201
|
Kuo TT, Shih LY, Tsang NM. Nasal NK/T cell lymphoma in Taiwan: a clinicopathologic study of 22 cases, with analysis of histologic subtypes, Epstein-Barr virus LMP-1 gene association, and treatment modalities. Int J Surg Pathol 2005; 12:375-87. [PMID: 15494863 DOI: 10.1177/106689690401200410] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nasal NK/T cell lymphoma is a distinctive type of extranodal lymphoma with an unique immunophenotype and a strong association with Epstein-Barr virus (EBV). It is one of the common extranodal lymphomas in Taiwan. We studied 22 cases of nasal NK/T cell lymphoma to characterize their clinicopathologic features and to explore the possible differences between histologic subtypes and their clinical behavior as well as the prevalence of 30-base pair (bp) deleted latent membrane protein-1 (LMP-1) gene of the EBV. They consisted of 5 cases of small cell type (SC), 6 cases of medium-sized cell type (MC), 6 cases of large cell type (LC), and 5 cases of pleomorphic cell type (PC). Twelve patients were men and 10 were women (1.2 to 1), and their ages ranged from 34 to 75 years with a median age of 55.5 years. The median ages of the LC type and PC type were older than the other 2 types. No other clinical features differed significantly among the 4 subtypes. Nasal obstruction was the most common initial presenting symptom. All but 1 case had stage IE disease at the time of diagnosis. Five cases developed extranasal involvement and skin was the most common site. No bone marrow involvement was detected. The majority of patients received local radiotherapy and chemotherapy. Local irradiation was more effective than chemotherapy alone. We achieved an overall survival of 63.6% at 5 years as estimated by the Kaplan-Meier analysis, which was better than other series. All cases displayed an immunophenotypic profile of CD3(epsilon)+, CD20-, CD56+, and TIA-1+ except that 1 case was CD3(epsilon)-. Fourteen of 22 cases (64%) expressed LMP-1. Nine cases of various cell types (41%) were also CD30+. Among the 4 histologic subtypes, the SC type differed from the other types by the absence of angiodestruction and necrosis, although angioinvasive growth was seen in 2 of them. Pseudoepitheliomatous hyperplasia was seen in only 3 cases of the SC type, and all 5 cases of the SC type were CD30-. No statistical difference in survival was found among the 4 histologic subtypes or between CD30+ and CD30- cases. All 22 cases were positive for EBV by polymerase chain reaction and Epstein-Barr virus early RNA (EBER) in-situ hybridization. A high prevalence rate of 86% (19/22) of the 30-base pair (bp) deleted LMP-1 gene was found, but 81.5% (22/27) of the EBV-positive control reactive lymphoid tissues also had the 30-bp deleted LMP-1 gene. Therefore, the high prevalence of the 30-bp deleted LMP-1 gene found in NK/T cell lymphoma could be due to the high prevalence of the deleted variant in this geographic region. However, it remains possible that the high prevalence of the deleted LMP-1 gene contributed to the increased incidence of EBV-associated nasal NK/T cell lymphoma in Taiwan.
Collapse
MESH Headings
- Adult
- Aged
- Female
- Gene Deletion
- Herpesvirus 4, Human
- Humans
- In Situ Hybridization
- Killer Cells, Natural/pathology
- Killer Cells, Natural/virology
- Lymphoma, T-Cell, Peripheral/mortality
- Lymphoma, T-Cell, Peripheral/pathology
- Lymphoma, T-Cell, Peripheral/virology
- Male
- Middle Aged
- Nose Neoplasms/mortality
- Nose Neoplasms/pathology
- Nose Neoplasms/virology
- RNA, Viral/analysis
- Survival Rate
- Taiwan/epidemiology
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/isolation & purification
Collapse
Affiliation(s)
- Tseng-Tong Kuo
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University School of Medicine, Kwei San, Tao Yuan, Taiwan
| | | | | |
Collapse
|
202
|
May KF, Roychowdhury S, Bhatt D, Kocak E, Bai XF, Liu JQ, Ferketich AK, Martin EW, Caligiuri MA, Zheng P, Liu Y. Anti–human CTLA-4 monoclonal antibody promotes T-cell expansion and immunity in a hu-PBL-SCID model: a new method for preclinical screening of costimulatory monoclonal antibodies. Blood 2005; 105:1114-20. [PMID: 15486062 DOI: 10.1182/blood-2004-07-2561] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractWhen adopting basic principles learned in mice to clinical application in humans, it is often difficult to distinguish whether a “translation” fails because of an invalid target in the human disease or because the therapeutic agents are not optimal for the human target. It is, therefore, desirable to develop preclinical models to optimize therapies for human targets using in vivo settings. Although anti–mouse CTLA-4 antibodies are known to enhance immune responses in vivo, their effect on T-cell activation in vitro ranges from enhancement to inhibition. Here we use the hu-PBL-SCID mouse model of Epstein-Barr virus (EBV)–associated lymphoma development to screen a panel of anti–human CTLA-4 monoclonal antibodies (mAbs) for their effect on human lymphocytes in an in vivo “humanized” environment. We report significant heterogeneity of anti–human CTLA-4 mAbs in enhancing the expansion of human T cells in mice, and this heterogeneity cannot be attributed to immunoglobulin isotypes or affinity for CTLA-4. These data validate the development of additional screening tools, such as the one described, to further characterize functional activity of antihuman antibodies before proceeding with clinical translation to human studies.
Collapse
Affiliation(s)
- Kenneth F May
- Department of Pathology, Division of Cancer Immunology, The Ohio State University Medical Center and Comprehensive Cancer Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Sheu LF, Chen A, Lee HS, Hsu HY, Yu DS. Cooperative interactions among p53, bcl-2 and Epstein-Barr virus latent membrane protein 1 in nasopharyngeal carcinoma cells. Pathol Int 2004; 54:475-85. [PMID: 15189500 DOI: 10.1111/j.1440-1827.2004.01654.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Interactions among p53, bcl-2 and Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) in nasopharyngeal carcinoma (NPC) cells were evaluated by gene cotransfections. The data showed that bcl-2 expression was not only able to prevent the growth suppression induced by wild-type p53 but was also paradoxically able to inhibit the growth enhancement induced by mutant p53. Latent membrane protein 1 was shown to be capable of overcoming the growth inhibition induced by wild-type p53 and the synergistic cooperation with bcl-2 to enhance cellular growth. Latent membrane protein 1 could also cooperate with mutant p53 to provide a growth advantage for NPC cells. Most NPC revealed detectable overexpression of p53, and the majority of those were a wild type possibly responding to EBV infection. The coexpression of bcl-2 and LMP1 was thought to inhibit the growth suppression induced by wild-type p53 in NPC. But there was no associated expression between LMP1 and bcl-2 because we demonstrated that transfected LMP1 failed to induce bcl-2 expression in NPC cells in contrast to the findings in B cells. It is theorized that the cooperative expression of bcl-2 and LMP1 exists in the majority of NPC, while a minority of NPC have cooperative expression of LMP1 and mutant p53. Each cooperative interaction could play an important role in the development and progression of NPC.
Collapse
Affiliation(s)
- Lai-Fa Sheu
- Department of Pathology, Tri-Service General Hospital, National Defense Center, Taipei, Taiwan, Republic of China.
| | | | | | | | | |
Collapse
|
204
|
Chang Y, Lee HH, Chang SS, Hsu TY, Wang PW, Chang YS, Takada K, Tsai CH. Induction of Epstein-Barr virus latent membrane protein 1 by a lytic transactivator Rta. J Virol 2004; 78:13028-36. [PMID: 15542654 PMCID: PMC525024 DOI: 10.1128/jvi.78.23.13028-13036.2004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Latent membrane protein 1 (LMP1) of Epstein-Barr virus (EBV) is a transforming protein that affects multiple cell signaling pathways and contributes to EBV-associated oncogenesis. LMP1 can be expressed in some states of EBV latency, and significant induction of full-length LMP1 is also observed frequently during virus reactivation into the lytic cycle. It is still unknown how LMP1 expression is regulated during the lytic stage and whether any EBV lytic protein is involved in the induction of LMP1. In this study, we first identified that LMP1 expression is associated with the spontaneous virus reactivation in EBV-infected 293 cells and that its expression is a downstream event of the lytic cycle. We further found that LMP1 can be induced by ectopic expression of Rta, an EBV immediate-early lytic protein. The Rta-mediated LMP1 induction is independent of another immediate-early protein, Zta. Northern blotting and reverse transcription-PCR analysis revealed that Rta upregulates LMP1 at the RNA level. Reporter gene assays further demonstrated that Rta activates both the proximal and distal promoters of the LMP1 gene in EBV-negative cells. Both the amino and carboxyl termini of the Rta protein are required for the induction of LMP1. In addition, Rta transactivates LMP1 not only in epithelial cells but also in B-lymphoid cells. This study reveals a new mechanism to upregulate LMP1 expression, expanding the knowledge of LMP1 regulation in the EBV life cycle. Considering an equivalent case of Kaposi's sarcoma-associated herpesvirus, induction of a transforming membrane protein by a key lytic transactivator during virus reactivation is likely to be a conserved event for gammaherpesviruses.
Collapse
Affiliation(s)
- Yao Chang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Room 714, Number 1, Section 1, Jen-Ai Rd., Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
205
|
Panagopoulos D, Victoratos P, Alexiou M, Kollias G, Mosialos G. Comparative analysis of signal transduction by CD40 and the Epstein-Barr virus oncoprotein LMP1 in vivo. J Virol 2004; 78:13253-61. [PMID: 15542676 PMCID: PMC524978 DOI: 10.1128/jvi.78.23.13253-13261.2004] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
There is much evidence, based primarily on in vitro studies, indicating that the Epstein-Barr virus oncoprotein latent membrane protein 1 (LMP1) mimics an activated CD40 receptor. In order to investigate the extent of similarity between LMP1 and CD40 functions in vivo, we analyzed the cytoplasmic signaling properties of LMP1 and CD40 in B cells in a directly comparable manner. For this purpose, we generated transgenic mice expressing either LMP1 or a chimeric LMP1CD40 molecule, which constitutively activates the CD40 pathway, under the control of the CD19 promoter. LMP1 and LMP1CD40 were expressed at similar levels in a B-lymphocyte-specific manner. Similar to LMP1, LMP1CD40 suppressed germinal center (GC) formation and antibody production in response to thymus-dependent antigens, albeit to a greater extent than LMP1. Furthermore, the avidity of the antibodies produced against thymus-dependent antigens was lower for LMP1CD40 transgenic mice than for wild-type and LMP1 transgenic mice. GC suppression was linked to the ability of LMP1CD40 and LMP1 to downregulate mRNA and protein levels of BCL6 and to suppress the activity of the BCL6 promoter. In contrast to LMP1, LMP1CD40 caused an upregulation of CD69, CD80, and CD86 in B cells and a dramatic increase in serum immunoglobulin M. In addition, LMP1CD40 but not LMP1 transgenic mice had elevated numbers of marginal-zone B cells and increased populations of polymorphonuclear cells and/or neutrophils. Consistent with these findings, LMP1CD40 but not LMP1 transgenic mice showed signs of spontaneous inflammatory reactions and the potential for autoimmunity.
Collapse
Affiliation(s)
- Dimitris Panagopoulos
- Institute of Immunology, Biomedical Sciences Research Center Al. Fleming, 34 Al. Fleming St., Vari 16672, Greece
| | | | | | | | | |
Collapse
|
206
|
Xie P, Bishop GA. Roles of TNF Receptor-Associated Factor 3 in Signaling to B Lymphocytes by Carboxyl-Terminal Activating Regions 1 and 2 of the EBV-Encoded Oncoprotein Latent Membrane Protein 1. THE JOURNAL OF IMMUNOLOGY 2004; 173:5546-55. [PMID: 15494504 DOI: 10.4049/jimmunol.173.9.5546] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
TNFR-associated factor (TRAF)3, an adaptor protein that binds the cytoplasmic domains of both CD40 and the EBV-encoded oncoprotein latent membrane protein (LMP)1, is required for positive signaling by LMP1 but not CD40 in B lymphocytes. The present study further investigated how TRAF3 participates in LMP1 signaling. We found that TRAF3 mediates signaling both through direct interactions with the C-terminal activating region (CTAR)1 of LMP1 and through indirect interactions with the CTAR2 region of LMP1 in mouse B cells. Notably, our results demonstrated that the CTAR2 region appears to inhibit the recruitment of TRAF1 and TRAF2 to membrane rafts by the CTAR1 region. Additionally, the absence of TRAF2 in B cells resulted in only a modest reduction in CTAR1-mediated signals and no detectable effect on CTAR2-mediated signals. CTAR1 and CTAR2 cooperated to achieve the robust signaling activity of LMP1 when recruited to the same membrane microdomains in B cells. Interestingly, TRAF3 deficiency completely abrogated the cooperation between CTAR1 and CTAR2, supporting the hypothesis that TRAF3 participates in the physical interaction between CTAR1 and CTAR2 of LMP1. Together, our findings highlight the central importance of TRAF3 in LMP1-mediated signaling, which is critical for EBV persistent infection and EBV-associated pathogenesis.
Collapse
Affiliation(s)
- Ping Xie
- Department of Microbiology, University of Iowa, Iowa City, IA 52242 , USA
| | | |
Collapse
|
207
|
Siegler G, Meyer B, Dawson C, Brachtel E, Lennerz J, Koch C, Kremmer E, Niedobitek E, Gonnella R, Pilch BZ, Young LS, Niedobitek G. Expression of tumor necrosis factor receptor-associated factor 1 in nasopharyngeal carcinoma: possible upregulation by Epstein-Barr virus latent membrane protein 1. Int J Cancer 2004; 112:265-72. [PMID: 15352039 DOI: 10.1002/ijc.20367] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
EBV infection is associated with virtually all cases of undifferentiated NPC, and the EBV-encoded LMP1 is expressed in a proportion of cases. LMP1 has transforming functions similar to members of the TNF receptor family and activates intracellular signaling cascades through interaction with TRAFs. In B cells, expression of TRAF1 is in turn upregulated by LMP1. LMP1 signaling in epithelial cells may be affected by the presence or absence of TRAF1. By immunohistochemistry, we detected TRAF1 expression in 17 of 42 (40%) EBV+ undifferentiated NPCs. All 7 LMP1+ NPC biopsies were also TRAF1+. Using an RNAse protection assay, high-level TRAF1 expression was detected in an LMP1-expressing NPC-derived cell line (C15) and expression was weaker in 2 LMP1- cell lines (C17, C19). Finally, LMP1 upregulated TRAF1 expression in an EBV- keratinocyte cell line. Our results demonstrate that TRAF1 is expressed in NPC tumor cells in vivo and suggest that TRAF1 expression may be upregulated by LMP1 in NPC. An antiapoptotic function has been proposed for TRAF1, and this may be relevant for the pathogenesis of NPC.
Collapse
Affiliation(s)
- Gabriele Siegler
- Institute for Pathology, Friedrich-Alexander-University, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Morrison JA, Gulley ML, Pathmanathan R, Raab-Traub N. Differential signaling pathways are activated in the Epstein-Barr virus-associated malignancies nasopharyngeal carcinoma and Hodgkin lymphoma. Cancer Res 2004; 64:5251-60. [PMID: 15289331 DOI: 10.1158/0008-5472.can-04-0538] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
EBV is associated with the epithelial cancer, nasopharyngeal carcinoma (NPC), and the lymphoid malignancy, Hodgkin lymphoma (HL). The EBV latent membrane proteins 1 and 2A are expressed in these tumors. These proteins activate the phosphatidylinositol 3'-OH kinase (PI3K)/Akt pathway, which is commonly activated inappropriately in malignancy. In this study, the status of Akt activation and its targets, glycogen synthase kinase-3beta (GSK-3beta) and beta-catenin, was investigated in NPC and HL clinical specimens. In the majority of HL and NPC specimens, Akt was activated, indicating an important role for this kinase in the development and/or progression of these tumors. Akt phosphorylates and inactivates GSK-3beta, a negative regulator of the proto-oncoprotein beta-catenin that is aberrantly activated in many cancers. GSK-3beta was phosphorylated and inactivated with concomitant nuclear beta-catenin accumulation in the majority of NPC specimens. The malignant cells of the majority of HL cases, however, did not have inactivated GSK-3beta and lacked nuclear beta-catenin expression. These data indicate that this signaling arm of PI3K/Akt is universal and important in NPC pathogenesis but is apparently not affected in HL. These findings point to a divergence in pathways activated by EBV in different cellular contexts.
Collapse
Affiliation(s)
- Jennifer A Morrison
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | | | | | | |
Collapse
|
209
|
Stunz LL, Busch LK, Munroe ME, Sigmund CD, Tygrett LT, Waldschmidt TJ, Bishop GA. Expression of the Cytoplasmic Tail of LMP1 in Mice Induces Hyperactivation of B Lymphocytes and Disordered Lymphoid Architecture. Immunity 2004; 21:255-66. [PMID: 15308105 DOI: 10.1016/j.immuni.2004.07.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Revised: 06/18/2004] [Accepted: 06/23/2004] [Indexed: 01/20/2023]
Abstract
The oncogenic EBV protein LMP1 mimics a dysregulated CD40 receptor in vitro. To compare CD40 and LMP1-mediated events in vivo, transgenic mice were engineered to express mouse CD40 (mCD40tg) or a protein with extracellular mCD40 and cytoplasmic LMP1 (mCD40-LMP1tg). Transgenic and CD40(-/-) mice were bred so that only the transgenic CD40 molecule is expressed in B cells, macrophages, and dendritic cells. mCD40-LMP1tg mice had normal lymphocyte subsets, and immunization elicited an antibody response featuring normal isotype switching, affinity maturation, and germinal center (GC) formation. However, unimmunized mCD40-LMP1tg mice had expanded immature and germinal center B cells, produced autoantibodies, exhibited marked splenomegaly and lymphadenopathy, and elevated serum IL-6. Thus, signaling through the LMP1 cytoplasmic tail results in amplified and abnormal mimicry of CD40 functions in vivo, indicating possible ways in which LMP1 contributes to the pathogenesis of EBV-associated human disease.
Collapse
Affiliation(s)
- Laura L Stunz
- Department of Microbiology, Univeristy of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | |
Collapse
|
210
|
Burrows JM, Bromham L, Woolfit M, Piganeau G, Tellam J, Connolly G, Webb N, Poulsen L, Cooper L, Burrows SR, Moss DJ, Haryana SM, Ng M, Nicholls JM, Khanna R. Selection pressure-driven evolution of the Epstein-Barr virus-encoded oncogene LMP1 in virus isolates from Southeast Asia. J Virol 2004; 78:7131-7. [PMID: 15194789 PMCID: PMC421669 DOI: 10.1128/jvi.78.13.7131-7137.2004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The geographically constrained distribution of Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC) in southeast Asian populations suggests that both viral and host genetics may influence disease risk. Although susceptibility loci have been mapped within the human genome, the role of viral genetics in the focal distribution of NPC remains an enigma. Here we report a molecular phylogenetic analysis of an NPC-associated viral oncogene, LMP1, in a large panel of EBV isolates from southeast Asia and from Papua New Guinea, Africa, and Australia, regions of the world where NPC is and is not endemic, respectively. This analysis revealed that LMP1 sequences show a distinct geographic structure, indicating that the southeast Asian isolates have evolved as a lineage distinct from those of Papua New Guinea, African, and Australian isolates. Furthermore, a likelihood ratio test revealed that the C termini of the LMP1 sequences of the southeast Asian lineage are under significant positive selection pressure, particularly at some sites within the C-terminal activator regions. We also present evidence that although the N terminus and transmembrane region of LMP1 have undergone recombination, the C-terminal region of the gene has evolved without any history of recombination. Based on these observations, we speculate that selection pressure may be driving the LMP1 sequences in virus isolates from southeast Asia towards a more malignant phenotype, thereby influencing the endemic distribution of NPC in this region.
Collapse
Affiliation(s)
- Jacqueline M Burrows
- Tumour Immunology Laboratory, Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research, and Department of Molecular and Cellular Pathology, University of Queensland, Brisbane, Australia 4029
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
211
|
Snyder MJ, Stenzel TT, Buckley PJ, Lagoo AS, Rizzieri DA, Gasparetto C, Vredenburgh JJ, Chao NJ, Gong JZ. Posttransplant lymphoproliferative disorder following nonmyeloablative allogeneic stem cell transplantation. Am J Surg Pathol 2004; 28:794-800. [PMID: 15166672 DOI: 10.1097/01.pas.0000126052.88174.f] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Posttransplantation lymphoproliferative disorder (PTLD) is a well-recognized complication of conventional bone marrow/stem cell and solid organ transplantation. However, not much is known about PTLD following the more recently introduced nonmyeloablative allogeneic stem cell transplantation (NMST). This study reports the findings from two cases of PTLD following NMST and compares them to the one previously reported case. The donor origin of the PTLD was determined using short tandem repeat analysis, and B- and T-cell clonalities were evaluated by polymerase chain reaction. Two cases of PTLD evolved in a total of 70 patients who have undergone NMST at our institution from 1999 to 2003. Both patients received conditioning with Fludarabine/Cytoxan/Campath 1H (alemtuzumab, anti-CD52 antibody) and T-cell-depleted donor cells with Campath-1H. Both PTLDs were EBV positive (by immunohistochemistry and in situ hybridization) with diffuse large B-cell lymphoma morphology. Our findings indicate the incidence of PTLD following NMST is 3% (2 of 70 patients from our institution and 1 of 30 from the previously reported case). All three PTLDs arose 6 to 7 months after NMST and were rapidly fatal. The pathology of the PTLD in all cases was donor origin, EBV positive, diffuse large B-cell lymphoma.
Collapse
MESH Headings
- Adult
- Alemtuzumab
- Antibodies/therapeutic use
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal, Humanized
- Antibodies, Neoplasm/administration & dosage
- Antigens, CD/immunology
- Antigens, Neoplasm/immunology
- CD52 Antigen
- Female
- Glycoproteins/immunology
- Hematopoietic Stem Cell Transplantation/adverse effects
- Herpesvirus 4, Human/isolation & purification
- Humans
- In Situ Hybridization
- Lymphoma, B-Cell/pathology
- Lymphoma, B-Cell/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoproliferative Disorders/etiology
- Middle Aged
- Myelodysplastic Syndromes/therapy
- Polymerase Chain Reaction
- Tandem Repeat Sequences
- Transplantation Conditioning
Collapse
Affiliation(s)
- Matthew J Snyder
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Adriaenssens E, Mougel A, Goormachtigh G, Loing E, Fafeur V, Auriault C, Coll J. A novel dominant-negative mutant form of Epstein-Barr virus latent membrane protein-1 (LMP1) selectively and differentially impairs LMP1 and TNF signaling pathways. Oncogene 2004; 23:2681-93. [PMID: 14767477 DOI: 10.1038/sj.onc.1207432] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The latent membrane protein-1 (LMP1) is an integral membrane molecule expressed by Epstein-Barr virus (EBV) during viral latency and displays properties of a constitutively activated member of the TNF receptor family. LMP1 is required for B-cell or monocyte immortalization induced by EBV and is sufficient to transform rodent fibroblasts. Transforming potential of LMP1 is mediated by its cytoplasmic C-terminal domain, which activates various cellular signaling pathways including NFkappaB and JNK. In this report, we constructed mutants of LMP1 with preserved membrane spanning domain but mutated in the C-terminal domain and a second truncated C-terminal LMP1 fused to the enhanced green fluorescent protein. This latter mutant, termed LMP1-CT, impairs signaling by ectopic LMP1 as well as endogenous EBV-expressed wild-type (wt) LMP1. In contrast to dominant-negative mutants of LMP1 with preserved membrane spanning domains, LMP1-CT was unable to bind wt LMP1 to form an inactive complex. Its dominant-negative effects were due to binding and sequestration of LMP1 adapters TRAF2 and TRADD as assessed by coimmunoprecipitation experiments and confocal analysis. The effect was selective since LMP1-CT did not inhibit IL-1beta-induced signaling, whereas it impaired TNF-triggered NFkappaB and JNK signals without affecting TNF-induced apoptosis. In addition and in contrast to LMP1 constructs with membrane localization, LMP-CT did not display cytostatic properties in noninfected cells. Importantly, LMP1-CT inhibited survival induced by LMP1 in an EBV-transformed T-cell line expressing the type II viral latency commonly found in the majority of EBV-associated human tumors. These data demonstrate that LMP1-CT is a new tool to explore the differences between LMP1 and TNF signaling and may facilitate the design of molecules with potential therapeutic roles.
Collapse
Affiliation(s)
- Eric Adriaenssens
- CNRS UMR 8527, Institut de Biologie de Lille, BP 447, 59021 Lille Cedex, France
| | | | | | | | | | | | | |
Collapse
|
213
|
Rossi D, Gaidano G. Molecular heterogeneity of diffuse large B-cell lymphoma: implications for disease management and prognosis. ACTA ACUST UNITED AC 2004; 7:239-52. [PMID: 14972786 DOI: 10.1080/1024533021000024058] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) accounts for approximately 40% of all B-cell non-Hodgkin lymphomas of the Western world. According to the "WHO classification of tumours of the haematopoietic and lymphoid tissues", the term DLBCL is likely to include more than one disease entity, as suggested by the marked variability of the clinical presentation and response to treatment of this disease. Such heterogeneity may reflect the occurrence of distinct molecular subtypes of DLBCL as well as differences in the host's immune function. In immunocompetent hosts, approximately 50% DLBCL carry one of two primary molecular lesions defining two distinct genotypic subgroups, characterized by activation of either the BCL-6 or the BCL-2 proto-oncogene. Conversely, the remaining DLBCL of immunocompetent hosts display one of several molecular lesions, each associated with a small subset of cases and including activation of the proto-oncogenes REL, MUC-1, BCL-8 and c-MYC. The molecular pathogenesis of immunodeficiency-associated DLBCL differs substantially from that of DLBCL in immunocompetent hosts. In fact, EBV infection is present in a large fraction of immunodeficiency-associated DLBCL, whereas it is consistently negative in DLBCL of immunocompetent hosts, probably reflecting the critical role of disruption of the immune system in this disease. Finally, the application of DNA microarray technology to DLBCL has led to the distinction of two disease variants: a germinal center like DLBCL and an activated peripheral B-cell like DLBCL. Overall the molecular features of DLBCL may identify prognostic categories of the disease and may represent a powerful tool for therapeutic stratification.
Collapse
MESH Headings
- Adult
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- DNA-Binding Proteins/genetics
- Genes, bcl-2
- Genes, myc
- Genes, rel
- Genetic Heterogeneity
- Germinal Center/pathology
- Humans
- Immunocompetence
- Immunocompromised Host
- Lymphoma, AIDS-Related/genetics
- Lymphoma, Large B-Cell, Diffuse/classification
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Mucin-1/genetics
- Neoplasm Proteins/genetics
- Postoperative Complications
- Prognosis
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins c-bcl-6
- Proto-Oncogenes
- Somatic Hypermutation, Immunoglobulin
- Transcription Factors/genetics
- Transplantation
Collapse
Affiliation(s)
- Davide Rossi
- Hematology Unit, Division of Internal Medicine, Department of Medical Sciences and IRCAD, Amedeo Avogadro University of Eastern Piedmont, Via Solaroli 17, I-28100, Novara, Italy
| | | |
Collapse
|
214
|
Abstract
Epstein-Barr virus (EBV), a human herpesvirus associated with lymphoid and epithelial cell tumors, encodes several proteins that exploit the ubiquitin-proteasome system to regulate latency and allow the persistence of infected cells in immunocompetent hosts. Further modifications of ubiquitin-dependent proteolysis by activated cellular oncogenes contribute to malignant transformation. A detailed understanding of these processes may lead to the development of new therapeutic strategies for EBV-associated cancers.
Collapse
Affiliation(s)
- Maria G Masucci
- Microbiology and Tumor Biology Center, Karolinska Institutet, Box 280, S-171 77 Stockholm, Sweden.
| |
Collapse
|
215
|
D'Souza BN, Edelstein LC, Pegman PM, Smith SM, Loughran ST, Clarke A, Mehl A, Rowe M, Gélinas C, Walls D. Nuclear factor kappa B-dependent activation of the antiapoptotic bfl-1 gene by the Epstein-Barr virus latent membrane protein 1 and activated CD40 receptor. J Virol 2004; 78:1800-16. [PMID: 14747545 PMCID: PMC369510 DOI: 10.1128/jvi.78.4.1800-1816.2004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Suppression of the cellular apoptotic program by the oncogenic herpesvirus Epstein-Barr virus (EBV) is central to both the establishment of latent infection and the development of EBV-associated malignancies. We have previously shown that expression of the EBV latent membrane protein 1 (LMP1) in Burkitt's lymphoma cell lines leads to increased mRNA levels from the cellular antiapoptotic bfl-1 gene (also known as A1). Furthermore, ectopic expression of Bfl-1 in an EBV-positive cell line exhibiting a latency type 1 infection protects against apoptosis induced by growth factor deprivation (B. N. D'Souza, M. Rowe, and D. Walls, J. Virol. 74:6652-6658, 2000). We now report that LMP1 drives bfl-1 promoter activity through interactions with components of the tumor necrosis factor receptor (TNFR)/CD40 signaling pathway. We present evidence that this process is NF-kappa B dependent, involves the recruitment of TNFR-associated factor 2, and is mediated to a greater extent by the carboxyl-terminal activating region 2 (CTAR2) relative to the CTAR1 domain of LMP1. Activation of CD40 receptor also led to increased bfl-1 mRNA levels and an NF-kappa B-dependent increase in bfl-1 promoter activity in Burkitt's lymphoma-derived cell lines. We have delineated a 95-bp region of the promoter that functions as an LMP1-dependent transcriptional enhancer in this cellular context. This sequence contains a novel NF-kappa B-like binding motif that is essential for transactivation of bfl-1 by LMP1, CD40, and the NF-kappa B subunit protein p65. These findings highlight the role of LMP1 as a mediator of EBV-host cell interactions and may indicate an important route by which it exerts its cellular growth transforming properties.
Collapse
Affiliation(s)
- Brendan N D'Souza
- School of Biotechnology and National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Lam N, Sandberg ML, Sugden B. High physiological levels of LMP1 result in phosphorylation of eIF2 alpha in Epstein-Barr virus-infected cells. J Virol 2004; 78:1657-64. [PMID: 14747531 PMCID: PMC369503 DOI: 10.1128/jvi.78.4.1657-1664.2004] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
LMP1 is an Epstein-Barr virus (EBV)-encoded membrane protein essential for the proliferation of EBV-infected lymphoblasts (E. Kilger, A. Kieser, M. Baumann, and W. Hammerschmidt, EMBO J. 17:1700-1709, 1998). LMP1 also inhibits gene expression and induces cytostasis in transfected cells when it is expressed at levels as little as twofold higher than the average for EBV-positive lymphoblasts (M. Sandberg, A. Kaykas, and B. Sugden, J. Virol. 74:9755-9761, 2000; A. Kaykas and B. Sugden, Oncogene 19:1400-1410, 2000). We have found that in three different clones of EBV-infected lymphoblasts the levels of expression of LMP1 in individual cells in each clone ranged over 100-fold. This difference is due to a difference in levels of the LMP1 transcript. In these clones, cells expressing high levels of LMP1 incorporated less BrdU. We also found that induction of expression of LMP1 or of a derivative of LMP1 with its transmembrane domain fused to green fluorescent protein instead of its carboxy-terminal signaling domain resulted in phosphorylation of eIF2 alpha in EBV-negative Burkitt's lymphoma cells. This induction of phosphorylation of eIF2 alpha was also detected in EBV-infected lymphoblasts, in which high levels of LMP1 correlated with high levels of phosphorylation of eIF2 alpha. Our results indicate that inhibition of gene expression and of cell proliferation by LMP1 occurs normally in EBV-infected cells.
Collapse
Affiliation(s)
- Ngan Lam
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
217
|
Xie P, Hostager BS, Bishop GA. Requirement for TRAF3 in signaling by LMP1 but not CD40 in B lymphocytes. ACTA ACUST UNITED AC 2004; 199:661-71. [PMID: 14981114 PMCID: PMC2213302 DOI: 10.1084/jem.20031255] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
CD40, a member of the tumor necrosis factor receptor family, and the Epstein-Barr virus–encoded oncoprotein latent membrane protein 1 (LMP1) share several tumor necrosis factor receptor–associated factor (TRAF) adaptor proteins for signaling. Among these, TRAF3 was the first identified to directly bind both receptors, yet its role remains a mystery. To address this, we generated B cell lines deficient in TRAF3 by homologous recombination. We found that CD40 signals were normal in the absence of TRAF3, with the exception of moderately enhanced c-Jun NH2-terminal kinase (JNK) activation and antibody secretion. In sharp contrast, LMP1 signaling was markedly defective in TRAF3−/− B cells. LMP1-induced activation of JNK and nuclear factor κB, up-regulation of CD23 and CD80, and antibody secretion were substantially affected by TRAF3 deficiency. Reconstitution of TRAF3 expression decreased CD40-induced JNK activation and antibody secretion, and fully restored LMP1 signaling. Although TRAF2 is widely believed to be important for LMP1 function, LMP1 signaling was intact in TRAF2−/− B cells. Our data reveal that CD40 and LMP1 unexpectedly use TRAF3 in different ways, and that TRAF3 is required for LMP1-mediated activation of B cells.
Collapse
Affiliation(s)
- Ping Xie
- Department of Microbiology, University of Iowa, Iowa City 52242, USA
| | | | | |
Collapse
|
218
|
Wan J, Sun L, Mendoza JW, Chui YL, Huang DP, Chen ZJ, Suzuki N, Suzuki S, Yeh WC, Akira S, Matsumoto K, Liu ZG, Wu Z. Elucidation of the c-Jun N-terminal kinase pathway mediated by Estein-Barr virus-encoded latent membrane protein 1. Mol Cell Biol 2004; 24:192-9. [PMID: 14673155 PMCID: PMC303354 DOI: 10.1128/mcb.24.1.192-199.2004] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Epstein-Barr virus (EBV) is associated with several human diseases including infectious mononucleosis and nasopharyngeal carcinoma. EBV-encoded latent membrane protein 1 (LMP1) is oncogenic and indispensable for cellular transformation caused by EBV. Expression of LMP1 in host cells constitutively activates both the c-Jun N-terminal kinase (JNK) and NF-kappaB pathways, which contributes to the oncogenic effect of LMP1. However, the underlying signaling mechanisms are not very well understood. Based mainly on overexpression studies with various dominant-negative constructs, LMP1 was generally thought to functionally mimic members of the tumor necrosis factor (TNF) receptor superfamily in signaling. In contrast to the prevailing paradigm, using embryonic fibroblasts from different knockout mice and the small interfering RNA technique, we find that the LMP1-mediated JNK pathway is distinct from those mediated by either TNF-alpha or interleukin-1. Moreover, we have further elucidated the LMP1-mediated JNK pathway by demonstrating that LMP1 selectively utilizes TNF receptor-associated factor 6, TAK1/TAB1, and c-Jun N-terminal kinase kinases 1 and 2 to activate JNK.
Collapse
Affiliation(s)
- Jun Wan
- Department of Biochemistry, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Meyer RM, Ambinder RF, Stroobants S. Hodgkin's lymphoma: evolving concepts with implications for practice. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2004; 2004:184-202. [PMID: 15561683 DOI: 10.1182/asheducation-2004.1.184] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Hodgkin's lymphoma is a unique neoplasm of B lymphocytes. Recent data provide new understandings of the pathogenesis and options for staging and therapy of the disease. Three specific topics are addressed in this chapter. In Section I, Dr. Richard Ambinder reviews implications of the relationship of Epstein-Barr virus (EBV) and Hodgkin's lymphoma. This relation includes varying geographic epidemiologic associations, including varying associations with the clinical syndrome of infectious mononucleosis. There are plausible mechanisms, including processes initiated by viral proteins, by which EBV might lead to tumorigenesis. These mechanisms include promotion of genetic instability and alteration of normal processes of apoptosis. In addition to an epidemiologic association and potential role in pathogenesis, viral antigens may pose theoretical targets for anti-cancer therapies, including vaccination. In Section II, Dr. Sigrid Stroobants describes the potential role of positron emission tomographic (PET) scanning. By assessing differences in the metabolic activities of cancer cells, PET scanning may be superior to computerized tomographic scanning, which is limited to showing structural anatomical abnormalities. In patients with Hodgkin's and non-Hodgkin's lymphoma, PET scanning has been tested as an initial staging tool, to assess the rate of therapeutic response from a prognostic perspective, and to differentiate residual tumor from fibrotic masses in patients who have completed therapy. Particularly in assessing the nature of a residual mass seen with other post-therapeutic imaging modalities, PET scanning may provide unique information; very high negative predictive values have been reported. However, before this technology can be recommended for incorporation into standard management, properly conducted prospective trials are required to better evaluate the clinical utility of PET with respect to eventual patient outcomes. In Section III, Dr. Ralph Meyer reviews current data regarding the management of patients with limited-stage Hodgkin's lymphoma. Over the past decade, standard treatment has evolved to consist of combined-modality therapy that includes an abbreviated course of chemotherapy and involved-field radiation. As this therapy continues to include radiation therapy, patients will remain at risk of long-term toxicities that include the development of second cancers and cardiovascular events. These "late-effects" now account for more deaths than those attributed to progressive Hodgkin's lymphoma. Comparative data testing the role of chemotherapy alone are now available and demonstrate that omission of radiation therapy results in small but statistically significant reduction in disease control, but no detectable differences in overall survival. Further follow-up will clarify whether chemotherapy alone is the preferred treatment option; at present patients should be informed of the trade-offs involved in choosing between this option and combined modality therapy.
Collapse
Affiliation(s)
- Ralph M Meyer
- Juravinski Cancer Centre and McMaster University, Hamilton ONT, Canada
| | | | | |
Collapse
|
220
|
Tarantul VZ. Transgenic Mice as an In Vivo Model of Lymphomagenesis. INTERNATIONAL REVIEW OF CYTOLOGY 2004; 236:123-80. [PMID: 15261738 DOI: 10.1016/s0074-7696(04)36004-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This review covers multiple data obtained on genetically modified mice that help to elucidate various intricate molecular mechanisms of lymphomagenesis in humans. We are in a "golden age" of mouse genetics. The mouse is by far the most accessible mammalian system physiologically similar to humans. Transgenic mouse models have illuminated how different genes contribute to human lymphomagenesis. Multiple experiments with transgenic mice have not only confirmed the data obtained for human lymphomas but also gave additional evidence for the role of some genes and cooperative participation of their products in the development of human lymphomas. Genes and gene networks detected on transgenic mice can successfully serve as molecular targets for tumor therapy. This review demonstrates the extraordinary possibilities of transgenic technology, which is presently one of the readily available, efficient, and accurate tools to solve the problem of cancer.
Collapse
Affiliation(s)
- V Z Tarantul
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow 123182, Russia
| |
Collapse
|
221
|
Izumi KM. Epstein-Barr virus signal transduction and B-lymphocyte growth transformation. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2004; 36:269-88. [PMID: 15171616 DOI: 10.1007/978-3-540-74264-7_13] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Latent EBV growth transformation of resting B-cells into indefinitely proliferating cell lines is a successful viral strategy for survival in its host and the basis of several human malignancies. EBV transforms cell growth through viral proteins that modify cell gene expression at the level of transcription or by appropriating signaling pathways. Analyses of the EBV-transforming protein LMP1 have begun to reveal that this receptor transduces critical signals by appropriating the TNF receptor signal transduction pathway to activate NF-kappaB and MAPK. While this has brought an important aspect into clearer focus, future progress in delineating the underlying mechanism of transformation, which will be essential to devising effective therapies to treat EBV-associated malignancies, will depend on resolving the intricacies of TRAF signal transduction. Since expression of cytokines, receptors, and anti-apoptotic proteins are regulated by TRAF signaling, another critical issue is delineating the genes that are specifically targeted by LMP1 in order to transform B-lymphocyte growth.
Collapse
Affiliation(s)
- K M Izumi
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas 78229-3900, USA.
| |
Collapse
|
222
|
Affiliation(s)
- Jenny O'Nions
- Faculty of Medicine, Department of Virology and Ludwig Institute for Cancer Research, Imperial College London, Norfolk Place, London W2 1PG, UK
| | | |
Collapse
|
223
|
Luftig M, Prinarakis E, Yasui T, Tsichritzis T, Cahir-McFarland E, Inoue JI, Nakano H, Mak TW, Yeh WC, Li X, Akira S, Suzuki N, Suzuki S, Mosialos G, Kieff E. Epstein-Barr virus latent membrane protein 1 activation of NF-kappaB through IRAK1 and TRAF6. Proc Natl Acad Sci U S A 2003; 100:15595-600. [PMID: 14673102 PMCID: PMC307613 DOI: 10.1073/pnas.2136756100] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Epstein-Barr virus latent membrane protein 1 (LMP1) activation of NF-kappaB is critical for Epstein-Barr virus-infected B lymphocyte survival. LMP1 activates the IkappaB kinase complex and NF-kappaB through two cytoplasmic signaling domains that engage tumor necrosis factor receptor-associated factor (TRAF)1/2/3/5 or TRADD and RIP. We now use cells lacking expression of TRAF2, TRAF5, TRAF6, IKKalpha, IKKbeta, IKKgamma, TAB2, IL-1 receptor-associated kinase (IRAK)1, or IRAK4 to assess their roles in LMP1-mediated NF-kappaB activation. LMP1-induced RelA nuclear translocation was similar in IKKalpha knockout (KO) and WT murine embryo fibroblasts (MEFs) but substantially deficient in IKKbeta KO MEFs. NF-kappaB-dependent promoter responses were also substantially deficient in IKKbeta KO MEFs but were hyperactive in IKKalpha KO MEFs. More surprisingly, NF-kappaB responses were near normal in TRAF2 and TRAF5 double-KO MEFs, IKKgamma KO MEFs, TAB2 KO MEFs, and IRAK4 KO MEFs but were highly deficient in TRAF6 KO MEFs and IRAK1 KO HEK293 cells. Consistent with the importance of TRAF6, LMP1-induced NF-kappaB activation in HEK293 cells was inhibited by expression of dominant-negative TAB2 and Ubc13 alleles. These data extend a role for IKKalpha in IKKbeta regulation, identify an unusual IKKbeta-dependent and IKKgamma-independent NF-kappaB activation, and indicate that IRAK1 and TRAF6 are essential for LMP1-induced NF-kappaB activation.
Collapse
Affiliation(s)
- Micah Luftig
- Program in Virology, Department of Microbiology and Molecular Genetics, Harvard Medical School, Channing Laboratory/Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Tellam J, Connolly G, Webb N, Duraiswamy J, Khanna R. Proteasomal targeting of a viral oncogene abrogates oncogenic phenotype and enhances immunogenicity. Blood 2003; 102:4535-40. [PMID: 12920032 DOI: 10.1182/blood-2003-03-0870] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ability of viral or mutated cellular oncogenes to initiate neoplastic events and their poor immunogenicity have considerably undermined their potential use as immunotherapeutic tools for the treatment of human cancers. Using an Epstein-Barr virus-encoded oncogene, latent membrane protein 1 (LMP1), as a model, we report a novel strategy that both deactivates cellular signaling pathways associated with the oncogenic phenotype and reverses poor immunogenicity. We show that cotranslational ubiquitination combined with N-end rule targeting of LMP1 enhanced the intracellular degradation of LMP1 and total blockade of LMP1-mediated nuclear factor-kappaB (NF-kappaB) and signal transducer and activator of transcription (STAT) activation in human cells. In addition, although murine cells expressing LMP1 were uniformly tumorigenic, this oncogenicity was completely abrogated by covalent linkage of LMP1 with ubiquitin, while an enhanced CD8+ T cell response to a model epitope fused to the C-terminus of LMP1 was observed following immunization with ubiquitinated LMP1. These observations suggest that proteasomal targeting of tumor-associated oncogenes could be exploited therapeutically by either gene therapy or vaccination.
Collapse
Affiliation(s)
- Judy Tellam
- Queensland Institute of Medical Research, Bancroft Centre, 300 Herston Rd, Brisbane, Australia 4029
| | | | | | | | | |
Collapse
|
225
|
Eliopoulos AG, Caamano JH, Flavell J, Reynolds GM, Murray PG, Poyet JL, Young LS. Epstein-Barr virus-encoded latent infection membrane protein 1 regulates the processing of p100 NF-kappaB2 to p52 via an IKKgamma/NEMO-independent signalling pathway. Oncogene 2003; 22:7557-69. [PMID: 14576817 DOI: 10.1038/sj.onc.1207120] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The oncogenic Epstein-Barr virus (EBV)-encoded latent infection membrane protein 1 (LMP1) constitutively activates the 'canonical' NF-kappaB pathway that involves the phosphorylation and degradation of IkappaBalpha downstream of the IkappaB kinases (IKKs). In this study, we show that LMP1 also promotes the proteasome-mediated proteolysis of p100 NF-kappaB2 resulting in the generation of active p52, which translocates to the nucleus in complex with the p65 and RelB NF-kappaB subunits. LMP1-induced NF-kappaB transactivation is reduced in nf-kb2(-/-) mouse embryo fibroblasts, suggesting that p100 processing contributes to LMP1-mediated NF-kappaB transcriptional effects. This pathway is likely to operate in vivo, as the expression of LMP1 in primary EBV-positive Hodgkin's lymphoma and nasopharyngeal carcinoma biopsies correlates with the nuclear accumulation of p52. Interestingly, while the ability of LMP1 to activate the canonical NF-kappaB pathway is impaired in cells lacking IKKgamma/NEMO, the regulatory subunit of the IKK complex, p100 processing remains unaffected. As a result, nuclear translocation of p52, but not p65, occurs in the absence of IKKgamma. These data point to the existence of a novel signalling pathway that regulates NF-kappaB in LMP1-expressing cells, and may thereby play a role in both oncogenic transformation and the establishment of persistent EBV infection.
Collapse
Affiliation(s)
- Aristides G Eliopoulos
- Cancer Research UK Institute for Cancer Studies, The University of Birmingham Medical School, Birmingham B15 2TA, UK.
| | | | | | | | | | | | | |
Collapse
|
226
|
Atkinson PGP, Coope HJ, Rowe M, Ley SC. Latent Membrane Protein 1 of Epstein-Barr Virus Stimulates Processing of NF-κB2 p100 to p52. J Biol Chem 2003; 278:51134-42. [PMID: 14532284 DOI: 10.1074/jbc.m304771200] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recent studies have identified a limited number of cellular receptors that can stimulate an alternative NF-kappa B activation pathway that depends upon the inducible processing of NF-kappa B2 p100 to p52. Here it is shown that the latent membrane protein (LMP)-1 of Epstein-Barr virus can trigger this signaling pathway in both B cells and epithelial cells. LMP1-induced p100 processing, which is mediated by the proteasome and is dependent upon de novo protein synthesis, results in the nuclear translocation of p52.RelB dimers. Previous studies have established that LMP1 also stimulates the canonical NF-kappa B-signaling pathway that triggers phosphorylation and degradation of I kappa B alpha. Interestingly, LMP1 activation of these two NF-kappa B pathways is shown here to require distinct regions of the LMP1 C-terminal cytoplasmic tail. Thus, C-terminal-activating region 1 is required for maximal triggering of p100 processing but is largely dispensable for stimulation of I kappa B alpha phosphorylation. In contrast, C-terminal-activating region 2 is critical for maximal LMP1 triggering of I kappa B alpha phosphorylation and up-regulation of p100 levels but does not contribute to activation of p100 processing. Because p100 deletion mutants that constitutively produce p52 oncogenically transform fibroblasts in vitro, it is likely that stimulation of p100 processing by LMP1 will play an important role in its transforming function.
Collapse
Affiliation(s)
- Peter G P Atkinson
- Division of Immune Cell Biology, National Institute for Medical Research, Mill Hill, London, NW7 1AA, United Kingdom
| | | | | | | |
Collapse
|
227
|
Tang W, Pavlish OA, Spiegelman VS, Parkhitko AA, Fuchs SY. Interaction of Epstein-Barr Virus Latent Membrane Protein 1 with SCFHOS/β-TrCP E3 Ubiquitin Ligase Regulates Extent of NF-κB Activation. J Biol Chem 2003; 278:48942-9. [PMID: 14523018 DOI: 10.1074/jbc.m307962200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The Epstein-Barr virus latent membrane protein 1 (LMP1) is pivotal in the transforming activity of this virus. We found that the common LMP1-95-8 variant interacts with Homologue of Slimb (HOS), a receptor for the SCFHOS/betaTrCP ubiquitin-protein isopeptide ligase (E3) via one canonical and one cryptic HOS recognition site. These sites are mutated or deleted in the tumor-derived LMP1-Cao variant, which did not bind to HOS. Mutations within these sites on LMP1-95-8 abrogated HOS binding and increased transforming activity of LMP1. HOS did not regulate stability of LMP1-95-8 unless it was mutated to bear additional lysine residues near the cryptic motif. LMP1 proteins that could not bind to HOS exhibited an increased ability to induce IkappaB degradation and NF-kappaB-mediated transcription without further increase in activation of IkappaB kinases. Expression of LMP1-95-8 reduced the levels of endogenous HOS available to interact with phosphorylated IkappaBalpha. Degradation of IkappaBalpha and dose dependence of NF-kappaB activation by LMP1-95-8 were promoted by co-expression of HOS. Our data suggest that LMP1-95-8 is a pseudo-substrate of SCFHOS/betaTrCP E3 ubiquitin ligase and that interaction between LMP1 and HOS restricts the extent of LMP1-induced NF-kappaB signaling. We discuss the potential role of this mechanism in transforming and cytostatic effects of LMP1 variants in cells and Epstein-Barr virus-associated tumors.
Collapse
Affiliation(s)
- Weigang Tang
- Department of Animal Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
228
|
Küppers R. B cells under influence: transformation of B cells by Epstein-Barr virus. Nat Rev Immunol 2003; 3:801-12. [PMID: 14523386 DOI: 10.1038/nri1201] [Citation(s) in RCA: 364] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Epstein-Barr virus (EBV) is an extremely successful virus, infecting more than 90% of the human population worldwide. After primary infection, the virus persists for the life of the host, usually as a harmless passenger residing in B cells. However, EBV can transform B cells, which can result in the development of malignant lymphomas. Intriguingly, the three main types of EBV-associated B-cell lymphoma - that is, Burkitt lymphoma, Hodgkin lymphoma and post-transplant lymphomas - seem to derive from germinal-centre B cells or atypical survivors of the germinal-centre reaction in most, if not all, cases, indicating that EBV-infected germinal-centre B cells are at particular risk for malignant transformation.
Collapse
Affiliation(s)
- Ralf Küppers
- Institute for Genetics, Department of Internal Medicine I, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
229
|
Demachi A, Nagata H, Morio T, Oyoshi MK, Zhang Y, Tabata N, Kimura N, Shimizu N, Yamamoto K. Characterization of Epstein-Barr virus (EBV)-positive NK cells isolated from hydroa vacciniforme-like eruptions. Microbiol Immunol 2003; 47:543-52. [PMID: 12953848 DOI: 10.1111/j.1348-0421.2003.tb03416.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Recently, the involvement of Epstein-Barr virus (EBV) in hydroa vacciniforme (HV)-like eruptions has been suggested. To elucidate the role of EBV in this disease, we isolated EBV-infected cell clones from peripheral blood mononuclear cells (PBMC) and the skin lesions of a patient with HV-like eruptions; cells isolated from PBMC were designated SNK-12, and those from the eruption SNK-11. Both cells expressed CD16, CD56, and HLA-DR and had germline configurations of the T-cell receptor and the immunoglobulin genes, indicating that the cell clones were of NK cell lineage. The analysis of EBV terminal repeats indicated that the cells were monoclonal, had identical clonality, and originated from EBV-positive cells in the PBMC and eruption. Both clones expressed EBNA-1, but not EBNA-2. Although LMP-1 was weakly detected in SNK-11, no LMP-1 was detected in SNK-12. Interestingly, EBV-infected cells required less IL-2 for in vitro growth in the later phase of this disease and this appeared to correlate with the expression of LMP-1, suggesting that the proliferative capacity of the EBV-positive NK cells increased during the time course of the disease, and LMP-1 expression might be responsible for that. This is the first report of the isolation of EBV-infected cells from the skin lesions of HV-like eruptions and strongly suggests that the HV-like eruption in the patient was caused by clonal NK cells with latent EBV infection.
Collapse
MESH Headings
- Cell Culture Techniques
- Cell Lineage
- Cell Separation
- Epstein-Barr Virus Infections/immunology
- Epstein-Barr Virus Infections/pathology
- Epstein-Barr Virus Infections/virology
- Epstein-Barr Virus Nuclear Antigens/biosynthesis
- Gene Expression
- Genes, Immunoglobulin
- Genes, T-Cell Receptor
- Genes, Viral
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/metabolism
- Herpesvirus 4, Human/pathogenicity
- Immunophenotyping
- Interleukin-2/metabolism
- Killer Cells, Natural/pathology
- Killer Cells, Natural/virology
- Skin Diseases, Vesiculobullous/immunology
- Skin Diseases, Vesiculobullous/pathology
- Skin Diseases, Vesiculobullous/virology
- Viral Matrix Proteins/biosynthesis
- Viral Proteins
Collapse
Affiliation(s)
- Ayako Demachi
- Department of Virology, Division of Medical Science, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Nash RA, Dansey R, Storek J, Georges GE, Bowen JD, Holmberg LA, Kraft GH, Mayes MD, McDonagh KT, Chen CS, Dipersio J, Lemaistre CF, Pavletic S, Sullivan KM, Sunderhaus J, Furst DE, McSweeney PA. Epstein-Barr virus-associated posttransplantation lymphoproliferative disorder after high-dose immunosuppressive therapy and autologous CD34-selected hematopoietic stem cell transplantation for severe autoimmune diseases. Biol Blood Marrow Transplant 2003; 9:583-91. [PMID: 14506660 PMCID: PMC2956744 DOI: 10.1016/s1083-8791(03)00228-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
High-dose immunosuppressive therapy followed by autologous hematopoietic stem cell transplantation (HSCT) is currently being evaluated for the control of severe autoimmune diseases. The addition of antithymocyte globulin (ATG) to high-dose chemoradiotherapy in the high-dose immunosuppressive therapy regimen and CD34 selection of the autologous graft may induce a higher degree of immunosuppression compared with conventional autologous HSCT for malignant diseases. Patients may be at higher risk of transplant-related complications secondary to the immunosuppressed state, including Epstein-Barr virus (EBV)-associated posttransplantation lymphoproliferative disorder (PTLD), but this is an unusual complication after autologous HSCT. Fifty-six patients (median age, 42 years; range, 23-61 years) with either multiple sclerosis (n = 26) or systemic sclerosis (n = 30) have been treated. The median follow-up has been 24 months (range, 2-60 months). Two patients (multiple sclerosis, n = 1; systemic sclerosis, n = 1) had significant reactivations of herpesvirus infections early after HSCT and then developed aggressive EBV-PTLD and died on days +53 and +64. Multiorgan clonal B-cell infiltrates that were EBV positive by molecular studies or immunohistology were identified at both autopsies. Both patients had positive screening skin tests for equine ATG (Atgam) and had been converted to rabbit ATG (Thymoglobulin) from the first dose. Of the other 54 patients, 2 of whom had partial courses of rabbit ATG because of a reaction to the intravenous infusion of equine ATG, only 1 patient had a significant clinical reactivation of a herpesvirus infection (herpes simplex virus 2) early after HSCT, and none developed EBV-PTLD. The T-cell count in the peripheral blood on day 28 was 0/microL in all 4 patients who received rabbit ATG; this was significantly less than in patients who received equine ATG (median, 174/microL; P =.001; Mann-Whitney ranked sum test). Although the numbers are limited, the time course and similarity of the 2 cases of EBV-PTLD and the effect on day 28 T-cell counts support a relationship between the development of EBV-PTLD and the administration of rabbit ATG. The differences between equine and rabbit ATG are not yet clearly defined, and they should not be considered interchangeable in this regimen without further study.
Collapse
Affiliation(s)
- Richard A Nash
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Rothenberger S, Burns K, Rousseaux M, Tschopp J, Bron C. Ubiquitination of the Epstein-Barr virus-encoded latent membrane protein 1 depends on the integrity of the TRAF binding site. Oncogene 2003; 22:5614-8. [PMID: 12944909 DOI: 10.1038/sj.onc.1206497] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The latent membrane protein 1 (LMP1) encoded by the Epstein-Barr virus functions as a constitutively activated receptor of the tumor necrosis factor receptor family. LMP1 is a short-lived protein that is ubiquitinated and degraded by the proteasome. We have previously shown that LMP1 recruits the adapter protein tumor necrosis factor receptor-associated factor 3 (TRAF3) to lipid rafts. To test if TRAFs are involved in LMP1's ubiquitination, we have mutated the LMP1 CTAR1 site that has been identified as a TRAF binding site. We show that the CTAR1 mutant (CTAR1(-)) is expressed after transfection at a similar level to wild-type LMP1, and behaves as wild-type LMP1 with respect to membrane localization. However, CTAR1(-) does not bind TRAF3. We demonstrate that ubiquitination of CTAR1(-) is significantly reduced when compared to wild-type LMP1. In addition, the expression of wild-type LMP1 induces the ubiquitination, an effect that is significantly reduced when the CTAR1(-) is expressed. Taken together, our results suggest that TRAF proteins are involved in the ubiquitination of LMP1, and that their binding to LMP1 may facilitate their own ubiquitination.
Collapse
Affiliation(s)
- Sylvia Rothenberger
- Institute of Biochemistry, University of Lausanne, ch. des Boveresses 155, 1066 Epalinges, Switzerland.
| | | | | | | | | |
Collapse
|
232
|
Hsieh YH, Wu CJ, Chow KP, Tsai CL, Chang YS. Electroporation-mediated and EBV LMP1-regulated gene therapy in a syngenic mouse tumor model. Cancer Gene Ther 2003; 10:626-36. [PMID: 12872144 DOI: 10.1038/sj.cgt.7700609] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Latent membrane protein 1 (LMP1) is an Epstein-Barr virus (EBV)-encoded oncogene expressed in EBV-associated nasopharyngeal carcinoma (NPC). Previous studies indicate that a strategy combining LMP1-mediated NF-kappaB activation and the HSV thymidine kinase/Ganciclovir (HSVtk/GCV) prodrug system leads to regression of tumor growth in nude mice. To improve the efficacy of this strategy in immunocompetent hosts, we developed a therapeutic cassette, p6kappaB-EDL1E-tk, containing six copies of the NF-kappaB binding motif and an epithelial-specific EBV promoter, ED-L1E. The cassette was tested in a murine CT-26 carcinoma model in syngenic Balb/c mice. Coinjection of an LMP1-expressing vector and p6kappaB-EDL1E-tk by in vivo electroporation in mouse muscle revealed at least two-fold higher TK enzymatic activity than that of previously tested pLTR-tk. Furthermore, growth was attenuated in a group of mice containing LMP1-positive tumors that were intratumorally injected with the p6kappaB-EDL1E-tk cassette and GCV via in vivo electroporation, but not in mice treated with p6kappaB-EDL1E-tk or GCV alone. Similarly, no retardation of tumor growth was observed in mice containing LMP1-negative CT-26 tumors injected with both the p6kappaB-EDL1E-tk cassette and GCV. We propose that intratumoral injection of therapeutic agents, such as DNA of transcription-regulated cassette and GCV, via in vivo electroporation may be used as an alternative treatment for EBV LMP1-expressing cancers.
Collapse
Affiliation(s)
- Yu-hua Hsieh
- Institute of Microbiology and Immunology, National Yang-Ming University, Shih-pai, Taipei 112, Republic of China.
| | | | | | | | | |
Collapse
|
233
|
Dolcetti R, Masucci MG. Epstein-Barr virus: induction and control of cell transformation. J Cell Physiol 2003; 196:207-18. [PMID: 12811813 DOI: 10.1002/jcp.10263] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Epstein-Barr virus (EBV), a ubiquitous human herpes virus, is associated with an increasing number of lymphoid and epithelial malignancies. The ability of the virus to establish life-long persistent infections and induce growth transformation is related to the function of a set of viral proteins that are variously expressed in both normal and malignant cells. Recent evidence indicates that these viral proteins are able to usurp cellular pathways that promote the cell growth and survival, while impairing anti-viral immune responses. Elucidation of the mechanisms by which EBV induces cell transformation and escapes host immune control provides the rational background for the design of new strategies of intervention for EBV-related malignancies.
Collapse
Affiliation(s)
- Riccardo Dolcetti
- Immunovirology and Biotherapy Unit, Department of Pre-Clinical and Epidemiological Research, C.R.O.-I.R.C.C.S., National Cancer Institute, Aviano (PN), Italy.
| | | |
Collapse
|
234
|
Samaniego F, Pati S, Karp JE, Prakash O, Bose D. Human herpesvirus 8 K1-associated nuclear factor-kappa B-dependent promoter activity: role in Kaposi's sarcoma inflammation? J Natl Cancer Inst Monogr 2003:15-23. [PMID: 11158202 DOI: 10.1093/oxfordjournals.jncimonographs.a024252] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The growing number of human immunodeficiency virus type 1 (HIV-1) infections worldwide and the increasing use of immunosuppressive modalities for organ transplantation have contributed to an epidemic of Kaposi's sarcoma (KS), which has been etiologically linked to human herpesvirus 8 (HHV8) or KS-associated virus. Since the onset of the acquired immunodeficiency syndrome epidemic, inflammation has been recognized as an essential component of KS pathology. HHV8 bears a gene (K1) encoding a transmembrane protein with an immunoreceptor tyrosine-based activation motif. This motif is present in receptors that mediate inflammation. PURPOSE To dissect the cellular effects of K1 function and the eventual role of K1 in KS, we developed a cell model for studying K1 expression. METHODS K1 was cloned from BC-3 lymphoma cells. To monitor transcriptional activation, K1 was coexpressed with plasmids containing luciferase under control of various promoters. K1 expression was monitored by indirect immunofluorescence and by combined immunoprecipitation/immunoblot analysis. Inflammatory cytokines were measured by enzyme-linked immunosorbent assay. RESULTS Cellular transfection of the K1 gene induced reporter expression under control of nuclear factor-kappa B (NF-kappaB), which controls the transcription of numerous proteins involved in inflammation. Treatment of cells with aspirin, an agent that targets this intracellular pathway and blocks cell inflammatory responses, blocked K1-induced NF-kappaB-dependent promoter activity. When a second KS cofactor, i.e., the HIV-1-transactivating gene tat, was coexpressed with K1, we observed an additive effect on NF-kappaB-dependent transcription. K1 transfection stimulated the secretion of cytokines interleukin (IL) 6, granulocyte-macrophage colony-stimulating factor, and IL-12. Cells treated with the conditioned media of K1 transfectants exhibited similar characteristics of K1 transfectants, indicating that a paracrine loop was being activated. CONCLUSION Thus, K1 may activate cells in which it is expressed, as well as other cells in a paracrine manner. K1 cooperates in signaling with HIV-1 Tat, suggesting that both of the proteins from these viruses converge to reach an enhanced level of inflammation that may underlie progressive KS.
Collapse
MESH Headings
- Amino Acid Motifs
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Aspirin/pharmacology
- COS Cells
- Chlorocebus aethiops
- Culture Media, Conditioned/pharmacology
- Cytokines/biosynthesis
- Cytokines/metabolism
- Dermatitis/etiology
- Gene Expression Regulation, Viral
- Gene Products, tat/physiology
- Genes, tat
- HIV-1/genetics
- Herpesvirus 8, Human/genetics
- Herpesvirus 8, Human/physiology
- Humans
- Hyperplasia
- Lymphoma, Non-Hodgkin/pathology
- Lymphoma, Non-Hodgkin/virology
- NF-kappa B/physiology
- Neovascularization, Pathologic/etiology
- Neovascularization, Pathologic/physiopathology
- Paracrine Communication
- Promoter Regions, Genetic/genetics
- Recombinant Fusion Proteins/physiology
- Sarcoma, Kaposi/blood supply
- Sarcoma, Kaposi/complications
- Sarcoma, Kaposi/virology
- Skin/pathology
- Skin/virology
- Skin Neoplasms/blood supply
- Skin Neoplasms/complications
- Skin Neoplasms/virology
- Transcriptional Activation
- Transfection
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/virology
- Viral Proteins/chemistry
- Viral Proteins/genetics
- Viral Proteins/physiology
- tat Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- F Samaniego
- Department of Lymphoma/Myeloma and Clinical Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
235
|
Imadome KI, Shirakata M, Shimizu N, Nonoyama S, Yamanashi Y. CD40 ligand is a critical effector of Epstein-Barr virus in host cell survival and transformation. Proc Natl Acad Sci U S A 2003; 100:7836-40. [PMID: 12805559 PMCID: PMC164674 DOI: 10.1073/pnas.1231363100] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Epstein-Barr virus (EBV), implicated in numerous human diseases, including lymphoid malignancies, persistently infects peripheral B cells and transforms them into lymphoblastoid cell lines. Here we found that EBV equally infected B cells from patients with X-linked hyper IgM syndrome and those from healthy donors; however, it hardly transformed X-linked hyper IgM syndrome B cells, because of the dysfunctional gene of CD40 ligand (CD40L) of the patients. Unlike CD40, CD40L is not usually expressed on B cells. However, we found that EBV infection of normal B cells induced CD40L expression as a critical effector in host cell transformation and survival. Moreover, chronic active EBV infection of peripheral T cells, implicated in T cell malignancies, was associated with ectopic expression of CD40, and, in Jurkat T cells, EBV infection induced CD40 expression. These results suggest that EBV infection induces CD40L/CD40 signaling in host cells, which appears to play an essential role in its persistent infection and malignancies of lymphocytes.
Collapse
Affiliation(s)
- Ken-Ichi Imadome
- Department of Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | | | | | | | | |
Collapse
|
236
|
Swaminathan S. Molecular biology of Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus. Semin Hematol 2003. [DOI: 10.1016/s0037-1963(03)70002-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
237
|
Eliopoulos AG, Waites ER, Blake SMS, Davies C, Murray P, Young LS. TRAF1 is a critical regulator of JNK signaling by the TRAF-binding domain of the Epstein-Barr virus-encoded latent infection membrane protein 1 but not CD40. J Virol 2003; 77:1316-28. [PMID: 12502848 PMCID: PMC140818 DOI: 10.1128/jvi.77.2.1316-1328.2003] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The oncogenic Epstein-Barr virus (EBV)-encoded latent infection membrane protein 1 (LMP1) mimics a constitutive active tumor necrosis factor (TNF) family receptor in its ability to recruit TNF receptor-associated factors (TRAFs) and TNF receptor-associated death domain protein (TRADD) in a ligand-independent manner. As a result, LMP1 constitutively engages signaling pathways, such as the JNK and p38 mitogen-activated protein kinases (MAPK), the transcription factor NF-kappaB, and the JAK/STAT cascade, and these activities may explain many of its pleiotropic effects on cell phenotype, growth, and transformation. In this study we demonstrate the ability of the TRAF-binding domain of LMP1 to signal on the JNK/AP-1 axis in a cell type- dependent manner that critically involves TRAF1 and TRAF2. Thus, expression of this LMP1 domain in TRAF1-positive lymphoma cells promotes significant JNK activation, which is blocked by dominant-negative TRAF2 but not TRAF5. However, TRAF1 is absent in many established epithelial cell lines and primary nasopharyngeal carcinoma (NPC) biopsy specimens. In these cells, JNK activation by the TRAF-binding domain of LMP1 depends on the reconstitution of TRAF1 expression. The critical role of TRAF1 in the regulation of TRAF2-dependent JNK signaling is particular to the TRAF-binding domain of LMP1, since a homologous region in the cytoplasmic tail of CD40 or the TRADD-interacting domain of LMP1 signal on the JNK axis independently of TRAF1 status. These data further dissect the signaling components used by LMP1 and identify a novel role for TRAF1 as a modulator of oncogenic signals.
Collapse
Affiliation(s)
- Aristides G Eliopoulos
- Cancer Research UK Institute for Cancer Studies, The University of Birmingham Medical School, United Kingdom.
| | | | | | | | | | | |
Collapse
|
238
|
Middeldorp JM, Brink AATP, van den Brule AJC, Meijer CJLM. Pathogenic roles for Epstein-Barr virus (EBV) gene products in EBV-associated proliferative disorders. Crit Rev Oncol Hematol 2003; 45:1-36. [PMID: 12482570 DOI: 10.1016/s1040-8428(02)00078-1] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Epstein-Barr virus (EBV) is associated with a still growing spectrum of clinical disorders, ranging from acute and chronic inflammatory diseases to lymphoid and epithelial malignancies. Based on a combination of in vitro and in vivo findings, EBV is thought to contribute in the pathogenesis of these diseases. The different EBV gene expression patterns in the various disorders, suggest different EBV-mediated pathogenic mechanisms. In the following pages, an overview of the biology of EBV-infection is given and functional aspects of EBV-proteins are discussed and their putative role in the various EBV-associated disorders is described. EBV gene expression patterns and possible pathogenic mechanisms are discussed. In addition, expression of the cellular genes upregulated by EBV in vitro is discussed, and a comparison with the in vivo situation is made.
Collapse
Affiliation(s)
- Jaap M Middeldorp
- Department of Pathology, Vrije Universiteit Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
239
|
Abstract
CD40 is an important regulator of diverse aspects of the immune response including the T-cell-dependent humoral immune response, the development of antigen-presenting cells (APCs) and inflammation. Latent membrane protein 1 (LMP1), a protein encoded by Epstein-Barr Virus (EBV), appears to mimic CD40 in multiple ways. CD40 and LMP1 bind similar sets of cellular signalling proteins and activate overlapping signalling pathways. Despite many similarities shared between CD40 and LMP1, they also differ substantively. In this review, we will compare and contrast the signalling mediated by CD40 and LMP1.
Collapse
Affiliation(s)
- Ngan Lam
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, 1400 University Avenue, Madison, WI 53713, USA
| | | |
Collapse
|
240
|
Chan BCL, To KF, Pang JCS, Chung YF, Lo KW, Tong JHM, Huang DWS, Lim PL, Chui YL. Generation of monoclonal antibodies against Hong Kong nasopharyngeal carcinoma-associated Epstein-Barr virus latent membrane protein 1 (LMP1). Int J Cancer 2002; 102:492-8. [PMID: 12432552 DOI: 10.1002/ijc.10773] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A panel of monoclonal antibodies specific to Hong Kong Chinese nasopharyngeal carcinoma (NPC)-associated Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) variants has been generated. These monoclonal antibodies not only differentiate the Hong Kong Chinese NPC-associated LMP1 variants from the prototype B95-8 LMP1, derived from Caucasian infectious mononucleosis, but also differentiate the 2 highly homologous LMP1 deletion variants commonly found in Hong Kong primary NPC. The predominant deletion type variant, DV-Asp335, is characterized by an aspartic acid at residue 335 located in the cytoplasmic C-terminal region, whereas the other minor deletion variant, DV-Gly335, has a glycine in the same residue position. 335D is hitherto found predominantly in LMP1 of the China 1 strain in association with NPC in the Chinese populations located in southern China and Malaysia. These antibodies, which are applicable in ELISA, immunofluorescence, immunoprecipitation, immunoblotting and immunohistochemistry on paraffin sections, are the first variant-specific anti-LMP1 monoclonal antibodies produced, and will be useful in investigating the functional significance of 335D in NPC.
Collapse
Affiliation(s)
- Ben Chung Lap Chan
- Clinical Immunology Unit and Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Vockerodt M, Belge G, Kube D, Irsch J, Siebert R, Tesch H, Diehl V, Wolf J, Bullerdiek J, Staratschek-Jox A. An unbalanced translocation involving chromosome 14 is the probable cause for loss of potentially functional rearranged immunoglobulin heavy chain genes in the Epstein-Barr virus-positive Hodgkin's lymphoma-derived cell line L591. Br J Haematol 2002; 119:640-6. [PMID: 12437638 DOI: 10.1046/j.1365-2141.2002.03894.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the vast majority of cases, Hodgkin-Reed Sternberg (H-RS) cells, the malignant cells in Hodgkin's lymphoma (HL), are derived from germinal centre B cells. In some cases, somatic mutations within the rearranged immunoglobulin heavy (IgH) chain genes were detected, rendering potentially functional gene rearrangements non-functional. In these H-RS cells the expression of high-affinity B-cell receptors (BCR) was prevented. As in other cases only one non-productive IgH chain gene rearrangement was amplified from H-RS cells, it was speculated whether, in these cases, the functionally rearranged IgH chain genes were lost. An alternative explanation might be that the rearranged genes could not be amplified owing to a high load of somatic mutations within the primer binding sites. Here, we showed that, in the HL-derived Epstein-Barr virus (EBV)-positive cell line L591, only one non-functional somatically mutated IgH gene rearrangement could be detected. The other potentially functional IgH gene rearrangement was lost as a result of an unbalanced translocation affecting the long arm of chromosome 14. Moreover, L591 cells express the EBV latent membrane proteins LMP1 and LMP2A, which might have contributed to the 'escape' of these cells from apoptosis within the germinal centre. We conclude that, apart from the introduction of 'crippling mutations' into the rearranged VDJ genes rearrangement, deletions of the IGH locus may be regarded as another mechanism to prevent the expression of a BCR in H-RS cells.
Collapse
Affiliation(s)
- Martina Vockerodt
- Department of Internal Medicine I, Universitity of Cologne, Cologne, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Park JH, Faller DV. Epstein-Barr virus latent membrane protein-1 induction by histone deacetylase inhibitors mediates induction of intercellular adhesion molecule-1 expression and homotypic aggregation. Virology 2002; 303:345-63. [PMID: 12490396 DOI: 10.1006/viro.2002.1638] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Epstein-Barr virus (EBV) latent membrane protein (LMP)-1 induces B lymphocyte immortalization and activates constitutive signal transduction, including NF-kappaB, JNK/p38, and JAK/STAT pathways. During EBV latency, LMP-1 expression induces several B lymphocyte activation markers, including intercellular adhesion molecule (ICAM)-1. We found that various structurally distinct histone deacetylase inhibitors (HDACI), as well as phorbol ester treatment, induced homotypic aggregation in EBV-positive Burkitt's lymphoma lines. Cell-surface expression of ICAM-1 was concurrently strongly up-regulated by both HDACI and phorbol ester treatments. Cell-surface expression of ICAM-1 was concurrently strongly induced by both HDACI and phorbol ester treatment. Among several ICAM family members, only ICAM-1 was up-regulated by both HDACI and phorbol ester treatments, suggesting that up-regulated ICAM-1 expression might mediate the observed increase in homotypic aggregation. HDACI-induced homotypic aggregation was blocked by exposure to a monoclonal antibody specific for the beta-chain (CD18) of an ICAM-1 ligand, LFA-1. Unexpectedly, HDAC inhibition, but not phorbol ester treatment, induced LMP-1 expression in EBV-positive cell lines, and this LMP-1 species was identified by RT-PCR and immunoblot analyses as the latent form of LMP-1. Control of EBV LMP-1 gene expression by HDACI inhibition occurs at the transcriptional level, as indicated by nuclear runoff studies and analysis of steady-state mRNA levels. Dominant-negative LMP-1 efficiently blocked HDACI-induced ICAM-1 up-regulation, and ectopic expression of LMP-1 activated expression of an ICAM-1 promoter-driven reporter gene. The HDACI-induced up-regulation of ICAM-1, and consequent homotypic aggregation, were efficiently blocked by the addition of N-acetyl-L-cysteine and by ectopic expression of a super-repressor IkappaBalpha, while LPM-1 induction was unaffected, suggesting that these effects are mediated by NF-kappaB. We demonstrate, therefore, that the latent isoform of LMP-1 is induced by HDAC inhibition, and that HDACI-induced latent LMP-1 expression, through NF-kappaB activation, is responsible for ICAM-1 expression up-regulation and homotypic adhesion.
Collapse
Affiliation(s)
- Jae Hong Park
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | |
Collapse
|
243
|
Yu JS, Tsai HC, Wu CC, Weng LP, Li HP, Chung PJ, Chang YS. Induction of inducible nitric oxide synthase by Epstein-Barr virus B95-8-derived LMP1 in Balb/3T3 cells promotes stress-induced cell death and impairs LMP1-mediated transformation. Oncogene 2002; 21:8047-61. [PMID: 12439755 DOI: 10.1038/sj.onc.1205990] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2001] [Revised: 08/20/2002] [Accepted: 08/20/2002] [Indexed: 11/09/2022]
Abstract
The latent membrane protein 1 (LMP1) of Epstein-Barr virus (EBV) causes cellular transformation and activation of several intracellular signaling events. In this report, we show that BLMP1 (encoded by the LMP1 gene derived from the B95-8 strain of EBV) triggers the expression of inducible nitric oxide synthase (iNOS) in Balb/3T3 fibroblasts. Intriguingly, NLMP1, a natural sequence variant of LMP1 identified in EBV-positive nasopharyngeal carcinoma biopsy, does not similarly induce iNOS expression. BLMP1-induced iNOS in Balb/3T3 cells is active to produce nitric oxide (NO), and NO production can be blocked by several iNOS inhibitors. When subjected to environmental stress, Balb/3T3 cells that produce NO lose viability more rapidly than non NO-producing cells. Blockage of NO generation by iNOS inhibitors enhances the viability of NO-producing cells under stress conditions. The activities of caspase-3 and c-Jun N-terminal kinase, two important regulators mediating stress-induced apoptosis, are significantly potentiated following heat shock treatment of BLMP1-expressing/NO-producing cells, compared to parental and NLMP1-expressing cells. Furthermore, treatment with iNOS inhibitor augmented the cloning efficiency (in culture) and tumor growth (in nude mice) of BLMP1-expressing/NO-producing cells. Collectively, the results demonstrate that BLMP1 induces iNOS expression and NO production in Balb/3T3 cells, which leads to the alteration of cell functions, including sensitivity to environmental stress, capability to colonize independent of anchorage and tumorigenicity in nude mice. Our data additionally implicate that the differential iNOS induction potential of the two LMP1 forms may represent the basis of a functional difference between the two LMP1 proteins.
Collapse
Affiliation(s)
- Jau-Song Yu
- Department of Cell and Molecular Biology, Institute of Basic Medical Science, Medical College of Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
| | | | | | | | | | | | | |
Collapse
|
244
|
Chung PJ, Chang YS, Liang CL, Meng CL. Negative regulation of Epstein-Barr virus latent membrane protein 1-mediated functions by the bone morphogenetic protein receptor IA-binding protein, BRAM1. J Biol Chem 2002; 277:39850-7. [PMID: 12181323 DOI: 10.1074/jbc.m206736200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The latent membrane protein 1 (LMP1) of Epstein-Barr virus causes cellular transformation and activates several intracellular signals, including NF-kappaB and c-Jun N-terminal kinase. Using yeast two-hybrid screening with the LMP1 C-terminal sequence as bait, we demonstrate that BRAM1 (bone morphogenetic protein receptor-associated molecule 1) is an LMP1-interacting protein. BRAM1 associates with LMP1, both in vitro and in vivo, as revealed by confocal microscopy, glutathione S-transferase pull-down, and co-immunoprecipitation assays. This association mainly involves the C-terminal half of BRAM1 comprising the MYND domain and the CTAR2 region of LMP1, which is critical in LMP1-mediated signaling pathways. We show that BRAM1 interferes with LMP1-mediated NF-kappaB activation but not the JNK signaling pathway. Because the CTAR2 region interacts with the tumor necrosis factor (TNF-alpha receptor-associated death domain protein, it is interesting to find that BRAM1 also interferes with NF-kappaB activation mediated by TNF-alpha. BRAM1 interferes LMP1-mediated and TNF-alpha-induced NF-kappaB activation by targeting IkappaBalpha molecules. Moreover, BRAM1 inhibits the resistance of LMP1-expressing cells to TNF-alpha-induced cytotoxicity. We therefore propose that the BRAM1 molecule associates with LMP1 and functions as a negative regulator of LMP1-mediated biological functions.
Collapse
Affiliation(s)
- Pei-Jung Chung
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | | | | | | |
Collapse
|
245
|
Abstract
The B-lymphotropic virus Epstein-Barr virus (EBV) has been implicated in the pathogenesis of B-cell malignancies, particularly in immunodeficient individuals. This review provides a brief overview of the EBV-encoded proteins involved in B-cell transformation, and the current state of knowledge about their roles in this process.
Collapse
Affiliation(s)
- Gail A Bishop
- Department of Microbiology, 3-570 Bowen Science Bldg., The University of Iowa, Iowa City, IA 52242, USA.
| | | |
Collapse
|
246
|
Chi LM, Yu JS, Chang YS. Identification of protein kinase CK2 as a potent kinase of Epstein-Barr virus latent membrane protein 1. Biochem Biophys Res Commun 2002; 294:586-91. [PMID: 12056807 DOI: 10.1016/s0006-291x(02)00515-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The C-terminus of latent membrane protein 1 (LMP1) can be phosphorylated in vivo. However, the protein kinase responsible for LMP1 phosphorylation has not yet been identified. In this study, GST fusion proteins containing the C-terminus of LMP1 were generated and used as substrates to survey the kinases that phosphorylate LMP1. Among several purified protein kinases tested, only protein kinase CK2 (CK2) could specifically phosphorylate LMP1. Using the in-gel kinase assay in the absence and presence of a selective CK2 inhibitor, 4,5,6,7-tetrabromobenzotriazole, CK2 was determined to be the major kinase to phosphorylate LMP1 in lymphoma and epithelial cell lines. This is the first study to show that CK2 is a potent kinase to phosphorylate LMP1 in vitro.
Collapse
Affiliation(s)
- Lang-Ming Chi
- Department of Medical Technology, Yuan-Pei Institute of Science and Technology, Hsinchu, Taiwan, ROC.
| | | | | |
Collapse
|
247
|
Masciarelli S, Mattioli B, Galletti R, Samoggia P, Chichiarelli S, Mearini G, Mattia E. Antisense to Epstein Barr Virus-encoded LMP1 does not affect the transcription of viral and cellular proliferation-related genes, but induces phenotypic effects on EBV-transformed B lymphocytes. Oncogene 2002; 21:4166-70. [PMID: 12037673 DOI: 10.1038/sj.onc.1205515] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2001] [Revised: 03/01/2002] [Accepted: 03/22/2002] [Indexed: 11/08/2022]
Abstract
It is generally accepted that Epstein-Barr virus (EBV) latent genes EBNA-2, EBNA-3A, -3C, EBNA-LP and LMP1 are essential for growth transformation and immortalization of B lymphocytes. Among these genes, LMP1 plays a key role in the survival and dissemination of the infected B cells by inducing anti-apoptotic genes and surface expression of several activation antigens and adhesion molecules. We have previously shown that antisense oligodeoxynucleotides directed to LMP1 mRNA, effectively suppress LMP1 gene expression and substantially reduce B95.8 cell proliferation. In this study, we have used antisense LMP1 oligomers to investigate whether LMP1 suppression might influence the expression of latent EBV genes with oncogenic potential, anti-apoptotic genes, or affect the phenotype of EBV-infected B95.8 cells. Our data show that LMP1 suppression does not affect the transcription of EBNA-2, EBNA-3A, -3B and -3C genes, or that of bcl-2 and mcl-1 anti-apoptotic genes. In contrast, consistent modifications in the expression of CD39, CD54, CD23, CD11 and CD10 molecules were observed in B95.8 cells after treatment with antisense LMP1. Our findings support the possibility for using LMP1 antisense oligomers as therapeutics in EBV-associated tumors.
Collapse
Affiliation(s)
- Silvia Masciarelli
- Department of Public Health Sciences, Medical Faculty, University of Rome La Sapienza, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
248
|
Xue SA, Labrecque LG, Lu QL, Ong SK, Lampert IA, Kazembe P, Molyneux E, Broadhead RL, Borgstein E, Griffin BE. Promiscuous expression of Epstein-Barr virus genes in Burkitt's lymphoma from the central African country Malawi. Int J Cancer 2002; 99:635-43. [PMID: 12115495 DOI: 10.1002/ijc.10372] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Primary BL in Malawian children has a very high frequency association, approaching 100%, with the human herpesvirus EBV. A detailed study carried out on viral gene expression in these tumours, using both fresh material and methanol-fixed FNAs, showed, contrary to prediction, that most belong to a variant "class II" latency category, with lytic cycle-related genes also expressed. That is, in addition to EBNA1 expression, membrane proteins (LMP1/2A), immediate early (BZLF1) and early (IR2 and IR4) genes, a putative viral oncogene (BARF1), CST (BART) antisense transcripts and the viral bcl-2 homologue are expressed in a high proportion of the BLs. Most, but not all, express the small viral (EBER) RNAs. Two other significant observations were made: (i) in addition to expression of cellular cytokine (IL-10) transcripts in all tumours investigated, the normally silent viral IL-10 homologue was expressed in some tumours; (ii) whereas EBNA1 expression from its restricted Qp promoter was generally observed, the nonrestricted Cp/Wp promoter was also active in some tumours. Viral gene expression in the Malawian [endemic (e)] BLs appears to be more promiscuous than predicted from other studies, but expression accords with the cytopathologic picture of eBLs as a rapidly proliferating cell population accompanied by considerable necrosis, and a clinically diverse disease. A small-scale study of relapse Malawian BLs revealed a different picture of viral association, more akin to systemic BL than eBL, where EBV appears to be absent or present only at very low levels. The significance of these findings is considered.
Collapse
Affiliation(s)
- Shao-an Xue
- Viral Oncology Unit, Faculty of Medicine, Imperial College of Science, Technology and Medicine, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Xu J, Ahmad A, Menezes J. Preferential localization of the Epstein-Barr virus (EBV) oncoprotein LMP-1 to nuclei in human T cells: implications for its role in the development of EBV genome-positive T-cell lymphomas. J Virol 2002; 76:4080-6. [PMID: 11907247 PMCID: PMC136072 DOI: 10.1128/jvi.76.8.4080-4086.2002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2001] [Accepted: 01/16/2002] [Indexed: 11/20/2022] Open
Abstract
The Epstein-Barr virus (EBV)-encoded latent membrane protein-1 (LMP-1) is thought to play a role in the EBV-induced B-cell transformation and immortalization. EBV has also been implicated in certain human T-cell lymphomas; however, the phenotypic effects of the expression of this oncoprotein in T cells are not known. To learn whether LMP-1 also induces phenotypic changes in T cells, we stably expressed it in human cell lines of T and B lineages and 25 LMP-1-expressing T-cell clones and 7 B-cell clones were examined. Our results show for the first time that, in sharp contrast to B cells, LMP-1 preferentially localizes to nuclei in T cells and does not induce the phenotypic changes in these cells that it induces in B cells, does not associate with TRAF proteins, and does not arrest the cell cycle in the G2/M phase. A computer-assisted analysis revealed that LMP-1 lacks the canonical nuclear localization signal. Our results suggest that this oncoprotein may not play the same role in the lymphomagenesis of T cells as it does in B cells.
Collapse
Affiliation(s)
- Jingwu Xu
- Laboratory of Immunovirology, Department of Microbiology and Immunology, University of Montreal and Ste-Justine Hospital, Montreal, Quebec, Canada H3T 1C5.
| | | | | |
Collapse
|
250
|
Prokova V, Mosialos G, Kardassis D. Inhibition of transforming growth factor beta signaling and Smad-dependent activation of transcription by the Latent Membrane Protein 1 of Epstein-Barr virus. J Biol Chem 2002; 277:9342-50. [PMID: 11781310 DOI: 10.1074/jbc.m109099200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Inhibition of transforming growth factor beta (TGFbeta) signaling by the Epstein-Barr virus Latent Membrane Protein 1 (LMP1) may account, at least in part, for the oncogenic activity of LMP1. We found that LMP1 is a potent inhibitor of TGFbeta signaling and Smad-dependent activation of transcription in 293 epithelial cells and COS-7 fibroblasts. LMP1 strongly inhibited the uninduced and the Smad-inducible activity of the promoters of the human p21/WAF1/Cip1 gene and the mouse Smad7 gene. Inhibition of TGFbeta signaling and Smad-dependent activation of transcription by LMP1 was greatly reduced by deletion of both C-terminal activating regions 1 and 2 of LMP1 as well as by overexpression of a non-degradable form of IkappaB. In contrast, specific inhibitors of p38 kinase or MEK kinase did not reverse the inhibitory activity of LMP1. TGFbeta signaling was enhanced by overexpression of dominant negative forms of the LMP1 effectors TRAF2, NIK, and IKKbeta and was abolished by overexpression of p65/RelA or a p50/p65 fusion protein. Deletion of the transactivation domain of p65 abolished its inhibitory activity. Immunoblotting and immunofluorescence microscopy indicated that suppression of TGFbeta signaling and Smad transcriptional activity by LMP1 was not due to Smad degradation or cytoplasmic retention suggesting that LMP1 affects the nuclear function of Smad proteins. Our data are consistent with an essential role of NF-kappaB activation by LMP1 in the inhibition of TGFbeta signaling and Smad-mediated transcriptional responses.
Collapse
Affiliation(s)
- Vassiliki Prokova
- Department of Basic Sciences, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Heraklion GR-71110, Greece
| | | | | |
Collapse
|