1
|
New Look of EBV LMP1 Signaling Landscape. Cancers (Basel) 2021; 13:cancers13215451. [PMID: 34771613 PMCID: PMC8582580 DOI: 10.3390/cancers13215451] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/01/2021] [Accepted: 10/26/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Epstein-Barr Virus (EBV) infection is associated with various lymphomas and carcinomas as well as other diseases in humans. The transmembrane protein LMP1 plays versatile roles in EBV life cycle and pathogenesis, by perturbing, reprograming, and regulating a large range of host cellular mechanisms and functions, which have been increasingly disclosed but not fully understood so far. We summarize recent research progress on LMP1 signaling, including the novel components LIMD1, p62, and LUBAC in LMP1 signalosome and LMP1 novel functions, such as its induction of p62-mediated selective autophagy, regulation of metabolism, induction of extracellular vehicles, and activation of NRF2-mediated antioxidative defense. A comprehensive understanding of LMP1 signal transduction and functions may allow us to leverage these LMP1-regulated cellular mechanisms for clinical purposes. Abstract The Epstein–Barr Virus (EBV) principal oncoprotein Latent Membrane Protein 1 (LMP1) is a member of the Tumor Necrosis Factor Receptor (TNFR) superfamily with constitutive activity. LMP1 shares many features with Pathogen Recognition Receptors (PRRs), including the use of TRAFs, adaptors, and kinase cascades, for signal transduction leading to the activation of NFκB, AP1, and Akt, as well as a subset of IRFs and likely the master antioxidative transcription factor NRF2, which we have gradually added to the list. In recent years, we have discovered the Linear UBiquitin Assembly Complex (LUBAC), the adaptor protein LIMD1, and the ubiquitin sensor and signaling hub p62, as novel components of LMP1 signalosome. Functionally, LMP1 is a pleiotropic factor that reprograms, balances, and perturbs a large spectrum of cellular mechanisms, including the ubiquitin machinery, metabolism, epigenetics, DNA damage response, extracellular vehicles, immune defenses, and telomere elongation, to promote oncogenic transformation, cell proliferation and survival, anchorage-independent cell growth, angiogenesis, and metastasis and invasion, as well as the development of the tumor microenvironment. We have recently shown that LMP1 induces p62-mediated selective autophagy in EBV latency, at least by contributing to the induction of p62 expression, and Reactive Oxygen Species (ROS) production. We have also been collecting evidence supporting the hypothesis that LMP1 activates the Keap1-NRF2 pathway, which serves as the key antioxidative defense mechanism. Last but not least, our preliminary data shows that LMP1 is associated with the deregulation of cGAS-STING DNA sensing pathway in EBV latency. A comprehensive understanding of the LMP1 signaling landscape is essential for identifying potential targets for the development of novel strategies towards targeted therapeutic applications.
Collapse
|
2
|
Zhang ZW, Zhang HL, Yu YH, Ouyang YM, Chen ZC, He XS, He ZM. Carboxyl terminal activating region 3 of latent membrane protein 1 encoded by the Epstein‑Barr virus regulates cell proliferation and protein expression in NP69 cells. Mol Med Rep 2019; 21:720-730. [PMID: 31974609 PMCID: PMC6947836 DOI: 10.3892/mmr.2019.10859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 04/23/2018] [Indexed: 01/14/2023] Open
Abstract
In the present study, the mechanism by which carboxyl terminal activating region 3 (CTAR3) of latent membrane protein 1 (LMP1), encoded by the Epstein-Barr virus, regulated cell proliferation and protein expression was investigated in the nasopharyngeal epithelial cell line NP69. The deletion mutant LMP1 (LMP1Δ232-351; amino acid residues including 232–351 codons in CTAR3 deleted) was generated by polymerase chain reaction. An NP69-LMP1Δ232-351 cell line was established by retroviral infection. Finally, cell proliferation and protein expression of NP69 cells expressing LMP1Δ232-351 were examined using a cell growth curve and western blot analysis. The results demonstrated: i) The proliferation of NP69-LMP1Δ232-351 cells was significantly decreased compared with cells expressing wild type LMP1 (LMP1WT; n=3; P<0.05); ii) 17 proteins exhibited differential protein expression (>2-fold change) in NP69-LMP1Δ232-351 cells compared with NP69-LMP1WT cells; and iii) LMP1WT was involved in activating the Janus kinase 3 (JAK3) promoter and regulating the expression of JAK3 protein, while LMP1Δ232-351 was almost defective in ability to activate the JAK promoter. These results suggested that LMP1-CTAR3 may be an important functional domain for regulating cell proliferation and protein expression in nasopharyngeal epithelial cells.
Collapse
Affiliation(s)
- Zhi-Wei Zhang
- Cancer Research Institute of Medical College, University of South China, Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan, Hengyang, Hunan 421001, P.R. China
| | - He-Liang Zhang
- Cancer Research Institute of Medical College, University of South China, Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan, Hengyang, Hunan 421001, P.R. China
| | - Yan-Hui Yu
- Cancer Research Institute, Central South University, Xiangya School of Medicine, Changsha, Hunan 410078, P.R. China
| | - Yong-Mei Ouyang
- Cancer Research Institute, Central South University, Xiangya School of Medicine, Changsha, Hunan 410078, P.R. China
| | - Zhu-Chu Chen
- Cancer Research Institute, Central South University, Xiangya School of Medicine, Changsha, Hunan 410078, P.R. China
| | - Xiu-Sheng He
- Cancer Research Institute of Medical College, University of South China, Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan, Hengyang, Hunan 421001, P.R. China
| | - Zhi-Min He
- Cancer Research Institute, Central South University, Xiangya School of Medicine, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
3
|
Wang J, Zheng X, Qin Z, Wei L, Lu Y, Peng Q, Gao Y, Zhang X, Zhang X, Li Z, Fu Y, Liu P, Liu C, Yan Q, Xiong W, Li G, Lu J, Ma J. Epstein-Barr virus miR-BART3-3p promotes tumorigenesis by regulating the senescence pathway in gastric cancer. J Biol Chem 2019; 294:4854-4866. [PMID: 30674552 DOI: 10.1074/jbc.ra118.006853] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/14/2019] [Indexed: 12/25/2022] Open
Abstract
Epstein-Barr virus-associated gastric cancer (EBVaGC) accounts for about 10% of all gastric cancer cases and has unique pathological and molecular characteristics. EBV encodes a large number of microRNAs, which actively participate in the development of EBV-related tumors. Here, we report that EBV-miR-BART3-3p (BART3-3p) promotes gastric cancer cell growth in vitro and in vivo Moreover, BART3-3p inhibits the senescence of gastric cancer cells induced by an oncogene (RASG12V) or chemotherapy (irinotecan). LMP1 and EBNA3C encoded by EBV have also been reported to have antisenescence effects; however, in EBVaGC specimens, LMP1 expression is very low, and EBNA3C is not expressed. BART3-3p inhibits senescence of gastric cancer cells in a nude mouse model and inhibits the infiltration of natural killer cells and macrophages in tumor by altering the senescence-associated secretory phenotype (SASP). Mechanistically, BART3-3p directly targeted the tumor suppressor gene TP53 and caused down-regulation of p53's downstream target, p21. Analysis from clinical EBVaGC samples also showed a negative correlation between BART3-3p and TP53 expression. It is well known that mutant oncogene RASG12V or chemotherapeutic drugs can induce senescence, and here we show that both RASG12V and a chemotherapy drug also can induce BART3-3p expression in EBV-positive gastric cancer cells, forming a feedback loop that keeps the EBVaGC senescence at a low level. Our results suggest that, although TP53 is seldom mutated in EBVaGC, its expression is finely regulated such that EBV-encoded BART3-3p may play an important role by inhibiting the senescence of gastric cancer cells.
Collapse
Affiliation(s)
- Jia Wang
- From the Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China.,Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China.,Department of Immunology, Changzhi Medical College, Changzhi, Shanxi 046000, China
| | - Xiang Zheng
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China.,Hunan Key Laboratory of Translational Radiation Oncology, Changsha 410013, China
| | - Zailong Qin
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Changsha 410013, China
| | - Lingyu Wei
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China.,National Health Commission Key Laboratory of Carcinogenesis (Central South University), Changsha 410078, China
| | - Yuanjun Lu
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha 410078, China, and
| | - Qiu Peng
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Yingxue Gao
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Xuemei Zhang
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Xiaoyue Zhang
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Zhengshuo Li
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Yuxin Fu
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Peishan Liu
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Can Liu
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Qun Yan
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Wei Xiong
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China.,National Health Commission Key Laboratory of Carcinogenesis (Central South University), Changsha 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha 410078, China, and
| | - Guiyuan Li
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China.,National Health Commission Key Laboratory of Carcinogenesis (Central South University), Changsha 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha 410078, China, and
| | - Jianhong Lu
- Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China,
| | - Jian Ma
- From the Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China, .,Cancer Research Institute, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Changsha 410013, China.,National Health Commission Key Laboratory of Carcinogenesis (Central South University), Changsha 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha 410078, China, and
| |
Collapse
|
4
|
BZLF1 Attenuates Transmission of Inflammatory Paracrine Senescence in Epstein-Barr Virus-Infected Cells by Downregulating Tumor Necrosis Factor Alpha. J Virol 2016; 90:7880-93. [PMID: 27334596 DOI: 10.1128/jvi.00999-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/16/2016] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Recent studies have shown that inflammatory responses trigger and transmit senescence to neighboring cells and activate the senescence-associated secretory phenotype (SASP). Latent Epstein-Barr virus (EBV) infection induces increased secretion of several inflammatory factors, whereas lytic infections evade the antiviral inflammatory response. However, the changes in and roles of the inflammatory microenvironment during the switch between EBV life cycles remain unknown. In the present study, we demonstrate that latent EBV infection in EBV-positive cells triggers the SASP in neighboring epithelial cells. In contrast, lytic EBV infection abolishes this phenotype. BZLF1 attenuates the transmission of paracrine senescence during lytic EBV infection by downregulating tumor necrosis factor alpha (TNF-α) secretion. A mutant BZLF1 protein, BZLF1Δ207-210, that cannot inhibit TNF-α secretion while maintaining viral transcription, fails to block paracrine senescence, whereas a neutralizing antibody against TNF-α is sufficient to restore its inhibition. Furthermore, latent EBV infection induces oxidative stress in neighboring cells, while BZLF1-mediated downregulation of TNF-α reduces reactive oxygen species (ROS) levels in neighboring cells, and ROS scavengers alleviate paracrine senescence. These results suggest that lytic EBV infection attenuates the transmission of inflammatory paracrine senescence through BZLF1 downregulation of TNF-α secretion and alters the inflammatory microenvironment to allow virus propagation and persistence. IMPORTANCE The senescence-associated secretory phenotype (SASP), an important tumorigenic process, is triggered and transmitted by inflammatory factors. The different life cycles of Epstein-Barr virus (EBV) infection in EBV-positive cells employ distinct strategies to modulate the inflammatory response and senescence. The elevation of inflammatory factors during latent EBV infection promotes the SASP in uninfected cells. In contrast, during the viral lytic cycle, BZLF1 suppresses the production of TNF-α, resulting in the attenuation of paracrine inflammatory senescence. This finding indicates that EBV evades inflammatory senescence during lytic infection and switches from facilitating tumor-promoting SASP to generating a virus-propagating microenvironment, thereby facilitating viral spread in EBV-associated diseases.
Collapse
|
5
|
Weiland T, Lampe J, Essmann F, Venturelli S, Berger A, Bossow S, Berchtold S, Schulze-Osthoff K, Lauer UM, Bitzer M. Enhanced killing of therapy-induced senescent tumor cells by oncolytic measles vaccine viruses. Int J Cancer 2013; 134:235-43. [PMID: 23797800 DOI: 10.1002/ijc.28350] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 06/07/2013] [Indexed: 01/07/2023]
Abstract
Therapy-induced senescence (TIS) as a permanent growth arrest can be induced by various stimuli, including anticancer compounds. TIS emerged as a promising strategy to overcome resistance phenomena. However, senescent cancer cells might regain proliferation activity in vivo or even secrete tumor-promoting cytokines. Therefore, successful exploitation of TIS in cancer treatment simultaneously requires the development of effective strategies to eliminate senescent cancer cells. Virotherapy aims to selectively hit tumor cells, thus a combination with senescence-inducing drugs was explored. As a model, we chose measles vaccine virus (MeV), which does not interfere with cellular senescence by itself. In different tumor cell types, such as hepatoma, pancreatic and mammary gland carcinoma, we demonstrate efficient viral replication and lysis after TIS by gemcitabine, doxorubicin or taxol. Applying real time imaging, we even found an accelerated lysis of senescent cancer cells, supporting an enhanced viral replication with an increase in cell-associated and released infectious MeV particles. In summary, we show as a proof-of-concept that senescent tumor cells can be efficiently exploited as virus host cells by oncolytic MeV. These observations open up a new field for preclinical and clinical research to further investigate TIS and oncolytic viruses as an attractive combinatorial future treatment approach.
Collapse
Affiliation(s)
- Timo Weiland
- Department of Internal Medicine I, Medical University Hospital, University of Tübingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
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: 25] [Impact Index Per Article: 1.6] [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
|
7
|
Lin X, Liu S, Luo X, Ma X, Guo L, Li L, Li Z, Tao Y, Cao Y. EBV-encoded LMP1 regulates Op18/stathmin signaling pathway by cdc2 mediation in nasopharyngeal carcinoma cells. Int J Cancer 2009; 124:1020-7. [PMID: 19048596 DOI: 10.1002/ijc.23767] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Oncoprotein 18/stathmin (Op18/stathmin) plays a crucial role in maintaining cell biological characteristics by regulating microtubule dynamics, especially entry into mitosis; phosphorylated Op18/stathmin promotes microtubule polymerization to form the mitotic spindle, which is essential for chromosome segregation and cell division. Cdc2 is a critical kinase in starting M phase events in cell-cycle progression and is a positive regulator of the cell cycle. Latent membrane protein 1 (LMP1) is an Epstein-Barr virus (EBV)-encoded oncogenic protein that is able to induce carcinogenesis via various signaling pathways. This study focused on regulation by LMP1 of Op18/stathmin signaling in nasopharyngeal carcinoma (NPC) cells and showed that LMP1 regulates Op18/stathmin signaling through cdc2 mediation, LMP1 upregulates cdc2 kinase activity, and Op18/stathmin phosphorylation promotes the interaction of cdc2 with Op18/stathmin and microtubule polymerization during mitosis, and inhibition of LMP1 expression attenuates the interaction of cdc2 and Op18/stathmin and promotes microtubule depolymerization. These results reveal a new pathway via which LMP1 regulates Op18/stathmin signaling by cdc2 mediation; this new signaling pathway not only perfects the LMP1 regulation network but also elucidates the molecular mechanism of LMP1 that leads to carcinogenesis.
Collapse
Affiliation(s)
- Xuechi Lin
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Zhang Q, Zhang Z, Wang C, Xiao Z, Yu Y, Yang F, Chen Z, He Z. Proteome analysis of the transformation potential of the Epstein-Barr virus-encoded latent membrane protein 1 in nasopharyngeal epithelial cells NP69. Mol Cell Biochem 2008; 314:73-83. [PMID: 18425422 DOI: 10.1007/s11010-008-9767-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2007] [Accepted: 04/07/2008] [Indexed: 11/28/2022]
Abstract
Latent membrane protein 1 (LMP1) of Epstein-Barr virus has been identified to be crucial in inducing cell transformation. However, the mechanism of LMP1-mediated epithelial cell transformation remains unclear. In this study, nasopharyngeal epithelial cells NP69 were infected with retrovirus with gene encoding wild type LMP1 or mutational LMP1 defective in binding to tumor necrosis factor receptor-associated death domain (TRADD). The NP69-LMP1(TRADD) lost some malignant phenotypes compared with the NP69-LMP1(WT). We performed proteomic approach to gain the differential protein expression profile associated with LMP1-mediated epithelial cell transformation. Furthermore, the differential expressional levels of partial identified proteins were confirmed by Western blot and real-time RT-PCR. Some were known to be related to the development of LMP1-induced transformation, and some were new LMP1-associated proteins. These data are valuable for further study of the mechanism of LMP1 in human nasopharyngeal carcinoma and provide some new clues for investigating other LMP1-associated tumors.
Collapse
Affiliation(s)
- Qiong Zhang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Ding L, Li L, Yang J, Zhou S, Li W, Tang M, Shi Y, Yi W, Cao Y. Latent membrane protein 1 encoded by Epstein-Barr virus induces telomerase activity via p16INK4A/Rb/E2F1 and JNK signaling pathways. J Med Virol 2007; 79:1153-63. [PMID: 17597480 DOI: 10.1002/jmv.20896] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Elevated telomerase activity is observed in about 90% of human cancers. This activity correlates strictly with human telomerase reverse transcriptase (hTERT). Previously, it was shown that the Epstein-Barr virus-encoded latent membrane protein 1 (LMP1) induced telomerase activity in nasopharyngeal carcinoma cells. In this study, it was indicated that LMP1 inhibited p16(INK4A) expression, promoted phosphorylation of p105 Rb and upregulated E2F1 expression as well as transactivation, and overexpression of E2F1 alone was sufficient to upregulate telomerase activity. The JNK kinase cascade could also promote telomerase activity modulated by LMP1, that inhibition of JNK by JIP and TAM 67 dominant negative mutant abrogated telomerase activity. The data show that p16(INK4A)/Rb/E2F1 and JNK signaling pathways are involved in the regulation of telomerase activity via LMP1. The present study provides new perspectives on carcinogenesis of nasopharyngeal carcinoma that may be exploited for novel therapeutic strategies.
Collapse
Affiliation(s)
- Lin Ding
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Brinkmann MM, Schulz TF. Regulation of intracellular signalling by the terminal membrane proteins of members of the Gammaherpesvirinae. J Gen Virol 2006; 87:1047-1074. [PMID: 16603506 DOI: 10.1099/vir.0.81598-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The human gamma(1)-herpesvirus Epstein-Barr virus (EBV) and the gamma(2)-herpesviruses Kaposi's sarcoma-associated herpesvirus (KSHV), rhesus rhadinovirus (RRV), herpesvirus saimiri (HVS) and herpesvirus ateles (HVA) all contain genes located adjacent to the terminal-repeat region of their genomes, encoding membrane proteins involved in signal transduction. Designated 'terminal membrane proteins' (TMPs) because of their localization in the viral genome, they interact with a variety of cellular signalling molecules, such as non-receptor protein tyrosine kinases, tumour-necrosis factor receptor-associated factors, Ras and Janus kinase (JAK), thereby initiating further downstream signalling cascades, such as the MAPK, PI3K/Akt, NF-kappaB and JAK/STAT pathways. In the case of TMPs expressed during latent persistence of EBV and HVS (LMP1, LMP2A, Stp and Tip), their modulation of intracellular signalling pathways has been linked to the provision of survival signals to latently infected cells and, hence, a contribution to occasional cellular transformation. In contrast, activation of similar pathways by TMPs of KSHV (K1 and K15) and RRV (R1), expressed during lytic replication, may extend the lifespan of virus-producing cells, alter their migration and/or modulate antiviral immune responses. Whether R1 and K1 contribute to the oncogenic properties of KSHV and RRV has not been established satisfactorily, despite their transforming qualities in experimental settings.
Collapse
Affiliation(s)
- Melanie M Brinkmann
- Institut für Virologie, Medizinische Hochschule Hannover, Carl-Neuberg Str. 1, D-30625 Hannover, Germany
| | - Thomas F Schulz
- Institut für Virologie, Medizinische Hochschule Hannover, Carl-Neuberg Str. 1, D-30625 Hannover, Germany
| |
Collapse
|
11
|
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
|
12
|
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: 1.6] [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
|
13
|
Li HP, Chang YS. Epstein-Barr virus latent membrane protein 1: structure and functions. J Biomed Sci 2003; 10:490-504. [PMID: 12928589 DOI: 10.1007/bf02256110] [Citation(s) in RCA: 149] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2003] [Accepted: 05/22/2003] [Indexed: 12/22/2022] Open
Abstract
The Epstein-Barr virus latent membrane protein (LMP) 1 is a versatile protein that has profound effects on target cells through its effect on constitutive cellular proteins, e.g. TRAFs, TRADD, RIP, JAK3, BRAM1, and p85. LMP1 can stimulate or inhibit signaling pathways, resulting in transformation of rodent fibroblast cell lines, blockade of differentiation in epithelial cells, upregulation of anti-apoptotic proteins, production of cytokines, upregulation of cell surface markers, upregulation of DNA methyltransferase activity, and downregulation of cell adhesion molecules and cyclin-dependent kinases. Overall, this results in greater transformation and survival in LMP1-expressing cells. Within nasopharyngeal carcinoma biopsy tissues, a naturally occurring LMP1 variant has been identified as having a 10-amino acid deletion in the C-terminus that seems to confer greater transformation potential than non-deleted LMP1. The role of LMP1 as a viral oncogene and its interaction with cellular factors are discussed.
Collapse
Affiliation(s)
- Hsin-Pai Li
- Graduate Institute of Basic Medical Sciences, Chang Gung University, Taoyuan, Taiwan, ROC
| | | |
Collapse
|
14
|
Ohtani N, Brennan P, Gaubatz S, Sanij E, Hertzog P, Wolvetang E, Ghysdael J, Rowe M, Hara E. Epstein-Barr virus LMP1 blocks p16INK4a-RB pathway by promoting nuclear export of E2F4/5. J Cell Biol 2003; 162:173-83. [PMID: 12860972 PMCID: PMC2172795 DOI: 10.1083/jcb.200302085] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2003] [Revised: 05/08/2003] [Accepted: 06/03/2003] [Indexed: 12/13/2022] Open
Abstract
The p16INK4a-RB pathway plays a critical role in preventing inappropriate cell proliferation and is often targeted by viral oncoproteins during immortalization. Latent membrane protein 1 (LMP1) of Epstein-Barr virus (EBV) is often present in EBV-associated proliferative diseases and is critical for the immortalizing and transforming activity of EBV. Unlike other DNA tumor virus oncoproteins, which possess immortalizing activity, LMP1 does not bind to retinoblastoma tumor suppressor protein, but instead blocks the expression of p16INK4a tumor suppressor gene. However, it has been unclear how LMP1 represses the p16INK4a gene expression. Here, we report that LMP1 promotes the CRM1-dependent nuclear export of Ets2, which is an important transcription factor for p16INK4a gene expression, thereby reducing the level of p16INK4a expression. We further demonstrate that LMP1 also blocks the function of E2F4 and E2F5 (E2F4/5) transcription factors through promoting their nuclear export in a CRM1-dependent manner. As E2F4/5 are essential downstream mediators for a p16INK4a-induced cell cycle arrest, these results indicate that the action of LMP1 on nuclear export has two effects on the p16INK4a-RB pathway: (1) repression of p16INK4a expression and (2) blocking the downstream mediator of the p16INK4a-RB pathway. These results reveal a novel activity of LMP1 and increase an understanding of how viral oncoproteins perturb the p16INK4a-RB pathway.
Collapse
Affiliation(s)
- Naoko Ohtani
- Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester M20 4BX, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
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: 2.2] [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
|
16
|
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
|
17
|
Tsao SW, Tramoutanis G, Dawson CW, Lo AKF, Huang DP. The significance of LMP1 expression in nasopharyngeal carcinoma. Semin Cancer Biol 2002; 12:473-87. [PMID: 12450733 DOI: 10.1016/s1044579x02000901] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Epstein-Barr virus (EBV)-encoded latent membrane protein 1 (LMP1) is a key effector of EBV-mediated B cell transformation. LMP1 displays potent oncogenic properties in rodent fibroblasts, and induces a wide range of effects in B cells and epithelial cells. LMP1 functions as a constitutively active tumor necrosis factor receptor (TNFR) engaging a multitude of signaling pathways that include NF-kappaB, the mitogen-activated protein kinases (MAPKs), JNK, p38, the JAK/STAT pathway and, more recently, the small Rho GTPases. The constitutive activation of these signaling cascades explains LMP1's ability to induce such a diverse array of morphological and phenotypic effects in cells and provides an insight into how LMP1 may induce cell transformation. The frequent expression of LMP1 in undifferentiated nasopharyngeal carcinoma (NPC) points to a role for this viral oncoprotein as a key effector molecule in NPC pathogenesis.
Collapse
Affiliation(s)
- Sai Wah Tsao
- Department of Anatomy, Faculty of Medicine, University of Hong Kong, Hong Kong, PR China.
| | | | | | | | | |
Collapse
|