201
|
NF-κB signaling modulation by EBV and KSHV. Trends Microbiol 2010; 18:248-57. [DOI: 10.1016/j.tim.2010.04.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 03/24/2010] [Accepted: 04/02/2010] [Indexed: 12/12/2022]
|
202
|
A role for protein kinase PKR in the mediation of Epstein–Barr virus latent membrane protein-1-induced IL-6 and IL-10 expression. Cytokine 2010; 50:210-9. [DOI: 10.1016/j.cyto.2010.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 12/10/2009] [Accepted: 01/20/2010] [Indexed: 01/29/2023]
|
203
|
Chen CC, Chen LC, Liang Y, Tsang NM, Chang YS. Epstein-Barr virus latent membrane protein 1 induces the chemotherapeutic target, thymidine phosphorylase, via NF-kappaB and p38 MAPK pathways. Cell Signal 2010; 22:1132-42. [PMID: 20214978 DOI: 10.1016/j.cellsig.2010.03.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 03/01/2010] [Indexed: 11/19/2022]
Abstract
High thymidine phosphorylase (TP) expression is significantly correlated with poor prognosis in patients with nasopharyngeal carcinoma (NPC). NPC is an Epstein-Barr Virus (EBV)-associated cancer in which the EBV-encoded oncogene product, latent membrane protein 1 (LMP1), is expressed in approximately 60% of tumor tissues. However, no previous study has examined whether LMP1 is involved in up-regulating TP expression in NPC tissues. We herein show that LMP1 expression is correlated with TP expression in tumor cells, as examined by quantitative RT-PCR and immunohistochemical staining. We further show that the CTAR1 and CTAR2 domains of LMP1 mediate TP induction, as demonstrated by quantitative RT-PCR and Western blot analyses using LMP1 deletion and site-specific mutants. Mechanistically, LMP1-mediated TP induction is abolished by inhibitors of NF-kappaB and p38 MAPK, dominant-negative IkappaB and p38, and siRNA-mediated knockdown of p38 MAPK. Clinically, there were significant correlations among the expression levels of TP, activated p65, and phospho-p38 MAPK in NPC biopsy samples. Functionally, LMP1-mediated induction of TP expression enhanced the sensitivity of NPC cells to the chemotherapeutic prodrug, 5'-DFUR. Our results provide new insights into the roles of LMP1-mediated NF-kappaB and p38 MAPK signaling pathways in TP induction, potentially suggesting new therapeutic strategies for the treatment of NPC.
Collapse
Affiliation(s)
- Chia-Chun Chen
- Graduate Institute of Biomedical Sciences, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Taoyuan 333, Taiwan.
| | | | | | | | | |
Collapse
|
204
|
Dual functions of interferon regulatory factors 7C in Epstein-Barr virus-mediated transformation of human B lymphocytes. PLoS One 2010; 5:e9459. [PMID: 20209099 PMCID: PMC2831998 DOI: 10.1371/journal.pone.0009459] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 02/07/2010] [Indexed: 11/19/2022] Open
Abstract
Epstein-Barr virus (EBV) infection is associated with several human malignancies. Interferon (IFN) regulatory factor 7 (IRF-7) has several splicing variants, and at least the major splicing variant (IRF-7A) has oncogenic potential and is associated with EBV transformation processes. IRF-7C is an alternative splicing variant with only the DNA-binding domain of IRF-7. Whether IRF-7C is present under physiological conditions and its functions in viral transformation are unknown. In this report, we prove the existence of IRF-7C protein and RNA in certain cells under physiological conditions, and find that high levels of IRF-7C are associated with EBV transformation of human primary B cells in vitro as well as EBV type III latency. EBV latent membrane protein 1 (LMP-1) stimulates IRF-7C expression in B lymphocytes. IRF-7C has oncogenic potential in rodent cells and partially restores the growth properties of EBV-transformed cells under a growth-inhibition condition. A tumor array experiment has identified six primary tumor specimens with high levels of IRF-7C protein—all of them are lymphomas. Furthermore, we show that the expression of IRF-7C is apparently closely associated with other IRF-7 splicing variants. IRF-7C inhibits the function of IRF-7 in transcriptional regulation of IFN genes. These data suggest that EBV may use splicing variants of IRF-7 for its transformation process in two strategies: to use oncogenic properties of various IRF-7 splicing variants, but use one of its splicing variants (IRF-7C) to block the IFN-induction function of IRF-7 that is detrimental for viral transformation. The work provides a novel relation of host/virus interactions, and has expanded our knowledge about IRFs in EBV transformation.
Collapse
|
205
|
Bieging KT, Swanson-Mungerson M, Amick AC, Longnecker R. Epstein-Barr virus in Burkitt's lymphoma: a role for latent membrane protein 2A. Cell Cycle 2010; 9:901-8. [PMID: 20160479 PMCID: PMC2855765 DOI: 10.4161/cc.9.5.10840] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Burkitt's lymphoma (BL) is characterized by translocation of the MYC gene to an immunoglobulin locus. Transgenic mouse models have been used to study the molecular changes that are necessary to bypass tumor suppression in the presence of translocated MYC. Inactivation of the p53 pathway is a major step to tumor formation in mouse models that is also seen in human disease. Human BL is often highly associated with Epstein-Barr virus (EBV). The EBV latency protein latent membrane protein 2A (LMP2A) is known to promote B cell survival by affecting levels of pro-survival factors. Using LMP2A transgenic mouse models, we have identified a novel mechanism that permits lymphomagenesis in the presence of an intact p53 pathway. This work uncovers a contribution of EBV to molecular events that have documented importance in BL pathogenesis, and may underlie the poorly understood link between EBV and BL.
Collapse
Affiliation(s)
- Kathryn T. Bieging
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611 USA
| | - Michelle Swanson-Mungerson
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois 60515 USA
| | - Alexandra C. Amick
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611 USA
| | - Richard Longnecker
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611 USA
| |
Collapse
|
206
|
Morris MA, Dawson CW, Young LS. Role of the Epstein-Barr virus-encoded latent membrane protein-1, LMP1, in the pathogenesis of nasopharyngeal carcinoma. Future Oncol 2010; 5:811-25. [PMID: 19663731 DOI: 10.2217/fon.09.53] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although frequently expressed in Epstein-Barr virus (EBV)-positive malignancies, the contribution of the oncogenic latent membrane protein-1 (LMP1) to the pathogenesis of nasopharyngeal carcinoma remains to be fully defined. As a key effector in EBV-driven B-cell transformation in vitro, LMP1 also displays oncogenic properties in rodent fibroblasts, and exhibits similar effects in epithelial cells. LMP1 functions as a viral mimic of the TNFR family member, CD40, engaging a plethora of signaling pathways including: NF-kappaB, JNK/p38 (SAPK), PI3-kinase and ERK-MPK. The constitutive activation of these pathways appears central in the ability of LMP1 to induce multiple morphological and phenotypic alterations. Here we review the effects of LMP1 on epithelial cell growth transformation, and its putative role in the pathogenesis of nasopharyngeal carcinoma, focusing on key areas of proliferation, survival, cell motility and invasion.
Collapse
Affiliation(s)
- Mhairi A Morris
- Cancer Research UK Institute for Cancer Studies, School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | | | | |
Collapse
|
207
|
Najjar I, Fagard R. STAT1 and pathogens, not a friendly relationship. Biochimie 2010; 92:425-44. [PMID: 20159032 PMCID: PMC7117016 DOI: 10.1016/j.biochi.2010.02.009] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 02/09/2010] [Indexed: 12/21/2022]
Abstract
STAT1 belongs to the STAT family of transcription factors, which comprises seven factors: STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B and STAT6. STAT1 is a 91 kDa protein originally identified as the mediator of the cellular response to interferon (IFN) α, and thereafter found to be a major component of the cellular response to IFNγ. STAT1 is, in fact, involved in the response to several cytokines and to growth factors. It is activated by cytokine receptors via kinases of the JAK family. STAT1 becomes phosphorylated and forms a dimer which enters the nucleus and triggers the transcription of its targets. Although not lethal at birth, selective gene deletion of STAT1 in mice leads to rapid death from severe infections, demonstrating its major role in the response to pathogens. Similarly, in humans who do not express STAT1, there is a lack of resistance to pathogens leading to premature death. This indicates a key, non-redundant function of STAT1 in the defence against pathogens. Thus, to successfully infect organisms, bacterial, viral or parasitic pathogens must overcome the activity of STAT1, and almost all the steps of this pathway can be blocked or inhibited by proteins produced in infected cells. Interestingly, some pathogens, like the oncogenic Epstein–Barr virus, have evolved a strategy which uses STAT1 activation.
Collapse
Affiliation(s)
- Imen Najjar
- INSERM Unité 978, SMBH, 74 rue Marcel Cachin, Bobigny-cedex 93017, France.
| | | |
Collapse
|
208
|
Lu JH, Tang YL, Yu HB, Zhou JH, Fu CY, Zeng X, Yu ZY, Yin HL, Wu MH, Zhang JY, Li XL, Li GY. Epstein-Barr virus facilitates the malignant potential of immortalized epithelial cells: from latent genome to viral production and maintenance. J Transl Med 2010; 90:196-209. [PMID: 19997065 DOI: 10.1038/labinvest.2009.130] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Epstein-Barr virus (EBV) is closely associated with several malignancies, including nasopharyngeal carcinoma. To investigate the EBV activity in tumor development, we tried to establish a malignant model of EBV-infected cells in nude mice. On the basis of the Maxi-EBV system, a human embryonic kidney epithelial cell line (293) with a low malignant potential was used for a stable EBV genome infection. The derived cell line, termed 293-EBV, exhibited obvious morphological transformation and significantly increased growth ability, with the cell cycle redistributed. The clonability and tumorigenicity were also substantially accelerated. In 293-EBV cells, the expression level of the transcription factor NF-kappaB and JNK2 were upregulated. The result suggested that latent membrane protein 1 (LMP1) was an important viral protein responsible for the enhanced malignant potential. Matured and budding virus particles were observed in tumor tissues, confirming the spontaneous reactivation of EBV from latent genome to lytic cycle at the site of tumor development. Primary culture of tumor tissues showed two patterns about the EBV maintenance or not in newly grown cells, and this was dependent on the thickness of the planted tissues. Moreover, the tumor cells lost EBV genome easily when subcultured at low density. Our findings revealed the cell-to-cell contact mechanism, which was required for the EBV maintenance in the tumor cells during the expansion of EBV-infected cells. This mechanism might give an explanation to the phenomenon that EBV genome in epithelial tumor cells becomes easily lost during subculture in vitro. Our results provided further evidence of a function for EBV in the etiology of tumor development.
Collapse
Affiliation(s)
- Jian-Hong Lu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan 410078, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Bibas M, Antinori A. EBV and HIV-Related Lymphoma. Mediterr J Hematol Infect Dis 2009; 1:e2009032. [PMID: 21416008 PMCID: PMC3033170 DOI: 10.4084/mjhid.2009.032] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 12/27/2009] [Indexed: 11/18/2022] Open
Abstract
HIV-associated lymphoproliferative disorders represent a heterogeneous group of diseases, arising in the presence of HIV-associated immunodeficiency. The overall prevalence of HIV-associated lymphoma is significantly higher compared to that of the general population and it continues to be relevant even after the wide availability of highly active antiretroviral therapy (HAART) (1). Moreover, they still represent one of the most frequent cause of death in HIV-infected patients. Epstein-Barr virus (EBV), a γ-Herpesviruses, is involved in human lymphomagenesis, particularly in HIV immunocompromised patients. It has been largely implicated in the development of B-cell lymphoproliferative disorders as Burkitt lymphoma (BL), Hodgkin disease (HD), systemic non Hodgkin lymphoma (NHL), primary central nervous system lymphoma (PCNSL), nasopharyngeal carcinoma (NC). Virus-associated lymphomas are becoming of significant concern for the mortality of long-lived HIV immunocompromised patients, and therefore, research of advanced strategies for AIDS-related lymphomas is an important field in cancer chemotherapy. Detailed understanding of the EBV lifecycle and related cancers at the molecular level is required for novel strategies of molecular-targeted cancer chemotherapy The linkage of HIV-related lymphoma with EBV infection of the tumor clone has several pathogenetic, prognostic and possibly therapeutic implications which are reviewed herein.
Collapse
Affiliation(s)
- Michele Bibas
- Clinical Department, National Institute for Infectious Diseases “Lazzaro Spallanzani”, IRCCS, Rome, Italy
| | - Andrea Antinori
- Clinical Department, National Institute for Infectious Diseases “Lazzaro Spallanzani”, IRCCS, Rome, Italy
| |
Collapse
|
210
|
Abstract
Epstein-Barr virus (EBV) encoded latent membrane protein 1 (LMP1) is noted for its transforming potential. Yet, it also acts as a cytostatic and growth-relenting factor in Burkitt's lymphoma (BL) cells. The underlying molecular mechanisms of the growth inhibitory property of LMP1 have remained largely unknown. In this study, we show that LMP1 negatively regulates a major oncogene, TCL1, in diffuse large B-cell lymphoma (DLBCL) and BL cells. MicroRNA (miR) profiling of LMP1 transfectants showed that among others, miR-29b, is upregulated. LMP1 diminished TCL1 by inducing miR-29b through C-terminus activation region 1 (CTAR1) and CTAR2. miR-29b locked nucleic acid (LNA) antisense oligonucleotide transfection into LMP1-expressing cells reduced miR-29b expression and consequently reconstituted TCL1, suggesting that LMP1 negatively regulates TCL1 through miR-29b upregulation. The miR-29b increase by LMP1 was due to an increase in the cluster pri-miR-29b1-a transcription, derived from human chromosome 7. Using pharmacological inhibitors, we found that p38 mitogen-activated protein kinase-activating function of LMP1 is important for this effect. The ability of LMP1 to negatively regulate TCL1 through miR-29b might underlie its B-cell lymphoma growth antagonistic property. As LMP1 is also important for B-cell transformation, we suggest that the functional dichotomy of this viral protein may depend on a combination of levels of its expression, lineage and differentiation of the target cells and regulation of miRs, which then directs the outcome of the cellular response.
Collapse
|
211
|
Synergism of BARF1 with Ras induces malignant transformation in primary primate epithelial cells and human nasopharyngeal epithelial cells. Neoplasia 2009; 11:964-73. [PMID: 19724690 DOI: 10.1593/neo.09706] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2009] [Revised: 06/13/2009] [Accepted: 06/15/2009] [Indexed: 12/26/2022] Open
Abstract
Although it is well known that nasopharyngeal carcinoma (NPC) is closely related with Epstein-Barr virus (EBV), few data are available about which and how EBV-expressed gene is involved in the carcinogenesis of human nasopharyngeal epithelial cells. EBV-encoded BARF1 (BamH I-A right frame 1) gene has been shown to be oncogenic and capable of inducing malignant transformation in BALB/c3T3 and NIH3T3 cells as well as in human B-cell lines Louckes and Akata. It remains unclear, however, whether BARF1 can transform primate or human epithelial cells. Here, we have shown that overexpression of H-Ras gene transformed BARF1-immortalized PATAS cells into malignant cell line. Furthermore, we found that cooperation of BARF1 with H-Ras and SV40 T antigens was sufficient to transform nonmalignant human nasopharyngeal epithelial NP69 cells when serially introduced BARF1 and H-Ras into the SV40 T antigens-immortalized NP69 cells. Taken together, these results demonstrated that the cooperation of BARF1 with Ras suffices to transform primary primate epithelial cell PATAS. Similarly, BARF1 together with H-Ras and SV40 T can transform human epithelial cell NP69, thereby indicating that BARF1 could be involved in the NPC pathogenesis in combination with additional genetic changes.
Collapse
|
212
|
Transcriptional downregulation of p27KIP1 through regulation of E2F function during LMP1-mediated transformation. J Virol 2009; 83:12671-9. [PMID: 19828622 DOI: 10.1128/jvi.01422-09] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
LMP1 induces the phenotypic transformation of fibroblasts and affects regulators of the cell cycle during this process. LMP1 decreases expression of the cyclin-dependent kinase inhibitor p27 and increases the levels and phosphorylation of cyclin-dependent kinase 2 and the retinoblastoma protein. In the present study, the effects of LMP1 on cell cycle progression and the mechanism of p27 downregulation by LMP1 were determined. Although p27 is frequently regulated at the posttranscriptional level during cell cycle progression and in cancer, LMP1 did not decrease ectopically expressed p27. However, LMP1 did decrease p27 RNA levels and inhibited the activity of p27 promoter reporters. The LMP1-regulated promoter element was mapped to a region containing two E2F sites. Electrophoretic mobility shift assays determined that the regulated cis element bound an inhibitory E2F complex containing E2F4 and p130. These findings indicate that LMP1 decreases p27 transcription through effects on E2F family transcription factors. This property likely contributes to the ability of LMP1 to stimulate cell cycle progression.
Collapse
|
213
|
Abstract
Human tumor viruses are associated with a variety of human malignancies, and it is estimated that 15% of all human cancers have a viral etiology. An abnormality in chromosomal ploidy or aneuploidy is a hallmark of cancers. In normal cells, euploidy is governed by several factors including an intact spindle assembly checkpoint, accurate centrosome duplication, and proper cytokinesis. Viral oncoproteins are suggested to perturb the cellular machineries for chromosomal segregation creating aneuploidy which can lead to the malignant transformation of infected cells. Here, we review in brief some of the mechanisms used by viruses that can cause cellular aneuploidy.
Collapse
Affiliation(s)
- Junichiro Yasunaga
- Molecular Virology Section, Laboratory of Molecular Microbiology, The National Institute of Allergy and Infectious Diseases/NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | | |
Collapse
|
214
|
Ndour PA, Ouk TS, Brocqueville G, Mougel A, Vanhecke E, Feuillard J, Coll J, Adriaenssens E. Inhibition of tumor necrosis factor-induced phenotypes by short intracellular versions of latent membrane protein-1. Cell Signal 2009; 22:303-13. [PMID: 19796681 DOI: 10.1016/j.cellsig.2009.09.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Accepted: 09/14/2009] [Indexed: 12/23/2022]
Abstract
Tumor necrosis factor (TNF) is a potent multi-functional cytokine with a homeostatic role in host defence. In case of deregulation, TNF is implicated in numerous pathologies. The latent membrane protein-1 (LMP1) is expressed by Epstein-Barr virus during viral latency and displaying properties of a constitutively activated member of the TNF receptor family. Both TNFR1 and LMP1 share a similar set of proximal adapters and signalling pathways although they display different biological responses. We previously demonstrated that the intracellular part of LMP1, LMP1-CT, a dominant-negative form of LMP1, inhibits LMP1 signalling. Here, we developed shorter versions derived from C-terminal part of LMP1 to investigate their roles on LMP1 and TNF signalling. We constructed several mutants of LMP1 containing a part of cytoplasmic signalling region fused to the green fluorescent protein. These mutants selectively impair signalling by LMP1 and TNF but not by IL-1beta which uses other adapters. Dominant-negative effect was due to binding and sequestration of LMP1 adapters RIP, TRAF2 and TRADD as assessed by coimmunoprecipitation experiments and confocal analysis. Expression of these mutants impairs the recruitment of these adapters by TNFR1 and TNF-associated phenotypes. These mutants did not display cytostatic properties but were able to modulate TNF-induced phenotypes, apoptosis or cell survival, depending on the cell context. Interestingly, these mutants are able to inhibit a pro-inflammatory response in endothelial cells. These data demonstrate that LMP1 derived molecules can be used to design compounds with potential therapeutic roles in diseases due to TNF overactivation.
Collapse
Affiliation(s)
- Papa Alioune Ndour
- CNRS UMR, Institut de Biologie de Lille, IFR, Université Lille-Nord de France, France
| | | | | | | | | | | | | | | |
Collapse
|
215
|
Shair KH, Schnegg CI, Raab-Traub N. Epstein-Barr virus latent membrane protein-1 effects on junctional plakoglobin and induction of a cadherin switch. Cancer Res 2009; 69:5734-42. [PMID: 19584275 PMCID: PMC2771661 DOI: 10.1158/0008-5472.can-09-0468] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Latent membrane protein-1 (LMP1) is considered the major oncoprotein of Epstein-Barr virus and is frequently expressed in nasopharyngeal carcinoma (NPC). LMP1 promotes growth and migration of epithelial cells, and the loss of plakoglobin has been identified as a contributing factor to LMP1-induced migration. Plakoglobin is a junctional protein that can also serve as a transcription factor in Tcf/Lef signaling. To determine the effects of LMP1 on the molecular and functional properties of plakoglobin, LMP1 was overexpressed in the NPC cell line C666-1. LMP1 did not affect plakoglobin stability but did decrease plakoglobin transcription. The resultant decreased levels of nuclear plakoglobin did not affect Tcf/Lef activity or the amount of plakoglobin bound to Tcf4. Although LMP1 induced and stabilized beta-catenin, a protein with common binding partners to plakoglobin, the loss of plakoglobin did not affect its association with Tcf4. However, LMP1 did induce a cadherin switch from E- to N-cadherin, a process involved in cancer progression, and enhanced the association of junctional beta-catenin with N-cadherin. LMP1 decreased overall levels of junctional plakoglobin but the remaining junctional plakoglobin was found associated with the induced N-cadherin. This increased association of junctional plakoglobin with N-cadherin was a distinguishing feature of LMP1-expressing cells that have reduced migration due to restoration of plakoglobin. Low levels of plakoglobin were also detected in human NPC tissues. These findings reveal that the effects of LMP1 on junctional plakoglobin and the initiation of a cadherin switch likely contribute to metastasis of NPC.
Collapse
Affiliation(s)
- Kathy H.Y Shair
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| | - Caroline I. Schnegg
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| | - Nancy Raab-Traub
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
- Department of Microbiology-Immunology, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| |
Collapse
|
216
|
Li DJ, Bei JX, Mai SJ, Xu JF, Chen LZ, Zhang RH, Yu XJ, Hong MH, Zeng YX, Kang T. The dominance of China 1 in the spectrum of Epstein-Barr virus strains from Cantonese patients with nasopharyngeal carcinoma. J Med Virol 2009; 81:1253-60. [PMID: 19475622 DOI: 10.1002/jmv.21503] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nasopharyngeal carcinoma is a disease with a remarkable geographic and ethnic distribution, and has a high incidence in southern China. Infection with Epstein-Barr virus (EBV) is an important contributing factor. The profile of EBV strains in Cantonese patients from Guangdong, the nasopharyngeal carcinoma endemic region in southern China, is described on the sequence variations in latent membrane protein 1 carboxyl-terminus. The results show that China 1 was the dominant EBV strain detected in both the tumor biopsies and samples of throat washings, whereas multiple strains, including China 1, China 2, B95-8, and Med, were detected in blood samples. In addition, a new strain named China 4 was found in blood samples. These findings suggest that the host population is susceptible to the predominant China 1 strain in the nasopharyngeal carcinoma endemic region of China, but its relationship with the host remains to be characterized further.
Collapse
Affiliation(s)
- Da-Jiang Li
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
217
|
Carbone, Riccardo Dolcetti, Ashok R A. What's New in the Biology and Treatment of Undifferentiated Carcinoma of Nasopharyngeal Type? Acta Otolaryngol 2009. [DOI: 10.1080/00016480127360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
218
|
Hadinoto V, Shapiro M, Sun CC, Thorley-Lawson DA. The dynamics of EBV shedding implicate a central role for epithelial cells in amplifying viral output. PLoS Pathog 2009; 5:e1000496. [PMID: 19578433 PMCID: PMC2698984 DOI: 10.1371/journal.ppat.1000496] [Citation(s) in RCA: 195] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Accepted: 06/03/2009] [Indexed: 01/27/2023] Open
Abstract
To develop more detailed models of EBV persistence we have studied the dynamics of virus shedding in healthy carriers. We demonstrate that EBV shedding into saliva is continuous and rapid such that the virus level is replaced in ≤2 minutes, the average time that a normal individual swallows. Thus, the mouth is not a reservoir of virus but a conduit through which a continuous flow stream of virus passes in saliva. Consequently, virus is being shed at a much higher rate than previously thought, a level too high to be accounted for by replication in B cells in Waldeyer's ring alone. Virus shedding is relatively stable over short periods (hours-days) but varies through 3.5 to 5.5 logs over longer periods, a degree of variation that also cannot be accounted for solely by replication in B cells. This variation means, contrary to what is generally believed, that the definition of high and low shedder is not so much a function of variation between individuals but within individuals over time. The dynamics of shedding describe a process governing virus production that is occurring independently ≤3 times at any moment. This process grows exponentially and is then randomly terminated. We propose that these dynamics are best explained by a model where single B cells sporadically release virus that infects anywhere from 1 to 5 epithelial cells. This infection spreads at a constant exponential rate and is terminated randomly, resulting in infected plaques of epithelial cells ranging in size from 1 to 105 cells. At any one time there are a very small number (≤3) of plaques. We suggest that the final size of these plaques is a function of the rate of infectious spread within the lymphoepithelium which may be governed by the structural complexity of the tissue but is ultimately limited by the immune response. Epstein-Barr virus is a human pathogen associated with several human cancers that nevertheless persists benignly as a latent infection in the majority of adults. EBV persistence is characterized by the presence of latently infected cells in the blood and the shedding of virus into saliva. We present the first systematic quantitative analysis of virus shedding. We show, contrary to what was previously thought, that shedding is continuous and at a high level for all subjects tested. This constant presence of infectious virus may be a crucial risk factor in the development of the EBV-associated tumor nasopharyngeal carcinoma. Unlike infected cells in the blood, which are maintained at very stable levels for years, we show that virus shedding is highly variable such that at any time any individual may be a relatively high or low shedder. We have analyzed these dynamics mathematically and with a simple simulation model. We find that they can be explained by a simple exponential function which we hypothesize is the expansion of 1–3 plaques of epithelial cells.
Collapse
Affiliation(s)
- Vey Hadinoto
- Department of Pathology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Michael Shapiro
- Department of Pathology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Chia Chi Sun
- Department of Pathology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - David A. Thorley-Lawson
- Department of Pathology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
219
|
Ho CH, Chen CL, Li WY, Chen CJ. Decoy receptor 3, upregulated by Epstein-Barr virus latent membrane protein 1, enhances nasopharyngeal carcinoma cell migration and invasion. Carcinogenesis 2009; 30:1443-51. [PMID: 19483191 DOI: 10.1093/carcin/bgp135] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Decoy receptor 3 (DcR3), a member of tumor necrosis factor receptor superfamily, has been implicated in tumorigenesis through its abilities to modulate immune responses and induce angiogenesis. Epstein-Barr virus (EBV), a ubiquitous gamma-herpesvirus, is associated with malignancies including nasopharyngeal carcinoma (NPC). Previous studies show that DcR3 is overexpressed in EBV-positive lymphomas and Rta, an EBV transcription activator, can upregulate DcR3 in Burkitt lymphoma cell lines. However, DcR3 expression has not been demonstrated in EBV-associated NPC nor have there been any EBV latent genes linked to DcR3 upregulation. Here, we showed DcR3 was overexpressed in NPC. Higher DcR3 expression score and DcR3-positive rate were found in metastatic NPC than in primary NPC tissues, suggesting DcR3 may enhance cell metastatic potential. This hypothesis is supported by our observation that NPC HONE-1 cells overexpressing DcR3 exhibited significant higher migration and invasion abilities in vitro. We found besides Rta, EBV latent membrane protein (LMP) 1 can upregulate DcR3 via nuclear factor-kappaB and phosphatidylinositol 3-kinase-signaling events. Approximate 75% of LMP1-positive NPC tissues overexpressed DcR3, suggesting LMP1 may enhance DcR3 expression in vivo. Data herein suggested that increasing DcR3 expression by LMP1 not only helps EBV-associated cancer cells gain survival advantage by preventing host immune detection but also increases the chance of cancer metastasis by enhancing cell migration and invasion. All these DcR3-mediated events facilitate normal cells to gain cancer hallmarks.
Collapse
Affiliation(s)
- Cheng-Hsun Ho
- Institute of Microbiology and Immunology, National Yang-Ming University, No. 155, Section 2, Linong Street, Taipei 112, Taiwan
| | | | | | | |
Collapse
|
220
|
Kieser A. Pursuing different 'TRADDes': TRADD signaling induced by TNF-receptor 1 and the Epstein-Barr virus oncoprotein LMP1. Biol Chem 2009; 389:1261-71. [PMID: 18713013 DOI: 10.1515/bc.2008.144] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The pro-apoptotic tumor necrosis factor (TNF)-receptor 1-associated death domain protein (TRADD) was initially identified as the central signaling adapter molecule of TNF-receptor 1 (TNFR1). Upon stimulation with the pro-inflammatory cytokine TNFalpha, TRADD is recruited to the activated TNFR1 by direct interaction between the death domains of both molecules. TRADD mediates TNFR1 activation of NF-kappaB and c-Jun N-terminal kinase (JNK), as well as caspase-dependent apoptosis. Surprisingly, TRADD is also recruited by latent membrane protein 1 (LMP1), the major oncoprotein of the human Epstein-Barr tumor virus. By mimicking a constitutively active receptor, LMP1 is essential for B-cell transformation by the virus, activating NF-kappaB, phosphatidylinositol 3-kinase, JAK/STAT and mitogen-activated protein kinase signaling. In contrast to TNFR1, LMP1's interaction with TRADD is independent of a functional death domain. The unique structure of the LMP1-TRADD complex dictates an unusual type of TRADD-dependent NF-kappaB signaling and subverts TRADD's potential to induce apoptosis. This article provides an overview of TNFR1 and LMP1 signal transduction with a focus on TRADD's functions in apoptotic and transforming signaling, incorporating recent results from TRADD RNAi and knockout studies.
Collapse
Affiliation(s)
- Arnd Kieser
- Abteilung Genvektoren, Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt, Marchioninistrasse 25, D-81377 München, Germany.
| |
Collapse
|
221
|
Association between human papillomavirus and Epstein-Barr virus infections in human oral carcinogenesis. Med Hypotheses 2009; 73:184-6. [PMID: 19361933 DOI: 10.1016/j.mehy.2009.02.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 02/18/2009] [Accepted: 02/21/2009] [Indexed: 11/21/2022]
Abstract
Infection by high-risk human papillomaviruses (HPVs) and Epstein-Barr virus (EBV) are very frequent in the adult human population, and have been associated with several human carcinomas, especially oral cancers. However, a small number of studies have examined the association between high-risk HPV and EBV in the progression of human oral cancers. Currently, the role of high-risk HPV and EBV co-infections in human oral cancers, particularly nasopharyngeal carcinomas, remain uncertain because of the limited number of investigations. This raises the question whether high-risk HPV and EBV co-infections play a significant role in the development of human nasopharyngeal carcinomas. In this paper, we propose the hypothesis that human oral normal epithelial cells, especially nasopharyngeal cells, are very susceptible to persistent HPV and EBV co-infections; therefore, high-risk HPV and EBV co-infections play an important role in the initiation of a neoplastic transformation of human oral epithelial cells. We believe that significant studies, using different cells and animal models as well as clinical samples, are necessary to answer these important questions.
Collapse
|
222
|
Lee JW, Liu PF, Hsu LP, Chen PR, Chang CH, Shih WL. EBV LMP-1 negatively regulates expression and pro-apoptotic activity of Par-4 in nasopharyngeal carcinoma cells. Cancer Lett 2009; 279:193-201. [PMID: 19250735 DOI: 10.1016/j.canlet.2009.01.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 12/24/2008] [Accepted: 01/27/2009] [Indexed: 12/19/2022]
Abstract
Latent membrane protein-1 (LMP-1) of the Epstein-Barr virus (EBV) is closely associated with nasopharyngeal carcinoma (NPC), and in this study we sought to determine whether the pro-apoptotic activity of prostate apoptosis response-4 (Par-4) is modulated by LMP-1 in NPC cells. We found that LMP-1 diminished the pro-apoptotic activity of Par-4 and negatively regulated Par-4 protein by de novo synthesis; moreover, although LMP-1 accelerated a Par-4 activator, PKA, we demonstrated that LMP-1 also activated the PI3K/Akt pathway and increased Bcl-2 expression to suppress the activity of Par-4. Consequently, our results revealed a novel negative action of LMP-1 on the pro-apoptosis protein Par-4 by the coordination of multiple signaling pathways.
Collapse
Affiliation(s)
- Jeng-Woei Lee
- Department and Institute of Life Science, College of Life Science, Tzu-Chi University, No. 701, Zhongyang Rd., Sec. 3, Hualien 97004, Taiwan.
| | | | | | | | | | | |
Collapse
|
223
|
MDM2-dependent inhibition of p53 is required for Epstein-Barr virus B-cell growth transformation and infected-cell survival. J Virol 2009; 83:2491-9. [PMID: 19144715 DOI: 10.1128/jvi.01681-08] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Epstein-Barr virus (EBV) growth transformation of primary B lymphocytes into indefinitely proliferating lymphoblastoid cell lines (LCLs) depends on the concerted activities of a subset of viral proteins expressed during latency. EBV drives quiescent B cells into S phase, and consequently, a host response is activated that includes expression of p53 and its target genes. Since LCLs retain wild-type p53, it was of interest to determine what contribution the p53 pathway may have in controlling established LCL growth and EBV-mediated transformation of primary B cells. We found that liberation of p53 through chemical antagonism of one of its major ubiquitin ligases, MDM2, using the small-molecule Nutlin-3 led to apoptosis of established LCLs and suppressed EBV-mediated transformation of primary B cells. The activation of latent p53 induced target genes associated with apoptosis. Furthermore, MDM2 antagonism synergized with NF-kappaB inhibition in killing LCLs. NF-kappaB was important to increase steady-state MDM2 protein levels rather than in affecting p53-dependent transcription, suggesting a unique mechanism by which LCLs survive in the presence of a primed p53 pathway. Nutlin sensitivity of EBV-infected cells provides a novel system for studying the pathways that dictate LCL survival and regulate EBV transformation. Finally, MDM2 antagonists may be considered for therapeutic intervention in EBV-associated malignancies expressing wild-type p53.
Collapse
|
224
|
Vaysberg M, Hatton O, Lambert SL, Snow AL, Wong B, Krams SM, Martinez OM. Tumor-derived variants of Epstein-Barr virus latent membrane protein 1 induce sustained Erk activation and c-Fos. J Biol Chem 2008; 283:36573-85. [PMID: 18986987 PMCID: PMC2605991 DOI: 10.1074/jbc.m802968200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Revised: 10/03/2008] [Indexed: 12/18/2022] Open
Abstract
Latent membrane protein 1 (LMP1) of Epstein-Barr virus (EBV) is a proven oncogene that is essential for transformation of human B cells by the virus. LMP1 induces constitutive activation of several signal transduction pathways involving nuclear factor kappaB, phosphatidylinositol 3-kinase/Akt, and the mitogen-activated protein kinases (MAPK) p38, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (Erk). Sequencing of LMP1 isolated from a panel of EBV+ B cell lymphomas identified three different variants of LMP1, each distinct from the B95.8 prototype isoform. All tumor variants of LMP1 as well as the B95.8 LMP1 isoform were able to induce rapid p38 phosphorylation as well as Akt and JNK activation. Additionally all variants showed similar ability to activate nuclear factor kappaB. In contrast, only tumor-derived LMP1 variants induced prolonged Erk activation and c-Fos expression. Sequence analysis revealed only two amino acids, 212 and 366, shared by the tumor variants but distinct from B95.8. Point mutation of either amino acids 212 (glycine to serine) or 366 (serine to threonine) from the B95.8 isoform to the tumor variant version of LMP1 was sufficient for gain of function characterized by sustained activation of Erk and subsequent c-Fos induction and binding to the AP1 site. Our results indicate that the enhanced ability of tumor-derived LMP1 to induce and stabilize the c-Fos oncogene can be localized to two amino acids in the C terminus of LMP1.
Collapse
Affiliation(s)
- Maria Vaysberg
- Program in Immunology, Department of Surgery, Division of Transplantation, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | |
Collapse
|
225
|
Thorley-Lawson DA, Allday MJ. The curious case of the tumour virus: 50 years of Burkitt's lymphoma. Nat Rev Microbiol 2008; 6:913-24. [PMID: 19008891 DOI: 10.1038/nrmicro2015] [Citation(s) in RCA: 201] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Burkitt's lymphoma (BL) was first described 50 years ago, and the first human tumour virus Epstein-Barr virus (EBV) was discovered in BL tumours soon after. Since then, the role of EBV in the development of BL has become more and more enigmatic. Only recently have we finally begun to understand, at the cellular and molecular levels, the complex and interesting interaction of EBV with B cells that creates a predisposition for the development of BL. Here, we discuss the intertwined histories of EBV and BL and their relationship to the cofactors in BL pathogenesis: malaria and the MYC translocation.
Collapse
Affiliation(s)
- David A Thorley-Lawson
- Department of Pathology, Jaharis Building, Tufts University School of Medicine, 150 Harrison Avenue, Boston, Massachusetts 02111, USA.
| | | |
Collapse
|
226
|
Latent membrane protein-1 of Epstein-Barr virus induces the expression of B-cell integration cluster, a precursor form of microRNA-155, in B lymphoma cell lines. Biochem Biophys Res Commun 2008; 377:579-583. [DOI: 10.1016/j.bbrc.2008.10.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Accepted: 10/03/2008] [Indexed: 12/12/2022]
|
227
|
Luo W, Yan G, Li L, Wang Z, Liu H, Zhou S, Liu S, Tang M, Yi W, Dong Z, Cao Y. Epstein–Barr virus latent membrane protein 1 mediates serine 25 phosphorylation and nuclear entry of annexin A2 via PI‐PLC–PKCα/PKCβ pathway. Mol Carcinog 2008; 47:934-46. [DOI: 10.1002/mc.20445] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Wei Luo
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Guangrong Yan
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Lili Li
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Zhenlian Wang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Haidan Liu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Shanghui Zhou
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Sufang Liu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Min Tang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Wei Yi
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, 55912
| | - Ya Cao
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| |
Collapse
|
228
|
Nuclear factor-kappaB binds to the Epstein-Barr Virus LMP1 promoter and upregulates its expression. J Virol 2008; 83:1393-401. [PMID: 19019967 DOI: 10.1128/jvi.01637-08] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The latent membrane protein 1 (LMP1) oncogene carried by Epstein-Barr virus (EBV) is essential for transformation and maintenance of EBV-immortalized B cells in vitro, and it is expressed in most EBV-associated tumor types. The activation of the NF-kappaB pathway by LMP1 plays a critical role in the upregulation of antiapoptotic proteins. The EBV-encoded EBNA2 transactivator is required for LMP1 activation in latency III, while LMP1 itself appears to be critical for its activation in the latency II gene expression program. In both cases, additional viral and cellular transcription factors are required in mediating transcription activation of the LMP1 promoter. Using DNA affinity purification and chromatin immunoprecipitation assay, we showed here that members of the NF-kappaB transcription factor family bound to the LMP1 promoter in vitro and in vivo. Electrophoretic mobility shift assay analyses indicated the binding of the p50-p50 homodimer and the p65-p50 heterodimer to an NF-kappaB site in the LMP1 promoter. Transient transfections and reporter assays showed that the LMP1 promoter is activated by exogenous expression of NF-kappaB factors in both B cells and epithelial cells. Exogenous expression of NF-kappaB factors in the EBNA2-deficient P3HR1 cell line induced LMP1 protein expression. Overall, our data are consistent with the presence of a positive regulatory circuit between NF-kappaB activation and LMP1 expression.
Collapse
|
229
|
Middeldorp JM, Pegtel DM. Multiple roles of LMP1 in Epstein-Barr virus induced immune escape. Semin Cancer Biol 2008; 18:388-96. [PMID: 19013244 DOI: 10.1016/j.semcancer.2008.10.004] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 10/16/2008] [Indexed: 12/21/2022]
Abstract
The life cycle of Epstein-Barr virus (EBV) is intriguing in that the virus resides within the immune system and utilizes distinct latency expression programs to establish a persistent infection yet escaping elimination. To achieve this EBV has hijacked cellular signaling pathways to its own benefit, but deregulated viral gene expression can turn into oncogenesis. EBV like many other persistent herpes viruses has evolved ingenious tricks to evade the immune system in part by mimicking host gene function(s). Latent membrane protein 1 (LMP1) mimics CD40 signaling as part of its "normal" biological function and when deregulated, functions as a viral oncogene. LMP1 also affects cell-cell contact, cytokine and chemokine production, Ag presentation and is secreted in the extracellular milieu via immunogenic exosomes. Thus, besides its well-known growth promoting properties LMP1 modulates immune responses. Herein we discuss current knowledge regarding the role of LMP1 in immune evasion of EBV and how this strategy for establishment of persistence contributes to immune escape of EBV+ tumors.
Collapse
Affiliation(s)
- J M Middeldorp
- VU University Medical Center, Department of Pathology and Cancer Center Amsterdam, The Netherlands.
| | | |
Collapse
|
230
|
Abstract
Among the most common HIV-associated lymphomas are Burkitt lymphoma (BL) and diffuse large B-cell lymphoma (DLBCL) with immunoblastic-plasmacytoid differentiation (also involving the central nervous system). Lymphomas occurring specifically in HIV-positive patients include primary effusion lymphoma (PEL) and its solid variants, plasmablastic lymphoma of the oral cavity type and large B-cell lymphoma arising in Kaposi sarcoma herpesvirus (KSHV)-associated multicentric Castleman disease. These lymphomas together with BL and DLBCL with immunoblastic-plasmacytoid differentiation frequently carry EBV infection and display a phenotype related to plasma cells. EBV infection occurs at different rates in different lymphoma types, whereas KSHV is specifically associated with PEL, which usually occurs in the setting of profound immunosuppression. The current knowledge about HIV-associated lymphomas can be summarized in the following key points: (1) lymphomas specifically occurring in patients with HIV infection are closely linked to other viral diseases; (2) AIDS lymphomas fall in a spectrum of B-cell differentiation where those associated with EBV or KSHV commonly exhibit plasmablastic differentiation; and (3) prognosis for patients with lymphomas and concomitant HIV infection could be improved using better combined chemotherapy protocols incorporating anticancer treatments and antiretroviral drugs.
Collapse
|
231
|
Abstract
The ubiquitin system of protein modification has emerged as a crucial mechanism involved in the regulation of a wide array of cellular processes. As our knowledge of the pathways in this system has grown, so have the ties between the protein ubiquitin and human disease. The power of the ubiquitin system for therapeutic benefit blossomed with the approval of the proteasome inhibitor Velcade in 2003 by the FDA. Current drug discovery activities in the ubiquitin system seek to (i) expand the development of new proteasome inhibitors with distinct mechanisms of action and improved bioavailability, and (ii) validate new targets. This review summarizes our current understanding of the role of the ubiquitin system in various human diseases ranging from cancer, viral infection and neurodegenerative disorders to muscle wasting, diabetes and inflammation. I provide an introduction to the ubiquitin system, highlight some emerging relationships between the ubiquitin system and disease, and discuss current and future efforts to harness aspects of this potentially powerful system for improving human health. Republished from Current BioData's Targeted Proteins database (TPdb; ).
Collapse
|
232
|
Gullo C, Low WK, Teoh G. Association of Epstein-Barr Virus with Nasopharyngeal Carcinoma and Current Status of Development of Cancer-derived Cell Lines. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2008. [DOI: 10.47102/annals-acadmedsg.v37n9p769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
It is well known that the Epstein-Barr virus (EBV) contributes directly to tumourigenesis in nasopharyngeal carcinoma (NPC), primarily in the undifferentiated form of NPC (WHO type III; UNPC or UC), which is commonly found in South East Asia. Unfortunately, research in NPC has been severely hampered by the lack of authentic EBV-positive (EBV+) human NPC cell lines for study. Since 1975, there have been more than 20 reported NPC cell lines. However, many of these NPC-derived cell lines do not express EBV transcripts in long-term culture, and therefore that finding may dispute the fundamental theory of NPC carcinogenesis. In fact, currently only one EBV+ human NPC cell line (C-666) in long-term culture has been reported. Hence, most of the NPC cell lines may not be representative of the disease itself. In order to better understand and treat NPC, there is an urgent need to develop more EBV+ human NPC cell lines. In this review, we discuss the authenticity of existing NPC cell lines and the impact of our understanding of NPC biology on the treatment of the disease and the relationship of EBV to NPC in the context of cell lines.
Key words: Carcinogenesis, Cell culture, Epstein-Barr virus, Hayflick’s limit
Collapse
|
233
|
Abstract
OBJECTIVE Serum and plasma contain species-specific factors that modulate cell population growth and function, and that are required for proliferation of most cell cultures. Foetal calf serum (FCS) is the most common source of these growth factors. We studied the effect of human plasma (HP) on the immortalization process of B lymphocytes by Epstein-Barr virus (EBV) was studied. MATERIALS AND METHODS The effect of HP as compared to FCS was done through assessment of cell proliferation. RESULTS It was found that HP (autologous and non-autologous plasma) is more effective than FCS in generating lymphoblastoid cell lines, regardless of EBV status of the donors: 65% of HP-supplemented cultures developed into lymphoblastoid cell lines by 7-14 culture days, as compared to 16% of cultures with FCS. In addition, 6% of HP-supplemented cultures did not achieve becoming lymphoblastoid cell lines by day 35 in comparison to 94% of cultures with FCS. The higher proliferative effect of HP was not altered by heat inactivation or filtration. HP maintained its proliferative activity at 4 degrees C over 8 months, thus indicating that HP contains a stable growth factor(s), which accelerates B-lymphocyte immortalization. CONCLUSION The results support other studies that recommend the use of autologous plasma for tissue culture, mainly in the case of autologous transplantation. Furthermore, the use of HP allows preparation of lymphoblastoid cell lines from a small amount of peripheral blood in a shorter period of time and with a higher rate of success.
Collapse
Affiliation(s)
- E Manor
- Genetics Laboratories, Institute of Genetics, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
234
|
Tsuji A, Wakisaka N, Kondo S, Murono S, Furukawa M, Yoshizaki T. Induction of Receptor for Advanced Glycation End Products by EBV Latent Membrane Protein 1 and Its Correlation with Angiogenesis and Cervical Lymph Node Metastasis in Nasopharyngeal Carcinoma. Clin Cancer Res 2008; 14:5368-75. [DOI: 10.1158/1078-0432.ccr-08-0198] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
235
|
Abstract
Latent membrane protein 1 (LMP1), the major oncoprotein of EBV, is likely responsible for many of the altered cellular growth properties in EBV-associated cancers, including nasopharyngeal carcinoma (NPC). In this study, the effects of LMP1 on cell growth and migration were studied in the context of the EBV-positive C666-1 NPC cell line. In the soft agar transformation and Transwell metastasis assays, LMP1 enhanced cell growth and migration through activation of phosphatidylinositol 3-kinase (PI3K)/Akt and nuclear factor-kappaB (NF-kappaB) signaling. Inhibitors of PI3K, Akt, and NF-kappaB signaling dramatically reduced these enhanced properties. An IkappaBalpha super-repressor also blocked these effects. However, constitutive activation of Akt alone did not alter cell growth, suggesting that both PI3K/Akt and NF-kappaB activation are required by LMP1. These enhanced effects required the full-length LMP1 encompassing both the PI3K/Akt-activating COOH-terminal activation region (CTAR) 1 and the nonredundant NF-kappaB-activating regions CTAR1 and CTAR2. LMP2A, a latent protein that is also frequently expressed in NPC, similarly activates the PI3K/Akt pathway; however, its overexpression in C666-1 cells did not affect cell growth or migration. LMP1 also decreased expression of the junctional protein plakoglobin, which was shown to be partially responsible for enhanced migration induced by LMP1. This study reveals that in epithelial cells the transforming properties of LMP1 require activation of both PI3K/Akt and NF-kappaB and shows that the loss of plakoglobin expression by LMP1 is a significant factor in the enhanced migration.
Collapse
Affiliation(s)
- Kathy H.Y Shair
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| | - Caroline I. Schnegg
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| | - Nancy Raab-Traub
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
- Department of Microbiology-Immunology, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| |
Collapse
|
236
|
Selgrad M, Malfertheiner P, Fini L, Goel A, Boland CR, Ricciardiello L. The role of viral and bacterial pathogens in gastrointestinal cancer. J Cell Physiol 2008; 216:378-88. [PMID: 18338378 PMCID: PMC2855192 DOI: 10.1002/jcp.21427] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The association of Helicobacter pylori (H. pylori) with gastric cancer is thus far the best understood model to comprehend the causal relationship between a microbial pathogen and cancer in the human gastrointestinal tract. Besides H. pylori, a variety of other pathogens are now being recognized as potential carcinogens in different settings of human cancer. In this context, viral causes of human cancers are central to the issue since these account for 10-20% of cancers worldwide. In the case of H. pylori and gastric cancer, as well as the human papillomavirus and anal cancer, the causal relationship between the infectious agent and the related cancer in the gastrointestinal tract has been clearly confirmed by epidemiological and experimental studies. Similarly, Epstein-Barr virus and the oncogenic JC virus are being suggested as possible causative agents for cancers in the upper and lower gastrointestinal tract. This review discusses various viral and microbial pathogens and their oncogenic properties in the evolution of gastrointestinal carcinogenesis and summarizes the available experimental data make a convincing agreement favoring the associations between infectious agents and specific human cancers.
Collapse
Affiliation(s)
- Michael Selgrad
- Department of Internal Medicine, Gastroenterology, Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Lucia Fini
- Department of Internal Medicine, Gastroenterology, Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| | - Ajay Goel
- Department of Internal Medicine, Gastroenterology, Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| | - C. Richard Boland
- Department of Internal Medicine, Gastroenterology, Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| | - Luigi Ricciardiello
- Department of Internal Medicine, Gastroenterology, Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| |
Collapse
|
237
|
Chow KPN, Wu CC, Chang HY, Chang C, Chang YS. A simplified tumour model established via Epstein-Barr virus-encoded, nasopharyngeal carcinoma-derived oncogene latent membrane protein 1 in immunocompetent mice. Lab Anim 2008; 42:193-203. [PMID: 18435877 DOI: 10.1258/la.2007.006037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The expression and immune modulation of Epstein-Barr virus-encoded oncogene latent membrane protein 1 (N-LMP1) is essential in the pathogenesis of nasopharyngeal carcinoma. In previous studies, cell transformation has been induced by the expression of EBV-encoded N-LMP1 in non-tumour BALB/c-3T3 cells and these cells have then been used to form tumours in T-cell-deficient nude mice. However, studies using this model have been limited by the lack of a competent immune system. To facilitate the study of immune components in N-LMP1-driven oncogenesis, we herein developed a simplified N-LMP1-derived tumour model in immunocompetent mice. Cell transformation was induced by the expression of N-LMP1 in BALB/c-3T3 cells, and these transformants were used to induce oncogenesis in BALB/c mice. In contrast to the 100% successful tumour-induction rate in nude mice treated with monodispersed transformed cells, the tumour incidence in BALB/c mice was only 5-36%. However, the transplantation of tumour fragments into BALB/c mice yielded a reproducible tumour-induction rate of >85%, which is acceptable for most of the research needs. This novel model of N-LMP1-directed oncogenesis in an immunocompetent environment may serve as an important platform for the future assessment of N-LMP1-targeted tumour therapies.
Collapse
Affiliation(s)
- Kai-Ping N Chow
- Department of Microbiology and Immunology, School of Medicine, Chang-Gung University, Kwei-shan, Taoyuan 333, Taiwan, Republic of China.
| | | | | | | | | |
Collapse
|
238
|
Everly DN, Mainou BA, Raab-Traub N. The ID proteins contribute to the growth of rodent fibroblasts during LMP1-mediated transformation. Virology 2008; 376:258-69. [PMID: 18456300 PMCID: PMC2901101 DOI: 10.1016/j.virol.2007.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 09/14/2007] [Accepted: 12/03/2007] [Indexed: 12/17/2022]
Abstract
LMP1 induces the expression of two members of the family of Id proteins, Id1 and Id3, and affects cell cycle regulation by decreasing the expression of the cyclin dependent kinase inhibitor, p27, and increasing levels and phosphorylation of cdk2 and Rb. In the present study, the contribution of the Id proteins to LMP1-mediated transformation was determined. Although LMP1 effectively inhibited p27 expression, the Id proteins alone did not affect expression of p27, cdk2, and Rb. Neither Id1 nor Id3 was sufficient to transform Rat-1 cells and inhibition of Id1 expression did not affect LMP1-induced morphologic transformation of Rat-1 cells or reduction of p27. However, reduced Id expression resulted in smaller foci and impaired the growth rate of Rat-1 cells. These data indicate that overexpression of the Id proteins is not sufficient for the effects of LMP1 on the cell cycle but that inhibition of Id expression does affect the growth of LMP1-transformed and parental Rat1 cells.
Collapse
Affiliation(s)
- David N Everly
- Lineberger Comprehensive Cancer Center, CB#7295, University of North Carolina-Chapel Hill, 450 West Drive, Chapel Hill, NC 27599-7295, USA.
| | | | | |
Collapse
|
239
|
Chou J, Lin YC, Kim J, You L, Xu Z, He B, Jablons DM. Nasopharyngeal carcinoma--review of the molecular mechanisms of tumorigenesis. Head Neck 2008; 30:946-63. [PMID: 18446839 PMCID: PMC3046044 DOI: 10.1002/hed.20833] [Citation(s) in RCA: 240] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a head and neck cancer rare throughout most of the world but common in certain geographic areas, such as southern Asia. While environmental factors and genetic susceptibility play important roles in NPC pathogenesis, the Epstein-Barr virus in particular has been implicated in the molecular abnormalities leading to NPC. There is upregulation of cellular proliferation pathways such as the Akt pathway, mitogen-activated protein kinases, and the Wnt pathway. Cell adhesion is compromised due to abnormal E-cadherin and beta-catenin function. Aberrations in cell cycle are due to dysregulation of factors such as p16, cyclin D1, and cyclin E. Anti-apoptotic mechanisms are also upregulated. There are multiple abnormalities unique to NPC that are potential targets for novel treatments.
Collapse
Affiliation(s)
- Josephine Chou
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, California 94115, USA
| | | | | | | | | | | | | |
Collapse
|
240
|
STAP-2 negatively regulates both canonical and noncanonical NF-kappaB activation induced by Epstein-Barr virus-derived latent membrane protein 1. Mol Cell Biol 2008; 28:5027-42. [PMID: 18573890 DOI: 10.1128/mcb.00194-08] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The signal-transducing adaptor protein 2 (STAP-2) is a recently identified adaptor protein that contains a pleckstrin homology (PH) and Src homology 2 (SH2)-like domains, as well as a proline-rich domain in its C-terminal region. In previous studies, we demonstrated that STAP-2 binds to MyD88 and IKK-alpha or IKK-beta and modulates NF-kappaB signaling in macrophages. In the present study, we found that ectopic expression of STAP-2 inhibited Epstein-Barr virus (EBV) LMP1-mediated NF-kappaB signaling and interleukin-6 expression. Indeed, STAP-2 associated with LMP1 through its PH and SH2-like domains, and these proteins interacted with each other in EBV-positive human B cells. We found, furthermore, that STAP-2 regulated LMP1-mediated NF-kappaB signaling through direct or indirect interactions with the tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) and TNFR-associated death domain (TRADD) proteins. STAP-2 mRNA was induced by the expression of LMP1 in human B cells. Furthermore, transient expression of STAP-2 in EBV-positive human B cells decreased cell growth. Finally, STAP-2 knockout mouse embryonic fibroblasts showed enhanced LMP1-induced cell growth. These results suggest that STAP-2 acts as an endogenous negative regulator of EBV LMP1-mediated signaling through TRAF3 and TRADD.
Collapse
|
241
|
Pich D, Humme S, Spindler MP, Schepers A, Hammerschmidt W. Conditional gene vectors regulated in cis. Nucleic Acids Res 2008; 36:e83. [PMID: 18566006 PMCID: PMC2490737 DOI: 10.1093/nar/gkn273] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Non-integrating gene vectors, which are stably and extrachromosomally maintained in transduced cells would be perfect tools to support long-term expression of therapeutic genes but preserve the genomic integrity of the cellular host. Small extrachromosomal plasmids share some of these ideal characteristics but are primarily based on virus blueprints. These plasmids are dependent on viral trans-acting factors but they can replicate their DNA molecules in synchrony with the chromosome of the cellular host and segregate to daughter cells in an autonomous fashion. On the basis of the concept of the latent origin of DNA replication of Epstein-Barr virus, oriP, we devised novel derivatives, which exclusively rely on an artificial replication factor for both nuclear retention and replication of plasmid DNA. In addition, an allosteric switch regulates the fate of the plasmid molecules, which are rapidly lost upon addition of doxycycline. Conditional maintenance of these novel plasmid vectors allows the reversible transfer of genetic information into target cells for the first time.
Collapse
Affiliation(s)
- Dagmar Pich
- Department of Gene Vectors, Helmholtz Center Munich, German Research Center for Environmental Health, Marchioninistr. 25, 81377 Munich, Germany
| | | | | | | | | |
Collapse
|
242
|
Fiorini S, Ooka T. Secretion of Epstein-Barr virus-encoded BARF1 oncoprotein from latently infected B cells. Virol J 2008; 5:70. [PMID: 18533018 PMCID: PMC2430953 DOI: 10.1186/1743-422x-5-70] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Accepted: 06/04/2008] [Indexed: 11/24/2022] Open
Abstract
Epstein-Barr virus (EBV) encodes two oncogenes, LMP1(Latent Membrane Protein-1) and BARF1 (BamH1-A Reading Frame-1). LMP1 belongs to latent gene family and BARF1 is considered so far as one of early gene family. However BARF1 oncogene was expressed highly in Nasopharyngeal (NPC) and gastric (GC) carcinoma as a type II latency, and in EBV-positive Akata cell and primary epithelial cell infected in vitro by EBV as type I latency. Its expression was also reported in Burkitt's lymphoma's biopsy frequent in Malawi in Africa as well as in nasal NK/T-cell lymphoma. We recently observed a massive secretion of BARF1 protein in serum and saliva of NPC patients. NPC-derived c666-1 epithelial cells also expressed and secreted BARF1 protein without other lytic genes expression. We asked whether this oncogene belongs to latent gene family. To investigate, we examined its transcriptional and translational expression in IB4 and Akata B cells where both cell lines belong to latent cell family. Transcriptional expression was analyzed by RT-PCR. As BARF1 protein is one of secreted proteins, its translational expression was analyzed by immunoblot after concentration of culture medium. Secreted BARF1 protein was futher purified by concanavalin A affinity column. BARF1 was transcribed in both EBV-positive AKATA and IB4 cells, and BARF1 protein was secreted from these latently infected human B cells. Its secretion does not depend EBV genome form in infected cells. Both episomal and integrated form of EBV genome were capable of expressing BARF1 gene. These results suggests that BARF1 is expressed in latent stage and increases its expression during lytic stage.
Collapse
Affiliation(s)
- Sylvie Fiorini
- Laboratoire de Virologie Moléculaire, Virologie et Pathogenèse Humaine, FRE 3011, CNRS, Faculté de Médecine Laennec, Université Claude Bernard Lyon-1, Rue Guillaume Paradin, 69372, Lyon Cedex 08, France
| | - Tadamasa Ooka
- Laboratoire de Virologie Moléculaire, Virologie et Pathogenèse Humaine, FRE 3011, CNRS, Faculté de Médecine Laennec, Université Claude Bernard Lyon-1, Rue Guillaume Paradin, 69372, Lyon Cedex 08, France
| |
Collapse
|
243
|
Epstein-Barr virus latent membrane protein 1 represses DNA repair through the PI3K/Akt/FOXO3a pathway in human epithelial cells. J Virol 2008; 82:8124-37. [PMID: 18524825 DOI: 10.1128/jvi.00430-08] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Latent membrane protein 1 (LMP1), an Epstein-Barr virus (EBV) oncoprotein, mimics a constitutively activated tumor necrosis factor receptor and activates various signaling pathways, including phosphatidylinositol 3-kinase (PI3K)/Akt. LMP1 is essential for EBV-mediated B-cell transformation and is sufficient to transform several cell lines. Cellular transformation has been associated strongly with genomic instability, while DNA repair plays an important role in maintaining genomic stability. Previously, we have shown that LMP1 represses DNA repair by the C-terminal activating region 1 (CTAR1) in human epithelial cells. In the present study, we demonstrate that the PI3K/Akt pathway is required for LMP1-mediated repression of DNA repair. Through the LMP1/PI3K/Akt pathway, FOXO3a, which can induce DNA repair, is inactivated because of phosphorylation and relocalization. Expression of a constitutively active FOXO3a mutant can rescue LMP1-mediated repression of DNA repair. Furthermore, LMP1 can decrease the expression of DNA damage-binding protein 1 (DDB1), which functions in nucleotide excision repair, through the PI3K/Akt/FOXO3a pathway. LMP1-mediated repression of DNA repair is restored by DDB1, although only partially. These results suggest that LMP1 triggers the PI3K/Akt pathway to inactivate FOXO3a and decrease DDB1, which can lead to repression of DNA repair and may contribute to genomic instability in human epithelial cells.
Collapse
|
244
|
Rohr JC, Wagner HJ, Lauten M, Wacker HH, Jüttner E, Hanke C, Pohl M, Niemeyer CM. Differentiation of EBV-induced post-transplant Hodgkin lymphoma from Hodgkin-like post-transplant lymphoproliferative disease. Pediatr Transplant 2008; 12:426-31. [PMID: 18466428 DOI: 10.1111/j.1399-3046.2007.00816.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The development of lymphomas after SOT is a well-known complication of the immunosuppressive therapy necessary to prevent graft rejection. Epstein-Barr virus plays a central role in the pathogenesis of lymphomas because of its ability to transform infected cells. Differentiating PTLD from malignant lymphomas, especially HL can be challenging. We report on two patients, who developed EBV-associated lymphomas several years after SOT. A histological examination of lymph nodes led to a diagnosis of HL in both patients, who were started on chemotherapy according to current treatment protocols. A rapid and complete remission in one patient prompted us to analyze the expression pattern of EBV-latency genes. In this patient, the EBV expression profile revealed a latency type III suggesting the diagnosis of Hodgkin-like PTLD. The other patient required six courses of chemotherapy plus radiotherapy to reach a complete remission. In his tumor cells, a restricted EBV-latency type II pattern was found, suggesting a diagnosis of classical HL. These two cases demonstrate that in post-transplant lymphomas with histological features of HL, an analysis of the expression pattern of EBV proteins might aid in the differentiation between PTLD and HL.
Collapse
Affiliation(s)
- Jan C Rohr
- Department of Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
245
|
Morris MA, Young LS, Dawson CW. DNA tumour viruses promote tumour cell invasion and metastasis by deregulating the normal processes of cell adhesion and motility. Eur J Cell Biol 2008; 87:677-97. [PMID: 18468721 DOI: 10.1016/j.ejcb.2008.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2007] [Revised: 03/17/2008] [Accepted: 03/17/2008] [Indexed: 01/11/2023] Open
Abstract
Approximately 15-20% of global cancer incidence is causally linked to viral infection, yet the low incidence of cancers in healthy infected individuals suggests that malignant conversion of virus-infected cells occurs after a long period as a result of additional genetic modifications. There are four families of viruses that are now documented to be involved in the development of human cancers which include members of the polyomavirus, hepadnavirus, papillomavirus and herpesvirus families. Although a number of these viruses are implicated in the aetiology of lymphomas or leukaemias, the vast majority are associated with malignancies of epithelial cells. In epithelial tissues, several classes of proteins are involved in maintaining tissue architecture, including those that promote cell-cell adhesion, and others, which mediate cell-matrix interactions. Proteins representative of all classes are frequently altered in malignant tumour cells that possess invasive and metastatic properties. Malignant tumour cells acquire mechanisms to degrade basement membranes and invade the underlying tissue. Many viruses encode proteins which engage signalling pathways that affect one or more of these mechanisms. It is believed that activation of these processes by chronic viral infection can, under certain circumstances, promote tumour cell invasion and metastasis. This review will take a brief look at the current knowledge of viral-induced alterations in cell motility and invasiveness in the context of tumour invasion and metastasis.
Collapse
Affiliation(s)
- Mhairi A Morris
- Cancer Research (UK) Institute for Cancer Studies, The Medical School, University of Birmingham, Birmingham B15 2TT, UK.
| | | | | |
Collapse
|
246
|
Deyrup AT. Epstein-Barr virus-associated epithelial and mesenchymal neoplasms. Hum Pathol 2008; 39:473-83. [PMID: 18342658 DOI: 10.1016/j.humpath.2007.10.030] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Revised: 10/29/2007] [Accepted: 10/30/2007] [Indexed: 01/22/2023]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous human pathogen that usually maintains a harmonious relationship with its host. Rarely, this host-virus balance is perturbed, causing a diverse group of malignancies in both immunocompetent and immunosuppressed patients. In addition to its role in hematologic malignancies (Burkitt lymphoma, subsets of Hodgkin and T-cell lymphomas, posttransplant lymphomas), EBV has been implicated in both epithelial (undifferentiated nasopharyngeal carcinoma, a subset of gastric adenocarcinomas) and mesenchymal (EBV-associated smooth muscle tumor, inflammatory pseudotumor-like follicular dendritic cell tumor) neoplasms. This review will focus on EBV-associated epithelial and mesenchymal neoplasms.
Collapse
Affiliation(s)
- Andrea T Deyrup
- Department of Pathology, Emory University Atlanta, GA 30322, USA.
| |
Collapse
|
247
|
Tiwawech D, Srivatanakul P, Karalak A, Ishida T. Association between EBNA2 and LMP1 subtypes of Epstein-Barr virus and nasopharyngeal carcinoma in Thais. J Clin Virol 2008; 42:1-6. [DOI: 10.1016/j.jcv.2007.11.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Revised: 10/17/2007] [Accepted: 11/11/2007] [Indexed: 11/28/2022]
|
248
|
Interferon regulatory factor 4 is involved in Epstein-Barr virus-mediated transformation of human B lymphocytes. J Virol 2008; 82:6251-8. [PMID: 18417578 DOI: 10.1128/jvi.00163-08] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Epstein-Barr virus (EBV) infection is associated with many human malignancies. In vitro, EBV transforms primary B lymphocytes into continuously growing lymphoblastoid cell lines. EBV latent membrane protein 1 (LMP-1) is required for EBV transformation processes. Interferon regulatory factor 4 (IRF-4) is a transcription factor and has oncogenic potential. We find that high levels of IRF-4 are associated with EBV transformation of human primary B cells in vitro and with EBV type III latency in which LMP-1 is expressed. We show that EBV LMP-1 stimulates IRF-4 expression in B lymphocytes. The stimulation of IRF-4 by LMP-1 requires signaling from LMP-1 and involves cellular NF-kappaB. The growth of EBV-transformed cells is inhibited when IRF-4 is specifically down-regulated. We further demonstrate that IRF-4 knockdown cells have lower proliferation but higher apoptotic rates than control cells. Finally, IRF-4 is expressed in significant numbers of specimens of primary central nervous system (CNS) lymphomas (12/27 [44.4%]), an EBV-associated malignancy. The association between the expression levels of LMP-1 and IRF-4 is statistically significant (P = 0.011) in these CNS lymphomas. Our data suggest that IRF-4 may be a critical factor in EBV transformation and a useful target in the therapy of EBV-mediated neoplasia.
Collapse
|
249
|
Horiguchi H, Matsui-Horiguchi M, Sakata H, Ichinose M, Yamamoto T, Fujiwara M, Ohse H. Inflammatory pseudotumor-like follicular dendritic cell tumor of the spleen. Pathol Int 2008; 54:124-31. [PMID: 14720144 DOI: 10.1111/j.1440-1827.2004.01589.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A case of so-called inflammatory pseudotumor (IPT), occurring in the spleen of a 77-year-old woman, is reported. The spleen contained a well-circumscribed mass with central hemorrhage and necrosis. Histologically, spindle cells were dispersed in a background of abundant inflammatory cells, predominantly lymphocytes and plasma cells. The cells possessed enlarged, sometimes twisted or irregularly folded, nuclei that contained vesicular chromatin, and small but distinct, centrally located nucleoli. Immunohistochemically, the spindle cells were diffusely positive for vimentin, and focally positive for follicular dendritic cell (FDC) markers (Ber-MAC-DRC for CD35 and CNA.42). The Epstein-Barr virus (EBV) was exclusively detected in the spindle cells by in situ hybridization analysis. The cells also expressed the latent membrane protein-1 (LMP-1) of EBV, and polymerase chain reaction (PCR) analysis revealed that the LMP-1 gene had a 30-bp deletion and three point mutations, although their significance remains controversial. Inflammatory pseudotumor is a descriptive term that encompasses several different entities, and recent investigations have revealed the existence of neoplastic entities among IPT. One of the neoplastic IPT, recently designated 'IPT-like FDC tumor', is characterized by proliferation of EBV-positive FDC and commonly occurs in the liver and spleen. Because such tumors are capable of recurrence and metastasis, it is important to consider the possibility of an IPT-like FDC tumor when making a diagnosis of a hepatic/splenic IPT-like lesion.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/analysis
- Dendritic Cells, Follicular/metabolism
- Dendritic Cells, Follicular/pathology
- Dendritic Cells, Follicular/virology
- Epstein-Barr Virus Infections/complications
- Epstein-Barr Virus Infections/metabolism
- Epstein-Barr Virus Infections/pathology
- Female
- Granuloma, Plasma Cell/metabolism
- Granuloma, Plasma Cell/pathology
- Granuloma, Plasma Cell/virology
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/immunology
- Herpesvirus 4, Human/isolation & purification
- Humans
- Immunoenzyme Techniques
- In Situ Hybridization
- RNA, Viral/analysis
- Splenectomy
- Splenic Neoplasms/metabolism
- Splenic Neoplasms/pathology
- Splenic Neoplasms/virology
- Tomography, X-Ray Computed
- Vimentin/analysis
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/metabolism
Collapse
Affiliation(s)
- Hisashi Horiguchi
- Center for Medical Sciences, Ibaraki Prefectural University of Health Sciences, Inashiki, Japan.
| | | | | | | | | | | | | |
Collapse
|
250
|
Schneider F, Neugebauer J, Griese J, Liefold N, Kutz H, Briseño C, Kieser A. The viral oncoprotein LMP1 exploits TRADD for signaling by masking its apoptotic activity. PLoS Biol 2008; 6:e8. [PMID: 18198944 PMCID: PMC2174972 DOI: 10.1371/journal.pbio.0060008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Accepted: 12/04/2007] [Indexed: 01/14/2023] Open
Abstract
The tumor necrosis factor (TNF)-receptor 1–associated death domain protein (TRADD) mediates induction of apoptosis as well as activation of NF-κB by cellular TNF-receptor 1 (TNFR1). TRADD is also recruited by the latent membrane protein 1 (LMP1) oncoprotein of Epstein-Barr virus, but its role in LMP1 signaling has remained enigmatic. In human B lymphocytes, we have generated, to our knowledge, the first genetic knockout of TRADD to investigate TRADD's role in LMP1 signal transduction. Our data from TRADD-deficient cells demonstrate that TRADD is a critical signaling mediator of LMP1 that is required for LMP1 to recruit and activate I-κB kinase β (IKKβ). However, in contrast to TNFR1, LMP1-induced TRADD signaling does not induce apoptosis. Searching for the molecular basis for this observation, we characterized the 16 C-terminal amino acids of LMP1 as an autonomous and unique virus-derived TRADD-binding domain. Replacing the death domain of TNFR1 by LMP1′s TRADD-binding domain converts TNFR1 into a nonapoptotic receptor that activates NF-κB through a TRAF6-dependent pathway, like LMP1 but unlike wild-type TNFR1. Thus, the unique interaction of LMP1 with TRADD encodes the transforming phenotype of viral TRADD signaling and masks TRADD's pro-apoptotic function. For viral infection to succeed, viral proteins must interact with the cellular signaling machinery of its target cell. An oncoprotein encoded by the Epstein-Barr virus (EBV) called latent membrane protein 1 (LMP1) is a primary contributor to the transformation of human B cells by the virus and the development of EBV-associated B cell malignancies by recruiting signaling molecules provided by the host. One such molecule, the cellular adapter protein TRADD, is among the few direct interaction partners of LMP1. But because TRADD promotes cell death (apoptosis) in the cellular tumor necrosis factor-receptor 1 (TNFR1) signaling pathway, it seems counterintuitive that TRADD could play a role in LMP1 biology, since LMP1 promotes cell survival and proliferation. We provide genetic evidence that TRADD is critical for LMP1 to assemble its transforming signaling network. LMP1 requires TRADD to recruit and activate I-κB kinase β and, thus, to induce canonical NF-κB signaling. Simultaneously, LMP1 masks TRADD's pro-apoptotic activity. We show that LMP1 carries a unique and autonomous viral TRADD-binding domain, which dictates an unusual structure of the LMP1-TRADD complex and the nonapoptotic phenotype of TRADD signaling, irrespective of the receptor context in which this domain is located. Thus, DNA tumor viruses alter the functional properties of cellular signaling molecules to exploit them for their own purpose of cell transformation. A unique Epstein Barr virus-derived protein interaction domain uses the cellular death domain protein TRADD to assemble its transforming signaling complex and dictates a transferable nonapoptitic phenotype of TRADD signaling.
Collapse
Affiliation(s)
- Frank Schneider
- Department of Gene Vectors, GSF–National Research Center for Environment and Health, Munich, Germany
| | - Julia Neugebauer
- Department of Gene Vectors, GSF–National Research Center for Environment and Health, Munich, Germany
| | - Janine Griese
- Department of Gene Vectors, GSF–National Research Center for Environment and Health, Munich, Germany
| | - Nicola Liefold
- Department of Gene Vectors, GSF–National Research Center for Environment and Health, Munich, Germany
| | - Helmut Kutz
- Department of Gene Vectors, GSF–National Research Center for Environment and Health, Munich, Germany
| | - Cinthia Briseño
- Department of Gene Vectors, GSF–National Research Center for Environment and Health, Munich, Germany
| | - Arnd Kieser
- Department of Gene Vectors, GSF–National Research Center for Environment and Health, Munich, Germany
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|