48
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Sequence analysis of Epstein-Barr virus (EBV) early genes BARF1 and BHRF1 in NK/T cell lymphoma from Northern China

      research-article
      , , , , ,
      Virology Journal
      BioMed Central

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          NK/T cell lymphoma is an aggressive lymphoma almost always associated with EBV. BamHI-A rightward open reading frame 1 (BARF1) and BamHI-H rightward open reading frame 1 (BHRF1) are two EBV early genes, which may be involved in the oncogenicity of EBV. It has been found that V29A strains, a BARF1 mutant subtype, showed higher prevalence in NPC, which may suggest the association between this variation and nasopharyngeal carcinoma (NPC). To characterize the sequence variation patterns of the Epstein-Barr virus (EBV) early genes and to elucidate their association with NK/T cell lymphoma, we analyzed the sequences of BARF1 and BHRF1 in EBV-positive NK/T cell lymphoma samples from Northern China.

          Methods

          In situ hybridization (ISH) performed for EBV-encoded small RNA1 (EBER1) with specific digoxigenin-labeled probes was used to select the EBV positive lymphoma samples. Nested-polymerase chain reaction (nested-PCR) and DNA sequence analysis technique were used to obtain the sequences of BARF1 and BHRF1. The polymorphisms of these two genes were classified according to the signature changes and compared with the known corresponding EBV gene variation data.

          Results

          Two major subtypes of BARF1 gene, designated as B95-8 and V29A subtype, were identified. B95-8 subtype was the dominant subtype. The V29A subtype had one consistent amino acid change at amino acid residue 29 (V → A). Compared with B95-8, AA change at 88 (L → V) of BHRF1 was found in the majority of the isolates, and AA79 (V → L) mutation in a few isolates. Functional domains of BARF1 and BHRF1 were highly conserved. The distributions of BARF1 and BHRF1 subtypes had no significant differences among different EBV-associated malignancies and healthy donors.

          Conclusion

          The sequences of BARF1 and BHRF1 are highly conserved which may contribute to maintain the biological function of these two genes. There is no evidence that particular EBV substrains of BARF1 or BHRF1 is region-restricted or disease-specific.

          Related collections

          Most cited references29

          • Record: found
          • Abstract: found
          • Article: not found

          Clinical differences between nasal and extranasal natural killer/T-cell lymphoma: a study of 136 cases from the International Peripheral T-Cell Lymphoma Project.

          Among 1153 new adult cases of peripheral/T-cell lymphoma from 1990-2002 at 22 centers in 13 countries, 136 cases (11.8%) of extranodal natural killer (NK)/T-cell lymphoma were identified (nasal 68%, extranasal 26%, aggressive/unclassifiable 6%). The disease frequency was higher in Asian than in Western countries and in Continental Asia than in Japan. There were no differences in age, sex, ethnicity, or immunophenotypic profile between the nasal and extranasal cases, but the latter had more adverse clinical features. The median overall survival (OS) was better in nasal compared with the extranasal cases in early- (2.96 vs 0.36 years, P < .001) and late-stage disease (0.8 vs 0.28 years, P = .031). The addition of radiotherapy for early-stage nasal cases yielded survival benefit (P = .045). Among nasal cases, both the International Prognostic Index (P = .006) and Korean NK/T-cell Prognostic Index (P < .001) were prognostic. In addition, Ki67 proliferation greater than 50%, transformed tumor cells greater than 40%, elevated C-reactive protein level (CRP), anemia (< 11 g/dL) and thrombocytopenia (< 150 x 10(9)/L) predicts poorer OS for nasal disease. No histologic or clinical feature was predictive in extranasal disease. We conclude that the clinical features and treatment response of extranasal NK/T-cell lymphoma are different from of those of nasal lymphoma. However, the underlying features responsible for these differences remain to be defined.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The Epstein-Barr virus and the pathogenesis of lymphoma.

            Since the discovery in 1964 of the Epstein-Barr virus (EBV) in African Burkitt lymphoma, this virus has been associated with a remarkably diverse range of cancer types. Because EBV persists in the B cells of the asymptomatic host, it can easily be envisaged how it contributes to the development of B-cell lymphomas. However, EBV is also found in other cancers, including T-cell/natural killer cell lymphomas and several epithelial malignancies. Explaining the aetiological role of EBV is challenging, partly because the virus probably contributes differently to each tumour and partly because the available disease models cannot adequately recapitulate the subtle variations in the virus-host balance that exist between the different EBV-associated cancers. A further challenge is to identify the co-factors involved; because most persistently infected individuals will never develop an EBV-associated cancer, the virus cannot be working alone. This article will review what is known about the contribution of EBV to lymphoma development. Copyright © 2014 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Structural Basis for Apoptosis Inhibition by Epstein-Barr Virus BHRF1

              Introduction To combat invading viruses, altruistic suicide of the infected host cells may be initiated to rapidly and efficiently eliminate the pathogen [1], [2]. Often, this response is a critical component of host defences [1], [2]. Consequently, many viruses have co-evolved adaptive mechanisms to subvert apopt osis, thereby ensuring their own survival and propagation. Some viruses, such as Epstein-Barr virus (EBV), encode homologs of the mammalian pro-survival protein Bcl-2 [3], [4], [5]. EBV was first identified in association with Burkitt lymphoma and it is also linked to other lymphoid malignancies (Hodgkin's lymphoma, post-transplant lymphoproliferative disorders) and nasopharyngeal carcinoma [6]. Whereas increased expression of Bcl-2 promotes malignancies such as human follicular lymphoma [7], the precise role of the EBV encoded Bcl-2 homolog BHRF1 in EBV-associated malignancies is less well defined. However, more recent studies link BHRF1 to the transformation of primary B lymphocytes [8] and to lymphomagenesis [9]. Since overactivity of the oncogene myc is obligatory for Burkitt lymphoma [10], [11], [12], expression of BHRF1 may be necessary to block myc-induced apoptosis, akin to the striking synergy observed between Bcl-2 and myc during B cell transformation [13], [14]. Of note, the constitutive expression of BHRF1 permits lymphoblastoid immortalization by EBV and their prolonged survival [9], and together with expression of BHRF1 during normal B cells transformation [8] suggests a role for BHRF1 in post-transplant lymphoproliferative disease. Although confirmed BHRF1 expression has been shown in only a subset of Burkitt lymphomas [9], [15], it is plausible that BHRF1 plays a central role in the maintenance of this subset of Burkitt lymphomas as Bcl-2 overexpression is rare in this disease. As BHRF1 may be central for developing and maintaining certain EBV-associated lymphomas, we investigated if BHRF1 can modulate responses to therapy in experimental models. If so, BHRF1 represents an attractive drug target since normal cells may well be spared by its selective antagonism. Here, we show that BHRF1 potently confers chemoresistance, and importantly, it adversely impacts upon survival in a mouse model of Burkitt lymphoma. BHRF1 acts by sequestering a subset of the pro-apoptotic Bcl-2 family proteins; we show here the 3D structures of it in complex with BH3 domains of two, Bim and Bak, which may provide the basis for developing small molecule inhibitors of BHRF1 to improve the generally poor prognosis in EBV-associated malignancies. Results/Discussion BHRF1 counters apoptosis induced by multiple chemotherapeutic agents Using cultured cell lines, we tested the ability of BHRF1 to confer resistance against a range of apoptotic stimuli, especially those used for cancer chemotherapy. Stable expression of BHRF1 in FDC-P1 mouse myelomonocytic cells conferred resistance to etoposide or γ-irradiation (Fig. 1A) comparable to that observed in cells expressing similar levels of Bcl-2, Bcl-xL or Bcl-w (Fig. 1D). It also inhibited apoptosis induced by other cytotoxics including cytosine arabinoside (Ara-C), doxorubicin, etoposide and staurosporine in other cell lines (Figs. 1B, 1C, S1 and data not shown), comparable to that observed in cells expressing similar levels of Bcl-2 (Fig. 1E). Thus, BHRF1, like its mammalian counterparts, inhibits apoptosis induced in multiple cell types by diverse chemotherapeutic agents. 10.1371/journal.ppat.1001236.g001 Figure 1 BHRF1 protects cells from diverse apoptotic stimuli. (A) Viability of FDC-P1 cells stably overexpressing BHRF1, Bcl-2, Bcl-xL, Bcl-w or vector, treated with 0-10 µM etoposide (left) or 0–30 Gy γ-irradiation (right) and cultured for 48 h. (B) Eµ-myc pre-B-cell tumor cells were stably transfected with BHRF1, Bcl-2 or vector were exposed to 0–10 µM cytosine arabinoside (Ara-C) for 24 h. (C) MEFs overexpressing BHRF1, Bcl-2, Bcl-xL or vector were treated with 0–100 µM etoposide for 24 h. (D) Bcl-2, Bcl-xL, Bcl-w and BHRF1 are expressed at comparable levels in FDC-P1 cells. FLAG-tagged wild-type Bcl-2, Bcl-xL, Bcl-w and BHRF1 were stably expressed in FDC-P1 cells. Protein expression was evaluated using flow cytometry after staining fixed cells with an anti-FLAG antibody, followed by an anti-mouse FITC secondary antibody. Controls (dotted lines) indicate staining of cells expressing empty vector. (E) BHRF1 and Bcl-2 are expressed at comparable levels in Eµ-myc pre-B-cell tumor cells. Pre-B-cell tumor cells derived from Eµ-myc transgenic mice were stably transfected with FLAG-tagged BHRF1, Bcl-2 or an empty control vector. Protein expression was evaluated using flow cytometry after staining fixed cells with an anti-FLAG antibody, followed by an anti-mouse FITC secondary antibody. Controls (dotted lines) indicate staining of cells expressing empty vector. Cell viability was determined by PI exclusion; data shown are means ±1 SEM of 3 independent experiments except for the representative γ-irradiation experiment shown in (A). BHRF1 preserves mitochondrial function by inhibiting the activation of Bax and Bak To ascertain precisely how BHRF1 interferes with apoptosis signaling, we examined mitochondrial outer membrane permeabilization (MOMP) [16] in FDC-P1 cells after treatment with staurosporine. Whereas MOMP occurred rapidly in the parental cells, as indicated by the reduced uptake of the mitochondrial dye DiOC6(3), it was inhibited in cells expressing BHRF1 or Bcl-2 (Fig. 2A), implicating an anti-apoptotic effect upstream of mitochondrial damage. When the mediators of mitochondrial damage (Bax and Bak) were examined, we found that BHRF1 inhibited translocation of Bax from the cytosol (Fig. 2B) and activation of Bax and Bak was abrogated, as their conformational alteration associated with activation is blocked (Fig. 2C). Consistent with these observations, BHRF1 also inhibited the release of cytochrome c from within the mitochondria (Fig. 2B). 10.1371/journal.ppat.1001236.g002 Figure 2 BHRF1 inhibits loss of mitochondrial transmembrane potential and Bax/Bak activation. (A) FDC-P1 cells expressing BHRF1, Bcl-2 or a control vector were treated with 25 nM staurosporine for 24 h. Outer mitochondrial transmembrane potential (Δψm) was assessed by DiOC6(3) uptake. (B) HeLa cells treated with 200 J/m2 UV-irradiation were analyzed at 4 h for Bax translocation and cytochrome c release by immunoblotting after fractionation into soluble (s) and pellet (p) fractions. (C) Bax and Bak activation in UV-irradiated HeLa cells were assessed at 4 h using the conformation-specific mouse anti-Bak clone Ab-1 (Calbiochem) or mouse anti-Bax clone 3 antibodies [53]. We conclude that BHRF1 must exert its anti-apoptotic effect at the level of, or prior to, Bax and Bak activation, consistent with a report that BHRF1 prevented Bax and Bak conformational change, oligomerization and activation of the initiator caspase, caspase-9 [17]. How then might BHRF1 inhibit activation of these essential cell death mediators? BHRF1 interacts with a sub-set of pro-apoptotic Bcl-2 family proteins It is most likely that BHRF1 acts to abort cell death initiation by sequestering the endogenous pro-apoptotic mammalian Bcl-2 family members. Thus, we assessed the ability of a recombinant C-terminally truncated form of BHRF1 to directly bind peptides spanning the BH3 domains of pro-apoptotic Bcl-2 proteins using isothermal titration calorimetry (ITC). Binding was observed with peptides from the BH3-only proteins Bim (KD = 18 nM), Puma (70 nM) or Bid (110 nM) (Fig. 3A). No detectable binding was observed with peptides from other BH3-only proteins, or with Mule and Beclin-1, other proteins harboring a BH3 domain. We confirmed the interaction of intact BHRF1 in mammalian cells with selected full-length BH3-only proteins in co-immunoprecipitation assays (Fig. 3B). These results closely mirror those obtained in solution competition assays, using either fluorescence polarization [18] or surface plasmon resonance [19]. Thus, BHRF1 probably antagonizes a subset of the BH3-only proteins by direct sequestration. Interestingly, the ones targeted (Bid, Bim, Puma) are potent inducers of apoptosis, either because they neutralize most, if not all, the mammalian pro-survival proteins [20], [21] or because they can directly activate Bax or Bak [22], [23], [24]. 10.1371/journal.ppat.1001236.g003 Figure 3 BHRF1 binds a subset of pro-apoptotic Bcl-2 family proteins to counter apoptosis. (A) The affinity of recombinant BHRF1ΔC31 for BH3 domain peptides (26-mers, except for a Bax 28-mer and a Bid 34-mer) was assessed using isothermal titration calorimetry (ITC). Kd values (in nM) are the means of 3 experiments ± SD. NB: no binding detected. (B) Lysates prepared from 35S-labeled HEK-293T cells co-expressing FLAG-tagged BHRF1 and EE-tagged BH3-only proteins were immunoprecipitated with mouse monoclonal antibodies recognizing the FLAG (FL), EE, or an irrelevant control (C) tag. **Endogenous 14-3-3 associating with Bad [56]. (C) Yeast co-transformed with constructs encoding Bax or Bak and the indicated pro-survival proteins, each under the control of an inducible (GAL) promoter, were spotted onto inducing galactose (“ON”) or repressing glucose (“OFF”) plates as 5-fold serial dilutions. Images are representative of 2 independent experiments. We also investigated if BHRF1, in a manner similar to some mammalian [21], [25] and viral Bcl-2 proteins [26], [27], might directly bind Bax and Bak, the downstream mediators of mitochondrial damage. Recombinant BHRF1 bound a 26-mer Bak BH3 peptide (KD = 150 nM), but only weakly (>1 µM) to Bax BH3 (Fig. 3A). Consistent with the binding data, we observed that BHRF1 could directly counter Bak (Fig. 3C), but not Bax, when these proteins were expressed in yeast [28]. This heterologous model system is suited for studying the functional interactions by circumventing potential complications due to the presence of endogenous Bcl-2 family proteins in mammalian cells and avoids the use of detergents that may artificially induce or disrupt interactions between Bcl-2 family proteins [29]. Our assay is based on the observation that overexpression of Bax or Bak in yeast is lethal, even though yeasts do not express Bcl-2 family members and do not undergo apoptosis. Nonetheless, co-expression of Bcl-2, Bcl-xL, Mcl-1, or A1 with Bax and Bak can suppress death induced in yeast [30], thus reconstituting key aspects of the mammalian apoptotic machinery. Our observation that BHRF1 could counter Bak (Fig. 3C), but not Bax-induced yeast death when these proteins were expressed in yeast suggests that BHRF1 is only able to directly neutralize Bak, but not Bax. Taken together with the previous reports that BHRF1 interacts with the full-length Bak, but not with Bax [17], [31], [32], we conclude that BHRF1 can keep Bak inactive by direct binding (Fig. 3A, 3C, [19]), but must inhibit Bax indirectly, presumably by its ability to sequester BH3-only proteins such as Bim (Fig. 3A, 3B, [31]). It will therefore be interesting to investigate which pro-apoptotic protein is the critical target for BHRF1 in diverse cell types, especially those targeted by EBV during oncogenesis. It is noteworthy that in some lymphoid cells, the pro-survival action of BHRF1 tracked with its ability to bind Bim [31], which is critical for apoptosis induced by multiple stimuli in this cell type [33] and plays a role in suppressing myc-driven lymphomagenesis [34]. Structural basis for the engagement of BH3 domains by BHRF1 Our data (Fig. 3) and previously published studies [18] suggest that BHRF1, like its mammalian counterparts, inhibits cell death by sequestering endogenous pro-apoptotic Bcl-2 proteins. However, the structural basis for this is unclear since a previously published structure of C-terminally truncated BHRF1 lacked the characteristic hydrophobic surface groove responsible for interaction with the BH3 domains [35]. We have therefore determined crystal structures of BHRF1 in complex with its key targets, the BH3 domains of Bim (Fig. 4A–B, Table 1) and Bak (Fig. 4C, Table 1). 10.1371/journal.ppat.1001236.g004 Figure 4 BH3 peptides bind BHRF1 in a canonical binding groove. (A–B) BHRF1 (blue) in complex with the Bim BH3 domain (yellow). BHRF1 helices are labeled α1, α1′, α2-8. The view in (A) is into the hydrophobic binding groove formed by helices α3-5, while (B) is rotated by 180° through the vertical axis to illustrate helix α1′. (C) BHRF1 (blue) in complex with the Bak BH3 domain (orange). The view is as in (A). (D) Bcl-xL (cyan) in complex with the Bim BH3 domain [36]. The view is as in (A). (E–F) Unliganded BHRF1 ([35]; in green). The view in (E) is similar to (A), while (F) is comparable to (B). The binding groove is occluded by the near parallel arrangement of helices α3 and α4. (G) Detailed view of the BHRF1:Bim complex interface. The BHRF1 surface, backbone and binding groove are shown in grey, cyan and magenta respectively, while Bim BH3 is shown in yellow. The four key hydrophobic residues of Bim (I58, L62, I65 and F69; numbering based on human BimL) protruding into the binding groove, as well as the conserved salt-bridge formed by Bim D67 and BHRF1 R100 are labeled. (H) Detailed view of the BHRF1:Bak complex interface. Colour scheme is as for (G) with Bak BH3 shown in orange. The four key hydrophobic residues of Bak (V75, L78, I81 and I85) protruding into the BHRF1 binding groove, as well as the conserved salt-bridge formed by Bak D83 and BHRF1 R100 are labeled. 10.1371/journal.ppat.1001236.t001 Table 1 Crystallographic statistics. Crystal BHRF1:Bim BH3 BHRF1:Bim BH3 BHRF1:Bak BH3 Data collection and phasing Derivative MeHg Native Native Spacegroup P3221 P3221 P3221 Resolution range (Å) 50 - 2.7 50 – 1.5 50 - 2.05 Unique reflections 6110 33924 13668 Multiplicity a 7.2 (5.5) 9.6 (6.7) 10.0 (5.1) Completeness (%) a 99.1 (94.3) 98.8 (93.1) 99.5 (95.8) I/σI 40.7 (2.4) 33.9 (2.2) Rmerge a,b 0.088 (0.346) 0.043 (0.568) 0.064 (0.497) Rderiv c 0.212 Rcullis (centric/acentric) d 0.653/0.609 Phasing power (centric/acentric) e 1.46/1.39 Refinement Resolution range (Å) 50 – 1.5 20 - 2.05 Reflections (working set/test set) 32174/1705 12945/668 Protein atoms 1466 1437 Solvent atoms 116 H2O, 9 Br 73 H2O, 4 NO3 Rcryst/Rfree f 0.198/0.205 0.184/0.217 r.m.s.d. bonds (Å) 0.015 0.021 r.m.s.d. angles (°) 1.7 1.8 Ramachandran plot (%) g 95.0/5.0/0.0/0.0 97.4/1.9/0.6/0.0 a Numbers in parentheses refer to the highest resolution shells. b Rmerge  =  ∑h∑i | Ii(h) - |/∑h∑iIi(h), where I i (h) is the ith measurement of reflection h and is the weighted mean of all measurements of h. c Rderiv  =  ∑h||FPH | - |FP||/∑h|FP|, where FP and FPH are the native and derivative structure factors, respectively. d Rcullis  =  ∑h|||FPH| - |FP|| - |FH||/∑h||FPH| - |FP|, where FH is the calculated heavy atom structure factor. e Phasing power is defined as (r.m.s FH/r.m.s lack-of closure). f R  =  ∑h|Fobs - Fcalc|/∑hFobs, where Fobs and Fcalc are the observed and calculated structure factor amplitudes, respectively. Rcryst and Rfree were calculated using the working and test set, respectively. g Residues in most favoured, additionally allowed, generously allowed and disallowed regions. The Bim BH3 peptide binds into a surface groove formed by helices α2-5 of BHRF1 (Fig. 4A), in a similar manner to that previously observed for mammalian pro-survival Bcl-2 members such as Bcl-xL [36] or the unrelated viral Bcl-2 protein M11L [27]. Bak BH3 binds in an equivalent manner (Fig. 4C), and the two complexes superimpose with an RMSD of only 0.6 Å over the entire BHRF1 backbone indicating their similarities. As the characteristic hydrophobic surface groove was absent in unliganded BHRF1 due to the close proximity of helices α3 and α4 (Fig. 4E; [35]), significant structural changes are required in order to accommodate Bim or Bak BH3 domains. These changes affect mainly α4, which is at a 120° angle to α3 in the BH3-bound form (Fig. 4A), compared to the near anti-parallel alignment in the free form (Fig. 4E). Overall, the free and bound BHRF1 structures (comparing Fig. 4A with 4B) superimpose with an RMSD of 3.5 Å, with most differences found within the BH3 binding groove. This is reminiscent of the movement observed in Bcl-xL, which in the ligand-free state displays a narrow binding groove [37]. However, upon ligand binding, both α3 and 4 helices move to widen the hydrophobic groove and allow binding [36]. In contrast, the movement of α3 that enables groove opening in BHRF1 upon ligand binding is much less pronounced. The side-chain interactions contributing to the BHRF1-BH3 complexes are equivalent to those observed for mammalian pro-survival proteins such as Bcl-xL, with the four conserved hydrophobic BH3 residues of Bim (I58, L62, I65 and F69; numbering based on human BimL) protruding into pockets within the BHRF1 hydrophobic binding groove (Fig. 4G). Similarly, Bak residues V74, L78, I81 and I85 (numbering based on human Bak) interact with the BHRF1 binding groove (Fig. 4H). In addition to the hydrophobic interactions, BHRF1 R100 forms a salt bridge with Bim D67 or with Bak D83 (Fig. 4G, 4H). This electrostatic interaction is also observed in complexes of Mcl-1 and Bcl-xL with pro-apoptotic BH3 domains [36], [38] and even in a complex of Bcl-xL with a peptide foldamer [39]. The conservation of aspartic acid in the mammalian BH3 ligands suggests that this interaction is of particular importance for complex formation, and indeed a BHRF1 R100A mutation reduces Bim binding and abolishes interaction with Bak [31]. BHRF1 confers potent chemoresistance in vivo Since BHRF1 engages BH3 domains using a hydrophobic groove (Fig. 4) in a manner equivalent to that of its mammalian counterparts, conserving key interactions, we asked whether ABT-737, a BH3 mimetic compound known to inhibit Bcl-2, Bcl-xL, and Bcl-w [40], [41], could also target BHRF1. In the absence of pro-survival Mcl-1, ABT-737 is a potent cytotoxic agent. However, cells expressing BHRF1 were completely insensitive to ABT-737, even at the highest dose tested (10 µM) (Fig. S2), and survived long-term (Fig. 5A). Similarly, recombinant BHRF1 did not bind ABT-737 in biosensor assays (IC50>20 µM, data not shown). As ABT-737 is ineffective and as BHRF1 can potently confer chemoresistance when tested in cultured cell lines (Fig. 1), we evaluated the impact of its expression in a transgenic mouse model of Burkitt lymphoma [11]. In the Eµ-myc mouse, myc is overexpressed in the B cell and drives an aggressive B cell leukemia/lymphoma syndrome that is very similar to human Burkitt lymphoma. 10.1371/journal.ppat.1001236.g005 Figure 5 BHRF1 inhibits ABT-737 induced apoptosis and confers chemoresistance in a mouse model of Burkitt lymphoma. (A) Colony formation of mcl-1−/− MEFs overexpressing BHRF1, Bcl-xL or a control vector, 6 d after treatment with 0-10 µM ABT-737. (B, C) Kaplan-Meier survival plots of mice inoculated with Eµ-myc tumor cells overexpressing BHRF1, Bcl-2 (or an empty vector) and treated on d 4, 5 and 6 (arrows) afterwards with (B) saline or (C) 100 mg/kg cytosine arabinoside (Ara-C; given intra-peritoneally). N = 5 mice in each group. Expression of BHRF1 or Bcl-2 conferred chemoresistance (p = 0.014 and p = 0.049 respectively by log-rank test when compared to saline treated controls). However, there is no significant difference whether or not BHRF1 or Bcl-2 is expressed as a transgene (p = 0.85 by log-rank test). Malignant cells derived from sick Eµ-myc mice are readily transplantable into syngeneic wild-type recipients which succumb within four weeks to a disseminated disease if left untreated [42]. Treatment with Ara-C (Fig. 5C, black line), an agent used in the clinic for treating patients with Burkitt lymphoma, resulted in a sustained disease remission and survival with all mice alive and disease-free at 100 days with normal peripheral blood counts and spleen weights. In striking contrast, only a handful of treated mice that were inoculated with tumor cells overexpressing BHRF1 or Bcl-2 survive long-term (Fig. 5C). Comparable results were observed when two other cytotoxic agents, cyclophosphamide and etoposide, were used in similar efficacy studies (data not shown). Therefore BHRF1, like Bcl-2 [42], can potently confer chemoresistance in a mouse model of Burkitt lymphoma. Thus, it is highly likely that expression of BHRF1 will attenuate the response during treatment for EBV-driven malignancies. Concluding remarks In this study, we have confirmed that EBV BHRF1 exerts its pro-survival function by directly inhibiting a sub-set of pro-apoptotic Bcl-2 family proteins Bid, Bim, Puma and Bak, presumably ones most critical for the virus. Our three-dimensional structures show that these interactions closely resemble those seen with mammalian pro-survival proteins such as Bcl-xL. In light of the importance of BHRF1 in certain Burkitt lymphomas [9], and its detection in other EBV-associated malignancies including nasopharyngeal carcinoma [43] and B cell lymphomas [44], the development of therapeutic inhibitors of BHRF1 may be highly desirable. The inability of current small molecule inhibitors of mammalian pro-survival proteins such as ABT-737 to inhibit BHRF1 will require novel small molecule antagonists to be developed. The successful development of small molecule inhibitors of mammalian pro-survival Bcl-2 family proteins [40] suggests that similar approaches might be applied to the development of BHRF1 inhibitors. Materials and Methods Recombinant proteins and binding experiments BHRF1ΔC31 was cloned into pET DUET (Invitrogen) using BamHI and EcoRI, and expressed in E.coli BL21 DE3 pLysS. Cells were homogenized using an Avestin EmulsiFlex homogenizer in lysis buffer (50 mM Tris-HCl, pH 8.0, 150 mM NaCl, 10 mM BME). His-tagged BHRF1ΔC31 was purified using a nickel charged Hi-Trap chelating column (Amersham), eluted in lysis buffer with 250 mM imidazole and subjected to gel-filtration chromatography in 25 mM Hepes pH 7.5, 150 mM NaCl using a Superdex 200 column (GE Healthcare). Calorimetry data were collected on a VP-ITC (MicroCal) with BHRF1ΔC31 as previously described [45]. All peptides were purified by reverse-phase HPLC and dissolved as 2–5 mM stock solutions in water. The accession numbers for the peptides were: human BimL (AAC39594), human Puma (AAK39542), mouse Bmf (AAK38747), human Bad (NP_004313), human Bik (NP_001188), human Hrk (NP_003797), human Bid (NP_001187), human Noxa (NP_066950), human Bax (NP_620119), human Bak (NP_001179), mouse Mule (UniProt Accession code Q7TMY8; residues 1969–1994; PGGTTQEVGQLLQDMGDDVYQQYRSL) and mmBeclin (UniProt Accession code O88597, residue 103–128; DGGTMENLSRRLKVTGDLFDIMSGQT). Crystallization and structure determination BHRF1 complexes with Bim or Bak BH3 were obtained by mixing BHRF1 with human Bim or Bak 26-mer peptide in a 1∶1.25 molar ratio and concentrating using a centricon (Millipore) to 10 mg/mL. Crystals were grown in sitting drops at 20°C in 1.2 M NaBr, 50 mM malic acid pH 4.0 (BHRF1:Bim) or 1.6 M NaNO3, 50 mM malic acid pH 4.4 (BHRF1:Bak). The crystals belong to space group P3221 with a = b = 62.75 Å, c = 92.38 Å, α = β = 90°, γ = 120° (BHRF1:Bim) or a = b = 62.39 Å, c = 93.73 Å, α = β = 90°, γ = 120° (BHRF1:Bak). The asymmetric units contain 1 BHRF1:peptide complex. Diffraction data were collected from flash frozen crystals at 100 K at the Australian Synchrotron (beamline 3BM1) or at the Swiss Light Source (beamlines X06SA, X10SA) and processed with HKL2000. For the BHRF1:Bim complex, a heavy atom derivative was obtained by soaking crystals in mother liquor supplemented with 1 mM MeHgCl for 2 h. Hg sites were found and refined with Sharp. Clear and continuous electron density was obtained for residues 2–158 of BHRF1 and 51–72 of Bim. The final model was built with Coot [46], refined with Refmac5 [47] to a resolution of 1.5 Å and has a final R-factor of 0.198 (R-free 0.205). 95.0% of the residues are in the core regions of the Ramachandran plot, and no residues are in disallowed regions. The BHRF1:Bak structure was solved by molecular replacement with PHASER [48] using the BHRF1:Bim structure as a search model. The final model was built with Coot and refined with Refmac5 to a resolution of 2.05 Å and has a final R-factor of 0.184 (R-free 0.217). 97.4% of the residues are in the core regions of the Ramachandran plot, and no residues are in disallowed regions. All data collection and refinement statistics are summarized in Table 1. The coordinates have been deposited in the Protein Data Bank (accession codes 2v6q, 2wh6, 2xpx). Figures were prepared using PyMol (DeLano Scientific). Mouse tumor model Eµ-myc transgenic mice on an inbred C57BL/6 background with clinical evidence of lymphoma were culled by CO2 asphyxiation and lymphomatous tissue excised. Single-cell suspensions were obtained by manual sieving and stable Eµ-myc tumor cell cultures were established in FMA medium. Retroviral transduction using a pMSCV-IRES-GFP vector containing BHRF1 or Bcl-2 and sorting of GFP positive cells were conducted as previously described [11]. 1×106 viable tumor cells in 100 µL PBS were injected intraperitoneally into syngeneic C57BL/6 mice. Ara-C or PBS was injected intraperitoneally in 100 µL total volume on days 4, 5 and 6. Mice were culled when sick (hindleg paralysis, tremor, lethargic, tumor nodule >1 cm diameter, >5% weight loss) and leukemia/lymphoma confirmed by the presence of peripheral blood leucocytosis and enlarged spleen and/or lymph nodes. Survival of cohorts of 5 mice was compared by log-rank test and Kaplan-Meier analysis using GraphPad Prism statistical software. All experiments were approved by an institutional ethics committee. Cell lines and tissue culture MEF, HEK-293T and Phoenix packaging 293 cells were cultured in Dulbecco's Modified Eagles medium (DMEM) supplemented with 10% FCS. FDC-P1 cells were additionally supplemented with mouse IL-3 (1000 U/mL). Eµ-myc tumor cells were harvested from a symptomatic Eµ-myc transgenic mouse and cultured in FMA, a high glucose version of DMEM supplemented with 10% FCS, 50 µM 2-mercaptoethanol and 250 µM asparagine. Mammalian expression constructs Epitope-tagged mammalian expression vectors for human Bcl-2 family proteins have been described previously [20], [49], [50], [51], [52]. All constructs were verified by sequencing. Details and constructs are available from the authors. Retrovirus production and transduction To produce retroviral supernatant, 2.5×106 ecotropic Phoenix packaging cells were seeded overnight in 10 cm tissue culture plates. Media was replaced with 5 ml serum-free DMEM containing 5 µg MSCV-based retroviral plasmid with 15 µL Lipofectamine (Invitrogen). After 24 h media was replaced with medium supplemented with 20% FCS and incubated for a further 24 h at 32°C. Viral supernatant was cleared of cell debris by centrifugation for 5 min at 1500 rpm. 500 µL filtered virus (0.45 µm, Millipore) was spin-infected onto target cells in a total volume of 1 mL media containing 4 µg/mL polybrene (Sigma) in 24 well plates at 32°C with 2500 rpm radial centrifugation for 45 min. Infection efficiency of MEFs were generally >90% and 20–30% for FDC-P1 cells. Cell survival assays and cytotoxic drugs Cell death was induced by 0–100 µM etoposide (Pharmacia-Upjohn), 0–30 Gy γ-irradiation, 0–10 µM Ara C (Pharmacia-Upjohn), 0–100 nM staurosprorine (Sigma-Aldrich) or 0–10 µM ABT-737 (Abbott Laboratories). Cell viability was quantified by flow cytometric analysis of cells excluding 5 µg/mL propidium iodide (PI) (Sigma-Aldrich) using a FACScan (Becton Dickinson). Each time point was performed at least three times. For long-term colony assays using MEFs, cells were infected with GFP-expressing retroviral constructs, then treated with qVD.OPH (Enzyme Systems) to prevent cell death. After culture for 24 h, 200 GFP+ve cells were sorted into 6-well plates. Colonies were stained and counted 6 d later. Immunofluorescence Cells expressing mammalian FLAG-tagged pro-survival Bcl-2 proteins, BHRF1 or empty vector were washed in PBS, fixed in 1% paraformaldehyde/PBS (10 min, 4°C) and washed twice in KDS-BSS. Cells were incubated with 1∶1,000 primary anti-FLAG M2 (Sigma) antibody for 20 minutes, washed in KDS-BSS/0.02% saponin and then incubated with 1∶100 goat anti-mouse FITC or PE antibody (Southern Biotechnology) for 20 minutes before analysis on a FACScan (BD) using Cell Quest software (BD). Cytofluorometric determination of mitochondrial transmembrane potential and Bax/Bak activation To assess mitochondrial transmembrane potential (Δψm), cells were incubated for 15 min at 37°C in buffer containing 40 nM 3,3′- dihexyloxacarbocyanine iodide (DiOC6[3]; Molecular Probes) before adding 10 µg/mL of PI. The cells were kept on ice until flow cytometric analysis. To assess the activation of Bax and Bak, HeLa cells were left untreated or pretreated with a proteasome inhibitor (10 µM MG-132; Calbiochem) or a wide-spectrum caspase inhibitor (100 µM zVAD.fmk; Bachem) for 1 h before treatment with 200 J/m2 UV-irradiation. Following UV irradiation, cells were fixed with 1% paraformaldehyde (5 min at room temperature) and then washed with buffer supplemented with 2% fetal bovine serum. Fixed cells were then incubated with the primary antibodies: 2 µg/mL anti-Bak Ab-1 (Calbiochem) or 5 µg/mL anti-Bax clone 3 (BD) diluted in FACS buffer supplemented with 0.3% saponin for 30 min on ice. Cells were then washed, before incubation with the appropriate secondary antibody, either FITC-conjugated goat-anti-mouse IgG (10 µg/ml; SouthernBiotech) to detect Bax activation or a biotin-conjugated anti-mouse (diluted 1∶200; SouthernBiotech) followed by Streptavidin-conjugated PE (diluted 1∶300; Caltag) to detect Bak activation. The samples were analyzed using a FACScan (BD). Subcellular fractionation Fractionation of whole cell lysates into the soluble and pellet fractions has been previously described [54]. In brief, cells lysed in HMKEE buffer (20 mM Hepes, pH 7.2, 5 mM MgCl2, 10 mM KCl, 1 mM EDTA, 1 mM EGTA, and protease inhibitors) containing 250 mM sucrose and 0.025% digitonin (Calbiochem) were left on ice for 10 min, and then the organelles, cytoskeleton, and membranes were pelleted by centrifugation (13,000 rpm, 5 min at 4°C). The pellet was solubilized in RIPA buffer (150 mM NaCl, 1% Triton X-100, 0.5% deoxycholic acid, 0.1% SDS, 50 mM Tris-HCl, pH 8.0, and protease inhibitors). The protease inhibitors used include Pefabloc SC, soybean trypsin inhibitor, leupeptin, aprotinin, E64, and pepstatin (Sigma-Aldrich or Roche). Transient transfection, immunoprecipitation and immunoblotting The transfection and metabolic labeling of HEK-293T cells with 35S-methionine/cysteine (NEN) as well as co-immunoprecipitation have been described [49], [50], [52]. Briefly, equivalent TCA-precipitable lysates were immunoprecipiated using the mouse monoclonal antibodies to FLAG (M2; Sigma), Glu-Glu (CRP) and control HA (HA.11; CRP) tags. The proteins were resolved by SDS:PAGE, transferred onto nitrocellulose membranes and detected by autoradiography after 20 h at −80°C. Immunoblotting was performed using mouse monoclonal antibodies to Bax (5B7; Sigma-Aldrich) cytochrome c (7H8.2C12; BD Pharmingen) and detected using HRP-conjugated secondary antibodies (Southern Biotechnology) revealed by enhanced chemiluminescence (ECL; Amersham Biosciences). Yeast colony assays Yeast expression vectors were made by subcloning the cDNAs for full-length human Bcl-xL and BHRF1, or human Bax and human Bak, respectively, into the pGALL(TRP1) and pGALS(LEU2) vectors [55]. Saccharomyces cerevisiae W303a cells were co-transformed with indicated plasmids and grown under selection. For the survival assays, the cells were spotted as 5-fold serial dilutions onto glucose (repressing, “OFF”) or galactose (inducing, “ON”) plates as previously described [28]. Plates were incubated for 48 h at 30°C and then photographed. Ethics statement This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Health and Medical Research Council. The protocol was approved by the Committee on the Ethics of Animal Experiments of the Walter and Eliza Hall Institute of Medical Research (Permit Number: NKOT_07_008). All surgery was performed under sodium pentobarbital anesthesia, and all efforts were made to minimize suffering. Supporting Information Figure S1 Pre-B-cell tumor cells derived from Eµ-myc transgenic mice were stably transfected with BHRF1, Bcl-2 or a control vector and exposed to etoposide (0–10 µM). Viability was assessed by PI staining after 24 h. (0.17 MB TIF) Click here for additional data file. Figure S2 BHRF1 is not inhibited by ABT-737. Mcl-1 deficient MEF stably expressing BHRF1, Bcl xL or a control vector were treated with ABT-737 (0–10 µM). Viability was assessed 8 h later by flow cytometry after propidium iodide staining. (0.13 MB TIF) Click here for additional data file.
                Bookmark

                Author and article information

                Contributors
                sunlingling761216@163.com
                chekui1988@163.com
                qdzzz1987@163.com
                liusongrr@163.com
                edithxing@hotmail.com
                86 0532 83812423 , qdluobing@163.com
                Journal
                Virol J
                Virol. J
                Virology Journal
                BioMed Central (London )
                1743-422X
                4 September 2015
                4 September 2015
                2015
                : 12
                : 135
                Affiliations
                [ ]Department of Pathology, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003 China
                [ ]Department of Medical Microbiology, Qingdao University Medical College, 38 Dengzhou Road, Qingdao, 266021 China
                [ ]Department of Clinical Laboratory, Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266555 China
                Article
                368
                10.1186/s12985-015-0368-3
                4558833
                26337172
                30e9062c-365f-4cb1-bdac-c8b7a51523db
                © Sun et al. 2015

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 10 March 2015
                : 27 August 2015
                Categories
                Research
                Custom metadata
                © The Author(s) 2015

                Microbiology & Virology
                Microbiology & Virology

                Comments

                Comment on this article