradiation (Figure 4B). A small but significant increase in expression of the p21 promoter was also evident in Z+ cells in the absence of cotransfected p53 (Figure4B) and also in Z− cells, but p53-dependent activation of WWP/Luc was not evident in Z− cells (Z− data not shown). These results demonstrated that stable expression of ZEBRA in Jurkat T-lymphoblastoid cells was associated with p53-dependent activation of the physiologic p21 promoter, and this activation could synergize with other activators of p21 transcription.

Fig. 4.

p53-Dependent transcription determined by luciferase activity of a reporter plasmid (WWP/Luc) regulated by the physiologic p21 promoter.

(A) Activation of the p21 promoter (WWP/Luc) was evident in Z+ cells but not Z− cells transfected with p53W expression plasmid (500 ng) and WWP/Luc reporter plasmid (200 ng) (*P < .05). (B) A significant increase in p21 transcription (*P < .05) was evident in Z+ cells transfected with WWP/Luc (200 ng) and exposed to ultraviolet irradiation without cotransfection of p53. With cotransfection of expression plasmid for p53W (200 ng) and WWP/Luc (200 ng) into Z+ cells, increased transcription of the p21 promoter was evident both in the absence (UV−) and the presence (UV+) of UV irradiation (P < .05 for either UV− [**] or UV+ [***] relative to Z+ cells not transfected with p53 [*]). Synergy between p21 promoter activation by p53W transfection in Z+ cells with ultraviolet irradiation (UV+) versus control cells (UV−) was noted (P < .05 for Z+ cells transfected with WWP/Luc and p53 (UV+ [***]) versus (UV−[**]). Increased transcription of WWP/Luc with transfection of 500 ng of p53 expression plasmid (A, Z+/p53W) relative to 200 ng of p53 expression plasmid (B, Z+/p53W/UV−) was also evident in these experiments.

Fig. 4.

p53-Dependent transcription determined by luciferase activity of a reporter plasmid (WWP/Luc) regulated by the physiologic p21 promoter.

(A) Activation of the p21 promoter (WWP/Luc) was evident in Z+ cells but not Z− cells transfected with p53W expression plasmid (500 ng) and WWP/Luc reporter plasmid (200 ng) (*P < .05). (B) A significant increase in p21 transcription (*P < .05) was evident in Z+ cells transfected with WWP/Luc (200 ng) and exposed to ultraviolet irradiation without cotransfection of p53. With cotransfection of expression plasmid for p53W (200 ng) and WWP/Luc (200 ng) into Z+ cells, increased transcription of the p21 promoter was evident both in the absence (UV−) and the presence (UV+) of UV irradiation (P < .05 for either UV− [**] or UV+ [***] relative to Z+ cells not transfected with p53 [*]). Synergy between p21 promoter activation by p53W transfection in Z+ cells with ultraviolet irradiation (UV+) versus control cells (UV−) was noted (P < .05 for Z+ cells transfected with WWP/Luc and p53 (UV+ [***]) versus (UV−[**]). Increased transcription of WWP/Luc with transfection of 500 ng of p53 expression plasmid (A, Z+/p53W) relative to 200 ng of p53 expression plasmid (B, Z+/p53W/UV−) was also evident in these experiments.

Close modal

Increased apoptosis of cells coexpressing ZEBRA protein and p53

Increased expression of p53 reporter genes including the p21 promoter was demonstrated in cells stably expressing ZEBRA protein transfected with p53W expression plasmid (Figures 3, 4). Although p21 promoter expression was activated by cotransfected p53 (Figure 4A), Western blotting was not sufficiently sensitive to detect a convincing increase in p21 expression in Z+ Jurkat cells transfected with p53W (data not shown), possibly because a small percentage (approximately 10%) of transfected cells expressed transiently transfected genes. Further experiments were designed to determine whether populations of Z+ cells demonstrated evidence of increased apoptosis (programmed cell death) in the presence of p53 expression because p53 induces apoptosis in many cell types through the transcription of gene products, including p21.1-3 

Z+ and Z− cells were transfected with 1 μg of either wild-type p53 (p53W) or DNA binding mutant p53 (p53N) expression plasmids. Cells were incubated for 48 hours and stained with both propidium iodide and annexin–FITC, independent markers for early and late stages of apoptotic cell death, respectively. Increased staining of approximately 10% of cells with both apoptotic markers was evident in populations of Z+ cells transfected with p53W expression plasmid relative to transfection with p53M expression plasmid or to transfection of Z− cells with either p53W or p53M expression plasmids, as shown in Figure 5A. The number of cells staining with either marker relative to total cells was quantitated in 3 independent experiments (Figure 5B). To obtain normalized data shown in Figure 5B, populations of Z+ and Z− cells were transfected with either p53N or p53W expression plasmids, as described above, or with a plasmid driven by a similar cytomegalovirus promoter lacking p53 protein coding sequences (pCMV5). The number of cells exhibiting apoptotic cell death in each population of cells transfected with either p53N or p53W was then determined and expressed as the ratio of apoptotic cells to apoptotic cells detected in a control population of cells transfected with control plasmid. As shown in Figure 5B, only the expression of p53W in Z+ cells resulted in a detectable increase in apoptotic cells compared with control cells (ratio to control cells greater than 1.0 by more than the standard error of measurement) for either annexin or propidium iodide markers. Cell death in either Z+ cells transfected with mutant p53 or Z− cells transfected with wild-type p53 was not different than that in control cells (ratio to pCMV = 1.0). Decreased cell death relative to control cells (ratio to pCMV less than 1.0) was evident in Z− cells transfected with mutant p53.

Fig. 5.

Increased apoptotic cell death in populations of Z+ Jurkat cells transfected with wild-type p53 expression plasmid.

(A) FACS analysis of populations of Z− and Z+ cells transfected with either p53N or p53W. Z+ cells transfected with p53W expression plasmid demonstrated a relative increase in cells staining with both propidium iodide and annexin-FITC antibody (quadrant 3) relative to Z+ cells transfected with p53N expression plasmid or Z− cells transfected with either p53W or p53N expression plasmids. (B) Cell death determined by staining with either propidium iodide (PI) or annexin-FITC antibody (AN) was increased in Z+ cells after transfection of p53W expression plasmid, but it was not increased in Z+ cells transfected with p53N expression plasmid or in Z− cells transfected with either p53N or p53W expression plasmids. Percentage of cells staining with either marker was normalized using equivalent cells transfected with a control plasmid (pCMV5) under identical conditions. In this analysis a ratio of greater than 1.0 indicates increased cell death relative to control, whereas a ratio of less than 1.0 indicates decreased cell death relative to control. (C) Cell death (percentage of cells staining with propidium iodide [PI] per 2000 analyzed cells) was similar in Z− and Z+ cells 24 hours after transfection of p53W expression plasmid, but significantly greater (P < .05 indicated) 48 hours after transfection in Z+ relative to Z− cells. During this interval, normalized p53-dependent transcription of pG13PYluc (inset) increased significantly in Z+ cells (*P < .05 for normalized luciferase activity levels 18 and 36 hours after transfection of p53W and reporter genes into Z+ cells).

Fig. 5.

Increased apoptotic cell death in populations of Z+ Jurkat cells transfected with wild-type p53 expression plasmid.

(A) FACS analysis of populations of Z− and Z+ cells transfected with either p53N or p53W. Z+ cells transfected with p53W expression plasmid demonstrated a relative increase in cells staining with both propidium iodide and annexin-FITC antibody (quadrant 3) relative to Z+ cells transfected with p53N expression plasmid or Z− cells transfected with either p53W or p53N expression plasmids. (B) Cell death determined by staining with either propidium iodide (PI) or annexin-FITC antibody (AN) was increased in Z+ cells after transfection of p53W expression plasmid, but it was not increased in Z+ cells transfected with p53N expression plasmid or in Z− cells transfected with either p53N or p53W expression plasmids. Percentage of cells staining with either marker was normalized using equivalent cells transfected with a control plasmid (pCMV5) under identical conditions. In this analysis a ratio of greater than 1.0 indicates increased cell death relative to control, whereas a ratio of less than 1.0 indicates decreased cell death relative to control. (C) Cell death (percentage of cells staining with propidium iodide [PI] per 2000 analyzed cells) was similar in Z− and Z+ cells 24 hours after transfection of p53W expression plasmid, but significantly greater (P < .05 indicated) 48 hours after transfection in Z+ relative to Z− cells. During this interval, normalized p53-dependent transcription of pG13PYluc (inset) increased significantly in Z+ cells (*P < .05 for normalized luciferase activity levels 18 and 36 hours after transfection of p53W and reporter genes into Z+ cells).

Close modal

Similar numbers of dying cells were present in populations of Z− and Z+ cells transfected with p53W expression plasmid after 24 hours of incubation (Figure 5C), as determined by staining with propidium iodide. Significantly more dying cells were evident 48 hours after transfection with p53 W in Z+ cells than in Z− cells. Thus increased cell death in Z+ cells transfected with p53W expression plasmid correlated with the time course of p53-dependent transcription in Z+ cells, which increased several times between 18 and 36 hours after transfection of p53 (Figure 5C, inset graph). These experiments demonstrated increased apoptotic cell death in populations of Jurkat cells stably expressing ZEBRA protein transfected with transcriptionally active p53 expression plasmid.

RNA transcripts encoding ZEBRA are expressed in EBV-infected T cells,42,43 but ZEBRA protein expression levels are low.43 A heterogeneous population of EBV-infected thymocytes contains infected and uninfected cells.43Thymocytes from normal patients obtained during surgery43also have endogenous p53 that may be in variable states of activation, depending on variable conditions such as the health of the tissue donor. In contrast, Jurkat cells stably expressing ZEBRA protein are a homogeneous population of cells lacking significant endogenous p53-dependent transcriptional activity. Therefore, we characterized p53-dependent transcription in Jurkat cells stably expressing ZEBRA protein (Figure 1) to test the hypothesis that ZEBRA expression modulates p53-dependent transcription in T cells. Remarkably, ZEBRA protein detected in these cells using several ZEBRA-specific antibodies (mouse monoclonal antibody, human and rabbit polyclonal antisera) was localized to the cell cytoplasm, as determined by both immunofluorescence (Figure 1C) and protein fractionation (Figure 1D). These small amounts of cytoplasmic ZEBRA protein were associated with an altered cellular morphology (Figure 1E).

Expression of ZEBRA in Jurkat T-lymphoblastoid cells was associated with increased nuclear levels of p53 protein detected by Western blot analysis (Figure 2) and increased p53-dependent transcriptional activity detected by luciferase reporter genes (Figures 3, 4). In T cells, nuclear p53 protein levels detected by Western blotting (Figure2) correlated with transcriptional activation of p53 in Jurkat cells (Figures 3, 4). Activation of p53-dependent transcription associated with ZEBRA expression required functional p53 DNA binding sites in the reporter gene. Deletion of the p53 carboxyl-terminus that regulates the physiologic activation of p53 was associated with decreased p53-dependent transcriptional activation (Figure 3). Increased transcription of p53 reporter genes was not evident in Jurkat cells transiently transfected with ZEBRA expression plasmid (Figure 3C). Activation of the p21 promoter, a physiologic target of p53, was also associated with the expression of ZEBRA protein in T cells (Figure 4). Transcriptional activation of the p21 promoter associated with ZEBRA expression did not require activation of p53 by physiologic signals, such as DNA damage or other cellular injury, but exhibited synergy with p53 activation by ultraviolet irradiation.

Collectively, these observations suggest that prolonged or high levels of ZEBRA protein expression, but not transient ZEBRA expression, activate p53-dependent transcription in Jurkat cells. A similar activation of p53-dependent transcription was noted previously29 in epithelial cell lines overexpressing ZEBRA protein from an inducible promoter in the absence of other viral gene products. The transient expression of p53 in Z+ Jurkat cells was associated with the increased apoptosis of Jurkat cells (Figure 5), consistent with the increased transcription of p53 response elements in reporter genes. Although increased p21 promoter expression (Figure 4) may contribute to this phenotype, delineation of the mechanism through which p53 mediates increased apoptosis in Z+ cells was not determined in this work. Because ZEBRA expression activates p53-dependent transcription in epithelial cells29 and, as described in this work, in Jurkat T lymphocytes because ZEBRA expression inactivates p53-dependent transcription in B-lymphoblastoid cells such as the Akata cell line,28 these discordant results require further investigation.

Because ZEBRA is a nuclear transcription factor in B lymphocytes,32 the cytoplasmic localization of ZEBRA and its corresponding transcriptional inactivity in Jurkat cells is novel. Possibly, mutant non-nuclear alleles of ZEBRA could be selected during the generation of stable cell lines because of the toxic nature of the protein. However, the remarkable similarity between our results and previously reported results in epithelial cells in which the inducible transient expression of ZEBRA also activated p53-dependent transcription30 suggests that the activation of p53-dependent transcription by ZEBRA is cell-type specific rather than related to stable or transient expression of the protein. In addition, using a variety of antibodies (Figure 1), we only detected a single species of ZEBRA protein in Jurkat cells, eliminating the possibility that a truncated or deleted protein was expressed.

Therefore, we propose that the cytoplasmic localization of the ZEBRA protein in some cell lineages, for example T lymphocytes, results from a fundamental difference in post-translational processing of the ZEBRA protein in T cells that could, in turn, account for the effects of the expressed protein on, for example, p53-dependent transcription (Figure6). As an example, a difference in post-translational phosphorylation53 could result in the altered cellular localization of ZEBRA protein. Thus, the cellular lineage and localization of ZEBRA protein, rather than the expression of ZEBRA itself, may be critical in determining the effects of ZEBRA on p53-dependent transcription (Figure 6). These results were also consistent with our recent studies of the effects of ZEBRA protein on both transient and stable expression of the NF-κB transcription system54 in which ZEBRA expression blocked the cytoplasmic-to-nuclear translocation of components of NF-κB, apparently through a cytoplasmic interaction between ZEBRA and components of NF-κB.

Fig. 6.

Model of the discordant effects of ZEBRA protein on p53-dependent transcription in different cell types.

In B lymphocytes in which ZEBRA is demonstrated to localize to the cell nucleus, binding between p53 and ZEBRA in the nucleus reduces transcriptional activity of both proteins through a direct protein–protein interaction. In T lymphocytes, interactions between ZEBRA and components of the cell cytoplasm or cytoskeleton (“cell stress”) activate cytoplasmic p53 activating kinases, such as ATM, independently of direct binding between ZEBRA and p53. “Cell stress” mediated by cytoplasmic ZEBRA may also contribute to the abnormal morphology of Jurkat cells stably expressing ZEBRA protein. An important prediction of this model is that relocation of ZEBRA to the cytoplasm by either post-translational modification or other mechanisms may reverse the effects of ZEBRA on p53-dependent transcription and cellular apoptosis in EBV-infected cells.

Fig. 6.

Model of the discordant effects of ZEBRA protein on p53-dependent transcription in different cell types.

In B lymphocytes in which ZEBRA is demonstrated to localize to the cell nucleus, binding between p53 and ZEBRA in the nucleus reduces transcriptional activity of both proteins through a direct protein–protein interaction. In T lymphocytes, interactions between ZEBRA and components of the cell cytoplasm or cytoskeleton (“cell stress”) activate cytoplasmic p53 activating kinases, such as ATM, independently of direct binding between ZEBRA and p53. “Cell stress” mediated by cytoplasmic ZEBRA may also contribute to the abnormal morphology of Jurkat cells stably expressing ZEBRA protein. An important prediction of this model is that relocation of ZEBRA to the cytoplasm by either post-translational modification or other mechanisms may reverse the effects of ZEBRA on p53-dependent transcription and cellular apoptosis in EBV-infected cells.

Close modal

We found in the present work that transiently transfected ZEBRA and p53 resulted in a trend toward increased p53-dependent transcription in most experiments in Jurkat cells (Figure 3C), though levels of ZEBRA expression in transiently transfected cells were below the level of detection by Western blot analysis and in situ studies. Because the ZEBRA expression plasmid pSV2-neo-Wzhet used in this work uses a native EBV promoter32 driven by the BamHI W promoter that we have demonstrated is poorly expressed in T cells,43,44it is likely that low levels of expression of the protein from this construct in Jurkat cells contributed to our ability to establish stable cell lines expressing an otherwise toxic protein product.54 In contrast, a significant decrease in p53-dependent transcription was confirmed with transient cotransfection of ZEBRA and p53 in B-lymphoblastoid cells, as reported previously.28 

Activation of p53-dependent transcription associated with cytoplasmic ZEBRA expression in Jurkat cells could be dependent or independent of binding between the 2 proteins.28 ZEBRA protein bound to p53 could displace factors such as mdm-2, which stimulates the degradation of p53,9 or could mask a p53 site targeting the protein for degradation by the ubiquitin pathway.48,49Alternatively, cytoplasmic ZEBRA could stabilize p53 indirectly the through activation of DNA-PK, ATM, or other kinases that phosphorylate the p53 amino terminus, thus interfering with the binding of mdm-2,9,11 or directly by displacing mdm-2 through a shared binding site. It would, therefore, be important to determine whether ZEBRA in part shares a p53-binding site with mdm-2 or interferes with mdm-2 binding and to what extent ZEBRA interacts directly or indirectly with other components of p53 activation, such as DNA-PK.

Because the ZEBRA protein binds at least in part to the carboxyl terminus of p53, as determined by in vitro binding studies,28 ZEBRA could, under some conditions, activate p53, which would be analogous to certain antibodies and small peptides binding to the p53 carboxyl terminus.10 This hypothesis is difficult to reconcile with the observations that deletion of the carboxyl terminus of p53 reduced, but did not eliminate, the activation of p53 detected by luciferase assay (Figure 3). If ZEBRA activates p53 directly by binding to the carboxyl terminus of p53, then deletion of the carboxyl regulatory terminus of p53 might be expected to eliminate p53 activation. We also did not find any evidence that purified ZEBRA protein could activate p53 DNA binding activity by a direct interaction with the carboxyl terminus of p53 using a gel-shift assay for p53-specific DNA binding55 and purified bacterially produced p5356 (data not shown).

As a result, we favor the hypothesis that ZEBRA expression in Jurkat cells indirectly activates cellular kinases or other factors that in turn stabilize and activate p53 partly or completely, independently of binding between the 2 proteins (Figure 6). For example, nonspecific toxic effects of ZEBRA on Jurkat cells could activate cytoplasmic kinases such as the ATM kinase11 or related components in DNA-PK leading to the stabilization and activation of p53. Toxic effects of cytoplasmic ZEBRA protein are also suggested by the altered appearance of Jurkat cells stably expressing ZEBRA (Figure 1E). Activated p53 would then be translocated to the nucleus, where it could interact with components of the p53-transcriptional pathway, including CBP/p300, a p53-binding regulatory protein57-59 possibly altered by ZEBRA expression.

EBV-encoded gene products expressed in T cells could contribute to altered T-cell signaling and apoptosis.44,54 Expression of EBV-encoded proteins such as ZEBRA could also alter the replication of HIV-1 in EBV-infected primary human T cells,45 in part through the altered activation of p53.21-25 Because p53 is expressed in primary T lymphocytes during normal cellular activation and proliferation but not in resting T cells,39 activated cells in particular could be targeted by ZEBRA expression because the ZEBRA promoter is activated through T-cell activation.60These observations may also be relevant to the pathogenesis of X-linked lymphoproliferative disease (XLP), an often fatal disorder of T-cell proliferation and malignancy in response to EBV infection.61 Because the defect in XLP resides in what is apparently a signaling protein related to the cytoplasmic SH2 adaptor family and this protein (denoted SH2D1A) is expressed primarily in human thymocytes, it is an interesting coincidence that these thymocytes are also the T-cell subset most readily infected by EBV with subsequent expression of ZEBRA and other EBV-encoded proteins.53,54 Possibly, interactions between cytoplasmic ZEBRA or other EBV-encoded proteins and SH2D1A could be altered in XLP, leading to the often fatal lymphoproliferation typical of this disease. Further studies in EBV-infected Jurkat cells should provide a useful model system for understanding the effects of EBV infection of T cells, particularly with regard to the role of this infection in altered p53-dependent transcription in the progression to T-cell lymphoma.40 

We thank Drs James F. Jones, Shannon Kenney, and Lucy Ghoda for helpful discussions. We also thank Dr Avi Kupfer and Hannah Kupfer for their assistance with immunofluorescence studies. We thank Diana Nabighian for her assistance in the preparation of the manuscript.

Supported by National Institutes of Health training grant T32-AI07365 (D.H.D.).

Reprints:Erwin W. Gelfand, Division of Basic Sciences, Department of Pediatrics, National Jewish Medical and Research Center, 1400 Jackson St, Denver, CO 80206.

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.

1
Ko
LJ
Prives
C
p53: puzzle and paradigm.
Genes Dev.
10
1996
1054
2
Levine
AJ
p53, the cellular gatekeeper for growth and division.
Cell.
88
1997
323
3
Hawley
RS
Friend
SH
Strange bedfellows in even stranger places: the role of ATM in meiotic cells, lymphocytes, tumors, and its functional links to p53.
Genes Dev.
10
1996
2383
4
el-Deiry
WS
Kern
SE
Pietenpol
JA
Kinzler
KW
Vogelstein
B
Definition of a consensus binding site for p53.
Nat Genet.
1
1992
45
5
Kern
SE
Pietenpol
JA
Thiagalingam
S
Seymor
A
Kinzler
KW
Vogelstein
B
Oncogenic forms of p53 inhibit p53-regulated gene expression.
Science.
256
1992
827
6
Pietenpol
JA
Tokino
T
Thiagalingam
S
el-Deiry
WS
Kinzler
KW
Vogelstein
B
Sequence-specific transcriptional activation is essential for growth suppression by p53.
Proc Natl Acad Sci U S A.
91
1994
1998
7
el-Deiry
WS
Tokino
T
Velculescu
VE
et al
WAF1, a potential mediator of p53 tumor suppression.
Cell.
75
1993
817
8
Macleod
KF
Sherry
N
Hannon
G
et al
p53-dependent and independent expression of p21 during cell growth, differentiation, and DNA damage.
Genes Dev.
9
1995
935
9
Shieh
SY
Ikeda
M
Taya
Y
Prives
C
DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2.
Cell.
91
1997
325
10
Hupp
TR
Sparks
A
Lane
DP
Small peptides activate the latent sequence-specific DNA binding function of p53.
Cell.
83
1995
237
11
Shiloh
Y
Ataxia–telangectasia and the nijmegen breakage syndrome: related disorders but genes apart.
Annu Rev Genet.
31
1997
635
12
Harris
CC
Hollstein
M
Clinical implications of the p53 tumor-suppressor gene.
N Engl J Med.
329
1993
1318
13
Teodoro
JG
Branton
PE
Regulation of apoptosis by viral gene products.
J Virol.
71
1997
1739
14
Kieff
E
Shenk
T
Modulation of apoptosis by herpes viruses.
Semin Virol.
8
1998
471
15
Sarnow
P
Ho
YS
Williams
J
Levine
AJ
Adenovirus E1b-58 kd tumor antigen and SV40 large tumor antigen are physically associated with the same 54 kd cellular protein in transformed cells.
Cell.
28
1982
387
16
Martin
MED
Berk
AJ
Adenovirus E1B 55K represses p53 activation in vitro.
J Virol.
72
1998
3146
17
Werness
BA
Levine
AJ
Howley
PM
Association of human papillomavirus types 16 and 18 E6 proteins with p53.
Science.
248
1990
76
18
Jones
DL
Thompson
DA
Munger
K
Destabilization of the RB tumor suppressor protein and stabilization of p53 contribute to HPV type 16 E7-induced apoptosis.
Virology.
239
1997
97
19
Daniels
PR
Sanders
CM
Maitland
NJ
Characterization of the interactions of human papillomavirus type 16 E6 with p53 and E6-associated protein in insect and human cells.
J Gen Virol.
79
1998
489
20
Pise-Masison
CA
Choi
KS
Radonovich
M
Dittmer
J
Kim
SJ
Brady
JN
Inhibition of p53 transactivation function by the human T-cell lymphotropic virus type 1 tax protein.
J Virol.
72
1998
1165
21
Duan
L
Ozaki
I
Oakes
JW
Taylor
JP
Khalili
K
Pomeranz
RJ
The tumor suppressor p53 strongly alters human immunodeficiency virus type 1 replication.
J Virol.
68
1994
4302
22
Subler
MA
Martin
DW
Deb
S
Activation of the human immunodeficiency virus type 1 long terminal repeat by transforming mutants of human p53.
J Virol.
68
1994
103
23
Greenway
A
Azad
A
McPhee
D
Human immunodeficiency virus type 1 Nef protein inhibits activation pathways in peripheral blood mononuclear cells and T-cell lines.
J Virol.
69
1995
1842
24
Li
CJ
Wang
C
Friedman
DJ
Pardee
AB
Reciprocal modulations between p53 and Tat of human immunodeficiency virus type 1.
Proc Natl Acad Sci U S A
92
1995
5461
25
Longo
F
Marchetti
MA
Castagnolli
L
Battaglia
PA
Gigliani
F
A novel approach to protein-protein interaction: complex formation between the p53 tumor suppressor and the HIV TAT proteins.
Biochem Biophys Res Comm.
206
1995
326
26
Fortunato
EA
Spector
DH
p53 and RPA are sequestered in viral replication centers in the nuclei of cells infected with human cytomegalovirus.
J Virol.
72
1998
2033
27
Kashanchi
F
Araujo
J
Doniger
J
et al
Human herpes virus 6 (HHV-6) ORF-1 transactivating gene exhibits malignant transforming activity and its protein binds to p53.
Oncogene.
14
1997
359
28
Zhang
Q
Gutsch
D
Kenney
S
Functional and physical interaction between p53 and BZLF-1: implications for Epstein-Barr virus latency.
Mol Cell Biol.
14
1994
1929
29
Cayrol
C
Flemington
EK
The Epstein-Barr virus bZIP transcription factor Zta causes G0/G1 cell cycle arrest through induction of cyclin dependent kinase inhibitors.
EMBO J.
15
1996
2748
30
Chen
W
Cooper
NR
Epstein-Barr virus nuclear antigen 2 and latent membrane protein independently transactivate p53 through induction of NF-kB activity.
J Virol.
70
1996
4849
31
Farrell
PJ
Allan
GJ
Shanahan
F
Vousdan
KH
Crook
T
p53 is frequently mutated in Burkitt's lymphoma cell lines.
EMBO J.
10
1991
2879
32
Countryman
J
Jenson
H
Seibl
R
Wolf
H
Miller
G
Polymorphic proteins encoded within BZLF-1 of defective and standard Epstein-Barr viruses disrupt latency.
J Virol.
61
1987
3672
33
Farrell
PJ
Rowe
DT
Rooney
CM
Kouzarides
T
Epstein-Barr virus BZLF-1 trans-activator specifically binds to a consensus AP-1 site and is related to c-fos.
EMBO J.
8
1989
127
34
Flemington
E
Speck
SH
Epstein-Barr virus BZLF1 trans-activator induces the promoter of a cellular cognate gene, c-fos.
J Virol.
64
1990
4549
35
Gutsch
DE
Holley-Guthrie
EA
Zhang
Q
et al
The bZIP transactivator of Epstein-Barr virus, BZLF-1, functionally and physically interacts with the p65 subunit of NF-κB.
Mol Cell Biol.
14
1994
1939
36
Sista
ND
Barry
C
Sampson
K
Pagano
JS
Physical and functional interaction of the Epstein– Barr virus BZLF1 transactivator with the retinoic acid receptors RARa and RXRa.
Nucl Acids Res.
23
1995
1729
37
Lieberman
PM
Berk
AJ
The Zta trans-activator protein stabilizes TFIID association with promoter DNA by direct protein-protein interaction.
Genes Dev.
5
1991
2441
38
Kouzarides
T
Packham
C
Cook
A
Farrell
PJ
The BZLF-1 protein of EBV has a coiled coil dimerisation domain without a heptad leucine repeat but with homology to the C/EBP leucine zipper.
Oncogene.
6
1991
195
39
Terada
N
Lucas
JJ
Gelfand
EW
Differential regulation of the tumor suppressor molecules, retinoblastoma susceptibility gene product (Rb) and p53, during cell cycle progression of normal human T cells.
J Immunol.
147
1991
698
40
Jones
JF
Shurin
S
Abramowsky
C
et al
T-cell lymphomas containing Epstein-Barr viral DNA in patients with chronic Epstein-Barr virus infections.
N Engl J Med.
318
1988
733
41
Chen
C
Sadler
RH
Walling
DM
Su
I
Hsieh
H
Raab-Traub
N
Epstein-Barr virus (EBV) gene expression in EBV-positive peripheral T-cell lymphomas.
J Virol.
67
1993
6303
42
Kanegane
H
Bhatia
K
Gutierrez
M
et al
A syndrome of peripheral blood T-cell infection with Epstein-Barr virus (EBV) followed by EBV-positive T-cell lymphoma.
Blood.
91
1998
2085
43
Kelleher
CA
Kaufman-Patterson
R
Dreyfus
DH
et al
Epstein-Barr virus replicative gene transcription during de novo infection of human thymocytes: simultaneous early expression of BZLF-1 and its repressor RAZ.
Virology.
208
1995
685
44
Dreyfus
DH
Kelleher
CA
Jones
JF
Gelfand
EW
Epstein-Barr virus infection of T cells: implicationsfor altered T-lymphocyte activation, repertoire development, and autoimmunity.
Immunol Rev.
152
1996
89
45
Guan
M
Zhang
RD
Wu
B
Henderson
EE
Infection of primary CD4+ and CD8+ T lymphocytes by Epstein-Barr virus enhances human immunodeficiency virus expression.
J Virol.
70
1996
7341
46
Katz
BZ
Berkman
AB
Shapiro
ED
Serologicevidence of active Epstein-Barr virus infection in Epstein-Barr virus-associated lymphoproliferative disorders of children with acquired immunodeficiency syndrome.
J Pediatr.
120
1992
228
47
Sinha
SK
Todd
SC
Hedrick
JA
Speiser
CL
Lambris
JD
Tsoukas
CD
Characterization of the EBV/C3d receptor on the human Jurkat T cell line.
J Immunol.
150
1993
5311
48
Li
X
Coffino
P
Identification of a region of p53 that confers lability.
J Biol Chem.
271
1996
4447
49
Ciechanover
A
Shkedy
D
Oren
M
Bercovich
B
Degradation of the tumor suppressor protein p53 by the ubiquitin-mediated proteolytic system requires a novel species of ubiquitin-carrier protein E2.
J Biol Chem.
269
1994
9582
50
Rickinson
AB
Kieff
E
Epstein-Barr virus. Fields Virology.
1996
2397
2446
Lippincott–Raven
Philadelphia
51
Schreiber
E
Matthias
P
Muller
MM
Schaffner
W
Rapid detection of octamer binding proteins with “mini-extracts”prepared from a small number of cells.
Nucl Acids Res.
17
1989
6419
52
Monks
CR
Kupfer
H
Tamir
I
Barlow
A
Kupfer
A
Selective modulation of protein kinase C during T-cell activation.
Nature.
385
1997
83
53
Daibata
M
Humphreys
RE
Sairenji
T
Phosphorylation of the Epstein-Barr virus immediate-early gene product BZLF-1.
Virology.
188
1992
916
54
Dreyfus
DH
Nagasawa
M
Pratt
JC
Kelleher
CA
Gelfand
EW
Inactivation of NF-κB by EBV BZLF-1 encoded ZEBRA protein in human T-cells.
J Immunol.
163
1999
6261
55
Furnari
FB
Zacny
V
Quinlivan
EB
Kenney
S
Pagano
JS
RAZ, an Epstein-Barr virus transdominant repressor that modulates the viral reactivation mechanism.
J Virol.
68
1994
1827
56
Selivanova
G
Iotsova
V
Okan
I
et al
Restoration of the growth suppression function of mutant p53 by a synthetic peptide derived from the p53 C-terminal domain.
Nat Med.
3
1997
632
57
Gu
W
Shi
XL
Roeder
RG
Synergistic activation of transcription by CBP and p53.
Nature.
387
1997
819
58
Lill
NL
Grossman
SR
Ginsberg
D
Decaprio
J
Livingston
DM
Binding and modulation of p53 by p300/CBP coactivators.
Nature.
387
1997
823
59
Avantaggiati
ML
Ogryzko
V
Gardner
K
Giordano
A
Levine
AS
Kelly
K
Recruitment of p300/CBP in p53-dependent signal pathways.
Cell.
89
1997
1175
60
Flamand
L
Menezes
J
Cyclic AMP-responsive element-dependent activation of Epstein-Barr virus Zebra promoter by human herpes virus 6.
J Virol.
70
1996
1784
61
Coffey
AJ
Brooksbank
RA
Brandau
O
et al
Host response to EBV infection in X-linked lymphoproliferative disease results from mutations in an SH2-domain encoding gene.
Nat Genet.
20
1999
129
Sign in via your Institution