51
|
EBNA1: Oncogenic Activity, Immune Evasion and Biochemical Functions Provide Targets for Novel Therapeutic Strategies against Epstein-Barr Virus- Associated Cancers. Cancers (Basel) 2018; 10:cancers10040109. [PMID: 29642420 PMCID: PMC5923364 DOI: 10.3390/cancers10040109] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 03/26/2018] [Accepted: 03/29/2018] [Indexed: 12/12/2022] Open
Abstract
The presence of the Epstein-Barr virus (EBV)-encoded nuclear antigen-1 (EBNA1) protein in all EBV-carrying tumours constitutes a marker that distinguishes the virus-associated cancer cells from normal cells and thereby offers opportunities for targeted therapeutic intervention. EBNA1 is essential for viral genome maintenance and also for controlling viral gene expression and without EBNA1, the virus cannot persist. EBNA1 itself has been linked to cell transformation but the underlying mechanism of its oncogenic activity has been unclear. However, recent data are starting to shed light on its growth-promoting pathways, suggesting that targeting EBNA1 can have a direct growth suppressing effect. In order to carry out its tasks, EBNA1 interacts with cellular factors and these interactions are potential therapeutic targets, where the aim would be to cripple the virus and thereby rid the tumour cells of any oncogenic activity related to the virus. Another strategy to target EBNA1 is to interfere with its expression. Controlling the rate of EBNA1 synthesis is critical for the virus to maintain a sufficient level to support viral functions, while at the same time, restricting expression is equally important to prevent the immune system from detecting and destroying EBNA1-positive cells. To achieve this balance EBNA1 has evolved a unique repeat sequence of glycines and alanines that controls its own rate of mRNA translation. As the underlying molecular mechanisms for how this repeat suppresses its own rate of synthesis in cis are starting to be better understood, new therapeutic strategies are emerging that aim to modulate the translation of the EBNA1 mRNA. If translation is induced, it could increase the amount of EBNA1-derived antigenic peptides that are presented to the major histocompatibility (MHC) class I pathway and thus, make EBV-carrying cancers better targets for the immune system. If translation is further suppressed, this would provide another means to cripple the virus.
Collapse
|
52
|
Zhang Y, Wang H, Liu Y, Wang C, Wang J, Long C, Guo W, Sun X. Baicalein inhibits growth of Epstein-Barr virus-positive nasopharyngeal carcinoma by repressing the activity of EBNA1 Q-promoter. Biomed Pharmacother 2018; 102:1003-1014. [PMID: 29710517 DOI: 10.1016/j.biopha.2018.03.114] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 03/19/2018] [Accepted: 03/19/2018] [Indexed: 12/12/2022] Open
Abstract
Epstein-Barr virus (EBV) can establish a life-long latent infection in the host and is associated with various human malignancies, including nasopharyngeal carcinoma (NPC), the most common cancer originated from nasopharynx. EBV nuclear antigen 1 (EBNA1) is the only viral protein absolutely demanded for segregation, replication, transcription and maintenance of EBV viral genome in host cells. Baicalein, a bioactive flavonoid compound purified from the root of Scutellariae baicaleinsis, displays anti-inflammatory, immunosuppressive, and anti-tumor properties. In this study, the therapeutic effects and functional mechanism of baicalein on EBV-positive human NPC were determined. Cell Counting Kit-8 assays and cell formation colony were performed to investigate that baicalein can suppress proliferation of EBV-infected human NPC cells. Flow cytometric and hoechst 33258 staining results indicated that baicalein induced cell cycle arrest and apoptosis. Western blotting results demonstrated that baicalein down-regulates EBNA1 expression but not reduces the stability and half-life of EBNA1 in EBV-infected NPC cells. Additionally, the mRNA level of EBNA1 was examined by real time-PCR, the activity of EBNA1 Q promoter (Qp) was determined by dual luciferase reporter assay. Considering that transcription factor specificity protein 1 (Sp1) can maintain EBNA1 Qp active. Further analyses also elucidated that baicalein inhibits the expression of Sp1 while knock-down Sp1 by specific shRNAs decreases the expression and transcription levels of EBNA1. Therefore, the results suggested that baicalein may decrease EBNA1 expression level in EBV-positive NPC cells via inhibiting the activity of EBNA1 Q-promoter while over-expression of EBNA1 attenuate the inhibitory effect of baicalein. Finally, it was found that baicalein may strongly reduce growth of tumor in the mouse xenograft model of EBV-positive NPC. These results indicated that baicalein inhibits growth of EBV-positive NPC by repressing the activity of EBNA1 Q-promoter. Baicalein may be used as a therapeutic agent to treat EBV-positive NPC.
Collapse
Affiliation(s)
- Yaqian Zhang
- Stat Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, PR China
| | - Huan Wang
- Stat Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, PR China
| | - Yu Liu
- Stat Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, PR China
| | - Chao Wang
- Stat Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, PR China
| | - Jingchao Wang
- Stat Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, PR China
| | - Cong Long
- Stat Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, PR China
| | - Wei Guo
- Department of Pathology and Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, PR China
| | - Xiaoping Sun
- Stat Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, PR China; State Key Laboratory of Virology, Wuhan University, Wuhan, 430072, PR China.
| |
Collapse
|
53
|
Dheekollu J, Malecka K, Wiedmer A, Delecluse HJ, Chiang AKS, Altieri DC, Messick TE, Lieberman PM. Carcinoma-risk variant of EBNA1 deregulates Epstein-Barr Virus episomal latency. Oncotarget 2018; 8:7248-7264. [PMID: 28077791 PMCID: PMC5352318 DOI: 10.18632/oncotarget.14540] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 12/26/2016] [Indexed: 12/27/2022] Open
Abstract
Epstein-Barr Virus (EBV) latent infection is a causative co-factor for endemic Nasopharyngeal Carcinoma (NPC). NPC-associated variants have been identified in EBV-encoded nuclear antigen EBNA1. Here, we solve the X-ray crystal structure of an NPC-derived EBNA1 DNA binding domain (DBD) and show that variant amino acids are found on the surface away from the DNA binding interface. We show that NPC-derived EBNA1 is compromised for DNA replication and episome maintenance functions. Recombinant virus containing the NPC EBNA1 DBD are impaired in their ability to immortalize primary B-lymphocytes and suppress lytic transcription during early stages of B-cell infection. We identify Survivin as a host protein deficiently bound by the NPC variant of EBNA1 and show that Survivin depletion compromises EBV episome maintenance in multiple cell types. We propose that endemic variants of EBNA1 play a significant role in EBV-driven carcinogenesis by altering key regulatory interactions that destabilize latent infection.
Collapse
Affiliation(s)
| | | | | | | | - Alan K S Chiang
- Department of Pediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | | | | | | |
Collapse
|
54
|
Sima J, Bartlett DA, Gordon MR, Gilbert DM. Bacterial artificial chromosomes establish replication timing and sub-nuclear compartment de novo as extra-chromosomal vectors. Nucleic Acids Res 2018; 46:1810-1820. [PMID: 29294101 PMCID: PMC5829748 DOI: 10.1093/nar/gkx1265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/27/2017] [Accepted: 12/06/2017] [Indexed: 12/11/2022] Open
Abstract
The role of DNA sequence in determining replication timing (RT) and chromatin higher order organization remains elusive. To address this question, we have developed an extra-chromosomal replication system (E-BACs) consisting of ∼200 kb human bacterial artificial chromosomes (BACs) modified with Epstein-Barr virus (EBV) stable segregation elements. E-BACs were stably maintained as autonomous mini-chromosomes in EBNA1-expressing HeLa or human induced pluripotent stem cells (hiPSCs) and established distinct RT patterns. An E-BAC harboring an early replicating chromosomal region replicated early during S phase, while E-BACs derived from RT transition regions (TTRs) and late replicating regions replicated in mid to late S phase. Analysis of E-BAC interactions with cellular chromatin (4C-seq) revealed that the early replicating E-BAC interacted broadly throughout the genome and preferentially with the early replicating compartment of the nucleus. In contrast, mid- to late-replicating E-BACs interacted with more specific late replicating chromosomal segments, some of which were shared between different E-BACs. Together, we describe a versatile system in which to study the structure and function of chromosomal segments that are stably maintained separately from the influence of cellular chromosome context.
Collapse
Affiliation(s)
- Jiao Sima
- Department of Biological Science, 319 Stadium Drive, Florida State University, Tallahassee, FL 32306, USA
| | - Daniel A Bartlett
- Department of Biological Science, 319 Stadium Drive, Florida State University, Tallahassee, FL 32306, USA
| | - Molly R Gordon
- Department of Biological Science, 319 Stadium Drive, Florida State University, Tallahassee, FL 32306, USA
| | - David M Gilbert
- Department of Biological Science, 319 Stadium Drive, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
55
|
Chen X, Gao B, Ponnusamy M, Lin Z, Liu J. MEF2 signaling and human diseases. Oncotarget 2017; 8:112152-112165. [PMID: 29340119 PMCID: PMC5762387 DOI: 10.18632/oncotarget.22899] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 09/09/2017] [Indexed: 01/01/2023] Open
Abstract
The members of myocyte Enhancer Factor 2 (MEF2) protein family was previously believed to function in the development of heart and muscle. Recent reports indicate that they are also closely associated with development and progression of many human diseases. Although their role in cancer biology is well established, the molecular mechanisms underlying their action is yet largely unknown. MEF2 family is closely associated with various signaling pathways, including Ca2+ signaling, MAP kinase signaling, Wnt signaling, PI3K/Akt signaling, etc. microRNAs also contribute to regulate the activities of MEF2. In this review, we summarize the known molecular mechanism by which MEF2 family contribute to human diseases.
Collapse
Affiliation(s)
- Xiao Chen
- School of Pharmacy, Qingdao University, Qingdao 266021, China.,Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Bing Gao
- School of Pharmacy, Qingdao University, Qingdao 266021, China.,School of Basic Medicine, Qingdao University, Qingdao 266021, China
| | - Murugavel Ponnusamy
- Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Zhijuan Lin
- Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Jia Liu
- School of Pharmacy, Qingdao University, Qingdao 266021, China.,School of Basic Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
56
|
Shannon-Lowe C, Rickinson AB, Bell AI. Epstein-Barr virus-associated lymphomas. Philos Trans R Soc Lond B Biol Sci 2017; 372:20160271. [PMID: 28893938 PMCID: PMC5597738 DOI: 10.1098/rstb.2016.0271] [Citation(s) in RCA: 247] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2017] [Indexed: 02/06/2023] Open
Abstract
Epstein-Barr virus (EBV), originally discovered through its association with Burkitt lymphoma, is now aetiologically linked to a remarkably wide range of lymphoproliferative lesions and malignant lymphomas of B-, T- and NK-cell origin. Some occur as rare accidents of virus persistence in the B lymphoid system, while others arise as a result of viral entry into unnatural target cells. The early finding that EBV is a potent B-cell growth transforming agent hinted at a simple oncogenic mechanism by which this virus could promote lymphomagenesis. In reality, the pathogenesis of EBV-associated lymphomas involves a complex interplay between different patterns of viral gene expression and cellular genetic changes. Here we review recent developments in our understanding of EBV-associated lymphomagenesis in both the immunocompetent and immunocompromised host.This article is part of the themed issue 'Human oncogenic viruses'.
Collapse
Affiliation(s)
- Claire Shannon-Lowe
- Institute of Immunology and Immunotherapy, The Medical School, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Alan B Rickinson
- Institute of Immunology and Immunotherapy, The Medical School, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Andrew I Bell
- Institute for Cancer and Genomic Sciences, The Medical School, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
57
|
Mine S, Hishima T, Suganuma A, Fukumoto H, Sato Y, Kataoka M, Sekizuka T, Kuroda M, Suzuki T, Hasegawa H, Fukayama M, Katano H. Interleukin-6-dependent growth in a newly established plasmablastic lymphoma cell line and its therapeutic targets. Sci Rep 2017; 7:10188. [PMID: 28860565 PMCID: PMC5579229 DOI: 10.1038/s41598-017-10684-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/14/2017] [Indexed: 12/26/2022] Open
Abstract
Plasmablastic lymphoma (PBL) is a rare, highly aggressive subtype of non-Hodgkin lymphoma with plasma-cell differentiation occurring typically in immune-suppressed patients such as those with AIDS. This study reports the establishment and characterization of a new cell line, PBL-1, derived from a patient with AIDS-associated PBL. Morphological assessment of PBL-1 indicated plasma-cell differentiation with a CD20(-) CD38(+) CD138(+) immunophenotype and IgH/c-myc translocation. The cell line harbours Epstein-Barr virus, but a 52.7-kbp length defect was identified in its genome, resulting in no expression of viral microRNAs encoded in the BamHI-A Rightward Transcript region. Importantly, supplementation of culture medium with >5 ng/mL of interleukin-6 (IL-6) was required for PBL-1 growth. Starvation of IL-6 or addition of tocilizumab, an inhibitory antibody for the IL-6 receptor, induced apoptosis of PBL-1. Transduction of IL-6 into PBL-1 by lentivirus vector induced autologous growth without IL-6 supplementation of culture medium. These data indicate the IL-6 dependency of PBL-1 for proliferation and survival. mTOR inhibitors induced cell death effectively, suggesting mTOR in the IL-6 signalling pathway is a potential therapeutic target for PBL. This established PBL cell line will be a useful tool to further understand the pathophysiology of PBL and aid the future development of PBL treatment.
Collapse
Affiliation(s)
- Sohtaro Mine
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tsunekazu Hishima
- Department of Pathology, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Akihiko Suganuma
- Department of Infectious Diseases, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Hitomi Fukumoto
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuko Sato
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Michiyo Kataoka
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tsuyoshi Sekizuka
- Pathogen Genomic Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Makoto Kuroda
- Pathogen Genomic Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Harutaka Katano
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan.
| |
Collapse
|
58
|
Okabe A, Funata S, Matsusaka K, Namba H, Fukuyo M, Rahmutulla B, Oshima M, Iwama A, Fukayama M, Kaneda A. Regulation of tumour related genes by dynamic epigenetic alteration at enhancer regions in gastric epithelial cells infected by Epstein-Barr virus. Sci Rep 2017; 7:7924. [PMID: 28801683 PMCID: PMC5554293 DOI: 10.1038/s41598-017-08370-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/11/2017] [Indexed: 12/29/2022] Open
Abstract
Epstein-Barr virus (EBV) infection is associated with tumours such as Burkitt lymphoma, nasopharyngeal carcinoma, and gastric cancer. We previously showed that EBV(+) gastric cancer presents an extremely high-methylation epigenotype and this aberrant DNA methylation causes silencing of multiple tumour suppressor genes. However, the mechanisms that drive EBV infection-mediated tumorigenesis, including other epigenomic alteration, remain unclear. We analysed epigenetic alterations induced by EBV infection especially at enhancer regions, to elucidate their contribution to tumorigenesis. We performed ChIP sequencing on H3K4me3, H3K4me1, H3K27ac, H3K27me3, and H3K9me3 in gastric epithelial cells infected or not with EBV. We showed that repressive marks were redistributed after EBV infection, resulting in aberrant enhancer activation and repression. Enhancer dysfunction led to the activation of pathways related to cancer hallmarks (e.g., resisting cell death, disrupting cellular energetics, inducing invasion, evading growth suppressors, sustaining proliferative signalling, angiogenesis, and tumour-promoting inflammation) and inactivation of tumour suppressive pathways. Deregulation of cancer-related genes in EBV-infected gastric epithelial cells was also observed in clinical EBV(+) gastric cancer specimens. Our analysis showed that epigenetic alteration associated with EBV-infection may contribute to tumorigenesis through enhancer activation and repression.
Collapse
Affiliation(s)
- Atsushi Okabe
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Sayaka Funata
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Keisuke Matsusaka
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroe Namba
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masaki Fukuyo
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Bahityar Rahmutulla
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Motohiko Oshima
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Iwama
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
59
|
Ribeiro J, Malta M, Galaghar A, Silva F, Afonso LP, Medeiros R, Sousa H. P53 deregulation in Epstein-Barr virus-associated gastric cancer. Cancer Lett 2017; 404:37-43. [PMID: 28729047 DOI: 10.1016/j.canlet.2017.07.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/07/2017] [Accepted: 07/07/2017] [Indexed: 12/27/2022]
Abstract
TP53 is a tumour suppressor gene frequently mutated in human cancers; nevertheless, in EBV-associated malignancies mutations are uncommon despite frequent deregulation of the p53 pathway. In this study, we aimed to investigate p53 expression, TP53 mRNA levels and TP53 mutations in EBV-associated gastric carcinoma (EBVaGC). A case-control study was performed using 46 patients: 15 EBVaGC and 31 EBV-negative GC (EBVnGC) cases. p53 expression was detected by immunohistochemistry (IHC), the evaluation of p53 mRNA levels was performed by RT-qPCR and TP53 mutations were investigated only in EBVaGC cases using the DNA sanger sequencing method. p53 expression was found in 97.8% (45/46) of all gastric cancer cases (including EBVaGC and EBVnGC groups). Despite the high frequency of p53 expression in both groups, the percentages of cells are significantly higher among EBVaGC cases (p = 0.027). Regarding the mRNA levels, we found a significantly increased expression of p53 mRNA in EBVnGC (2-ΔΔCt = 13.4 ± 2.4; p = 0.0029) when compared with EBVaGC. Furthermore, the sequencing analysis of TP53 gene revealed that only one of the 15 EBVaGC cases presented a missense mutation. Our results demonstrated that EBV-associated gastric carcinomas are characterized by a significant decrease of TP53 mRNA levels with a strong p53 expression and rare TP53 mutations when compared with EBV-negative cancers. Considering these results, EBV seems to induce a stabilization of p53 in the EBVaGC independently of the presence of mutations, which remains to be explained.
Collapse
Affiliation(s)
- Joana Ribeiro
- Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Molecular Oncology & Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portugal
| | - Mariana Malta
- Molecular Oncology & Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portugal; Abel Salazar Institute for the Biomedical Sciences, University of Porto, Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Ana Galaghar
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Fernanda Silva
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Luís Pedro Afonso
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology & Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portugal; Abel Salazar Institute for the Biomedical Sciences, University of Porto, Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal; Virology Service, Portuguese Oncology Institute of Porto FG EPE, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; Research Department, Portuguese League Against Cancer (LPCC-NRNorte), Estrada Interior da Circunvalação 6657, 4200 Porto, Portugal
| | - Hugo Sousa
- Molecular Oncology & Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portugal; Virology Service, Portuguese Oncology Institute of Porto FG EPE, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
| |
Collapse
|
60
|
Deschamps T, Bazot Q, Leske DM, MacLeod R, Mompelat D, Tafforeau L, Lotteau V, Maréchal V, Baillie GS, Gruffat H, Wilson JB, Manet E. Epstein-Barr virus nuclear antigen 1 interacts with regulator of chromosome condensation 1 dynamically throughout the cell cycle. J Gen Virol 2017; 98:251-265. [PMID: 28284242 DOI: 10.1099/jgv.0.000681] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1) is a sequence-specific DNA-binding protein that plays an essential role in viral episome replication and segregation, by recruiting the cellular complex of DNA replication onto the origin (oriP) and by tethering the viral DNA onto the mitotic chromosomes. Whereas the mechanisms of viral DNA replication are well documented, those involved in tethering EBNA1 to the cellular chromatin are far from being understood. Here, we have identified regulator of chromosome condensation 1 (RCC1) as a novel cellular partner for EBNA1. RCC1 is the major nuclear guanine nucleotide exchange factor for the small GTPase Ran enzyme. RCC1, associated with chromatin, is involved in the formation of RanGTP gradients critical for nucleo-cytoplasmic transport, mitotic spindle formation and nuclear envelope reassembly following mitosis. Using several approaches, we have demonstrated a direct interaction between these two proteins and found that the EBNA1 domains responsible for EBNA1 tethering to the mitotic chromosomes are also involved in the interaction with RCC1. The use of an EBNA1 peptide array confirmed the interaction of RCC1 with these regions and also the importance of the N-terminal region of RCC1 in this interaction. Finally, using confocal microscopy and Förster resonance energy transfer analysis to follow the dynamics of interaction between the two proteins throughout the cell cycle, we have demonstrated that EBNA1 and RCC1 closely associate on the chromosomes during metaphase, suggesting an essential role for the interaction during this phase, perhaps in tethering EBNA1 to mitotic chromosomes.
Collapse
Affiliation(s)
- Thibaut Deschamps
- Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon 69364, France.,CNRS, UMR5308, Lyon 69364, France.,CIRI, International Center for Infectiology Research, Oncogenic Herpesviruses Team, Université de Lyon, Lyon 69364, France.,Ecole Normale Supérieure de Lyon, Lyon 69364, France.,INSERM, U1111, Lyon 69364, France
| | - Quentin Bazot
- Ecole Normale Supérieure de Lyon, Lyon 69364, France.,CNRS, UMR5308, Lyon 69364, France.,Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon 69364, France.,Present address: Section of Virology, Department of Medicine, Imperial College London, St Mary's Campus, London, UK.,CIRI, International Center for Infectiology Research, Oncogenic Herpesviruses Team, Université de Lyon, Lyon 69364, France.,INSERM, U1111, Lyon 69364, France
| | - Derek M Leske
- Present address: University of Oxford, Ludwig Institute for Cancer Research, Oxford, UK.,College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Ruth MacLeod
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Dimitri Mompelat
- Present address: University Joseph Fourier, Pathogenesis and Lentiviral Vaccination Laboratory, Grenoble, France.,INSERM, U1111, Lyon 69364, France.,Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon 69364, France.,Ecole Normale Supérieure de Lyon, Lyon 69364, France.,CIRI, International Center for Infectiology Research, Oncogenic Herpesviruses Team, Université de Lyon, Lyon 69364, France.,CNRS, UMR5308, Lyon 69364, France
| | - Lionel Tafforeau
- CIRI, International Center for Infectiology Research, Cell Biology of Viral Infections Team, Université de Lyon, Lyon 69364, France.,INSERM, U1111, Lyon 69364, France.,Present address: Cell Biology Lab, University of Mons, Mons, Belgium.,Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon 69364, France
| | - Vincent Lotteau
- Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon 69364, France.,Ecole Normale Supérieure de Lyon, Lyon 69364, France.,CNRS, UMR5308, Lyon 69364, France.,INSERM, U1111, Lyon 69364, France.,CIRI, International Center for Infectiology Research, Cell Biology of Viral Infections Team, Université de Lyon, Lyon 69364, France
| | - Vincent Maréchal
- UPMC Université Paris 6, Inserm, Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), UMR 1135, ERL CNRS 8255, F-75013 Paris, France
| | - George S Baillie
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Henri Gruffat
- CNRS, UMR5308, Lyon 69364, France.,INSERM, U1111, Lyon 69364, France.,Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon 69364, France.,CIRI, International Center for Infectiology Research, Oncogenic Herpesviruses Team, Université de Lyon, Lyon 69364, France.,Ecole Normale Supérieure de Lyon, Lyon 69364, France
| | - Joanna B Wilson
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Evelyne Manet
- INSERM, U1111, Lyon 69364, France.,Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon 69364, France.,CIRI, International Center for Infectiology Research, Oncogenic Herpesviruses Team, Université de Lyon, Lyon 69364, France.,Ecole Normale Supérieure de Lyon, Lyon 69364, France.,CNRS, UMR5308, Lyon 69364, France
| |
Collapse
|
61
|
Wang C, Wang H, Zhang Y, Guo W, Long C, Wang J, Liu L, Sun X. Berberine inhibits the proliferation of human nasopharyngeal carcinoma cells via an Epstein-Barr virus nuclear antigen 1-dependent mechanism. Oncol Rep 2017; 37:2109-2120. [PMID: 28259949 DOI: 10.3892/or.2017.5489] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/22/2016] [Indexed: 11/05/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignancy derived from the epithelial cells of the nasopharynx cavity, and is closely associated with Epstein-Barr virus (EBV) infection. In addition to NPC, EBV causes various human malignancies, such as gastric cancer, hematological tumors and lymphoepithelioma-like carcinomas. Epstein-Barr nuclear antigen 1 (EBNA1) encoded by EBV is indispensable for replication, partition, transcription and maintenance of viral genomes. Berberine, a naturally occurring isoquinoline alkaloid, shows anti-inflammatory, anticholinergic, antioxidative, and anticancer activities. In the present study, the antitumor effect of berberine was studied. Cell Counting Kit-8 (CCK-8) assays were performed to demonstrate whether the proliferation of EBV-positive NPC cells was inhibited by berberine. Flow cytometric results revealed that berberine induced cell cycle arrest and apoptosis. Quantitative-PCR and western blotting results indicated that berberine decreased the expression of EBNA1 at both the mRNA and protein levels in the EBV-positive NPC cells. The function of EBNA1 promoter Qp which is to drive EBNA1 transcription in type Ⅱ latent infection was strongly suppressed by berberine. Overexpression of EBNA1 attenuated this inhibitory effect. Berberine also suppressed the activity of signal transducer and activator of transcription 3 which is a new therapeutic target in a series of malignancies, including NPC. Viral titer experiments demonstrated that berberine decreased the production of virions in HONE1 and HK1-EBV cells. In a mouse xenograft model of NPC induced by HONE1 cells, berberine significantly inhibited tumor formation. Altogether, these results indicate that berberine decreases the expression of EBNA1 and exhibits an antitumor effect against NPC both in vitro and in vivo.
Collapse
Affiliation(s)
- Chao Wang
- State Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Huan Wang
- State Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yaqian Zhang
- State Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wei Guo
- Department of Pathology and Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Cong Long
- State Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jingchao Wang
- State Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Limei Liu
- Corneal Disease Department of Weifang Eye Hospital, Weifang, Shandong 261041, P.R. China
| | - Xiaoping Sun
- State Key Laboratory of Virology, Department of Pathogen Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
62
|
Ribeiro J, Oliveira C, Malta M, Sousa H. Epstein-Barr virus gene expression and latency pattern in gastric carcinomas: a systematic review. Future Oncol 2017; 13:567-579. [PMID: 28118740 DOI: 10.2217/fon-2016-0475] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
METHODS A systematic review of literature was conducted to identify all published reports regarding the expression of Epstein-Barr Virus (EBV) proteins/transcripts and EBV latency patterns in EBV-associated gastric carcinomas (EBVaGC). RESULTS The literature search retrieved 247 papers, of which 25 papers matched the inclusion criteria. The analysis reveals that the most frequently expressed EBV latent proteins are EBNA1 (98.1%) and LMP2A (53.8%), while LMP1 and LMP2B are present in only 10% of cases. Lytic proteins, such as BARF0 and BARF1, and other lytic transcripts are present in almost half of cases. CONCLUSION EBVaGC seems to display a unique transcription/latency pattern that does not fit the 'standard' EBV latency patterns and therefore should be further studied to better understand EBVaGC carcinogenesis.
Collapse
Affiliation(s)
- Joana Ribeiro
- Molecular Oncology & Viral Pathology Group (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino Almeida, 4200-072 Porto, Portugal.,Virology Service, Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino Almeida, 4200-072 Porto, Portugal.,Faculty of Medicine of Porto University (FMUP), Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.,Research Department, Portuguese League Against Cancer (Liga Portuguesa Contra o Cancro - Núcleo Regional do Norte), Estrada Interior da Circunvalação 6657, 4200 Porto, Portugal
| | - Cláudia Oliveira
- Molecular Oncology & Viral Pathology Group (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino Almeida, 4200-072 Porto, Portugal.,Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Mariana Malta
- Molecular Oncology & Viral Pathology Group (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino Almeida, 4200-072 Porto, Portugal.,Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Hugo Sousa
- Molecular Oncology & Viral Pathology Group (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino Almeida, 4200-072 Porto, Portugal.,Virology Service, Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino Almeida, 4200-072 Porto, Portugal
| |
Collapse
|
63
|
Shen CL, Liu CD, You RI, Ching YH, Liang J, Ke L, Chen YL, Chen HC, Hsu HJ, Liou JW, Kieff E, Peng CW. Ribosome Protein L4 is essential for Epstein-Barr Virus Nuclear Antigen 1 function. Proc Natl Acad Sci U S A 2016; 113:2229-34. [PMID: 26858444 PMCID: PMC4776490 DOI: 10.1073/pnas.1525444113] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Epstein-Barr Virus (EBV) Nuclear Antigen 1 (EBNA1)-mediated origin of plasmid replication (oriP) DNA episome maintenance is essential for EBV-mediated tumorigenesis. We have now found that EBNA1 binds to Ribosome Protein L4 (RPL4). RPL4 shRNA knockdown decreased EBNA1 activation of an oriP luciferase reporter, EBNA1 DNA binding in lymphoblastoid cell lines, and EBV genome number per lymphoblastoid cell line. EBV infection increased RPL4 expression and redistributed RPL4 to cell nuclei. RPL4 and Nucleolin (NCL) were a scaffold for an EBNA1-induced oriP complex. The RPL4 N terminus cooperated with NCL-K429 to support EBNA1 and oriP-mediated episome binding and maintenance, whereas the NCL C-terminal K380 and K393 induced oriP DNA H3K4me2 modification and promoted EBNA1 activation of oriP-dependent transcription. These observations provide new insights into the mechanisms by which EBV uses NCL and RPL4 to establish persistent B-lymphoblastoid cell infection.
Collapse
Affiliation(s)
- Chih-Lung Shen
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
| | - Cheng-Der Liu
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
| | - Ren-In You
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Sec. 3, Hualien 97004, Taiwan
| | - Yung-Hao Ching
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Sec. 3, Hualien 97004, Taiwan
| | - Jun Liang
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115
| | - Liangru Ke
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115
| | - Ya-Lin Chen
- Department of Life Sciences, Tzu Chi University, Sec. 3, Hualien 97004, Taiwan
| | - Hong-Chi Chen
- Department of Life Sciences, Tzu Chi University, Sec. 3, Hualien 97004, Taiwan
| | - Hao-Jen Hsu
- Department of Life Sciences, Tzu Chi University, Sec. 3, Hualien 97004, Taiwan
| | - Je-Wen Liou
- Institute of Biochemical Sciences, Tzu Chi University, Sec. 3, Hualien 97004, Taiwan
| | - Elliott Kieff
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115;
| | - Chih-Wen Peng
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan; Department of Life Sciences, Tzu Chi University, Sec. 3, Hualien 97004, Taiwan;
| |
Collapse
|