1
|
Chan WJ, Li H. Recent advances in nano/micro systems for improved circulation stability, enhanced tumor targeting, penetration, and intracellular drug delivery: a review. Biomed Phys Eng Express 2024; 10:022001. [PMID: 38086099 DOI: 10.1088/2057-1976/ad14f0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 12/12/2023] [Indexed: 01/17/2024]
Abstract
In recent years, nanoparticles (NPs) have been extensively developed as drug carriers to overcome the limitations of cancer therapeutics. However, there are several biological barriers to nanomedicines, which include the lack of stability in circulation, limited target specificity, low penetration into tumors and insufficient cellular uptake, restricting the active targeting toward tumors of nanomedicines. To address these challenges, a variety of promising strategies were developed recently, as they can be designed to improve NP accumulation and penetration in tumor tissues, circulation stability, tumor targeting, and intracellular uptake. In this Review, we summarized nanomaterials developed in recent three years that could be utilized to improve drug delivery for cancer treatments.
Collapse
Affiliation(s)
- Wei-Jen Chan
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| | - Huatian Li
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| |
Collapse
|
2
|
Krishnan D, Babu S, Raju R, Veettil MV, Prasad TSK, Abhinand CS. Epstein-Barr Virus: Human Interactome Reveals New Molecular Insights into Viral Pathogenesis for Potential Therapeutics and Antiviral Drug Discovery. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:32-44. [PMID: 38190109 DOI: 10.1089/omi.2023.0241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Host-virus Protein-Protein Interactions (PPIs) play pivotal roles in biological processes crucial for viral pathogenesis and by extension, inform antiviral drug discovery and therapeutics innovations. Despite efforts to develop the Epstein-Barr virus (EBV)-host PPI network, there remain significant knowledge gaps and a limited number of interacting human proteins deciphered. Furthermore, understanding the dynamics of the EBV-host PPI network in the distinct lytic and latent viral stages remains elusive. In this study, we report a comprehensive map of the EBV-human protein interactions, encompassing 1752 human and 61 EBV proteins by integrating data from the public repository HPIDB (v3.0) as well as curated high-throughput proteomic data from the literature. To address the stage-specific nature of EBV infection, we generated two detailed subset networks representing the latent and lytic stages, comprising 747 and 481 human proteins, respectively. Functional and pathway enrichment analysis of these subsets uncovered the profound impact of EBV proteins on cancer. The identification of highly connected proteins and the characterization of intrinsically disordered and cancer-related proteins provide valuable insights into potential therapeutic targets. Moreover, the exploration of drug-protein interactions revealed notable associations between hub proteins and anticancer drugs, offering novel perspectives for controlling EBV pathogenesis. This study represents, to the best of our knowledge, the first comprehensive investigation of the two distinct stages of EBV infection using high-throughput datasets. This makes a contribution to our understanding of EBV-host interactions and provides a foundation for future drug discovery and therapeutic interventions.
Collapse
Affiliation(s)
- Deepak Krishnan
- Centre for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre (YRC), Yenepoya (Deemed to be University), Mangalore, India
| | - Sreeranjini Babu
- Centre for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre (YRC), Yenepoya (Deemed to be University), Mangalore, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | | | | | - Chandran S Abhinand
- Centre for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre (YRC), Yenepoya (Deemed to be University), Mangalore, India
| |
Collapse
|
3
|
Wang WT, Yang Y, Zhang Y, Le YN, Wu YL, Liu YY, Tu YJ. EBV-microRNAs as Potential Biomarkers in EBV-related Fever: A Narrative Review. Curr Mol Med 2024; 24:2-13. [PMID: 36411555 PMCID: PMC10825793 DOI: 10.2174/1566524023666221118122005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 07/31/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022]
Abstract
At present, timely and accurate diagnosis and effective treatment of Epstein- Barr Virus (EBV) infection-associated fever remain a difficult challenge. EBV encodes 44 mature microRNAs (miRNAs) that inhibit viral lysis, adjust inflammatory response, regulate cellular apoptosis, promote tumor genesis and metastasis, and regulate tumor cell metabolism. Herein, we have collected the specific expression data of EBV-miRNAs in EBV-related fevers, including infectious mononucleosis (IM), EBVassociated hemophagocytic lymphohistiocytosis (EBV-HLH), chronic active EBV infection (CAEBV), and EBV-related tumors, and proposed the potential value of EBVmiRNAs as biomarkers to assist in the identification, diagnosis, and prognosis of EBVrelated fever, as well as therapeutic targets for drug development.
Collapse
Affiliation(s)
- Wei-ting Wang
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai (201203), China
| | - Yun Yang
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai (201203), China
| | - Yang Zhang
- Information Center of Science and Technology, Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai (201203), China
| | - Yi-ning Le
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai (200433), China
| | - Yu-lin Wu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai (201203), China
| | - Yi-yi Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai (200032), China
| | - Yan-jie Tu
- Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai (201203), China
| |
Collapse
|
4
|
Sausen DG, Poirier MC, Spiers LM, Smith EN. Mechanisms of T cell evasion by Epstein-Barr virus and implications for tumor survival. Front Immunol 2023; 14:1289313. [PMID: 38179040 PMCID: PMC10764432 DOI: 10.3389/fimmu.2023.1289313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Epstein-Barr virus (EBV) is a prevalent oncogenic virus estimated to infect greater than 90% of the world's population. Following initial infection, it establishes latency in host B cells. EBV has developed a multitude of techniques to avoid detection by the host immune system and establish lifelong infection. T cells, as important contributors to cell-mediated immunity, make an attractive target for these immunoevasive strategies. Indeed, EBV has evolved numerous mechanisms to modulate T cell responses. For example, it can augment expression of programmed cell death ligand-1 (PD-L1), which inhibits T cell function, and downregulates the interferon response, which has a strong impact on T cell regulation. It also modulates interleukin secretion and can influence major histocompatibility complex (MHC) expression and presentation. In addition to facilitating persistent EBV infection, these immunoregulatory mechanisms have significant implications for evasion of the immune response by tumor cells. This review dissects the mechanisms through which EBV avoids detection by host T cells and discusses how these mechanisms play into tumor survival. It concludes with an overview of cancer treatments targeting T cells in the setting of EBV-associated malignancy.
Collapse
Affiliation(s)
- D. G. Sausen
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | | | | | | |
Collapse
|
5
|
Xu L, Zhang M, Tu D, Lu Z, Lu T, Ma D, Zhou Y, Zhang S, Ma Y, Yan D, Wang X, Sang W. Chidamide Induces Epstein-Barr Virus (EBV) Lytic Infection and Acts Synergistically with Tenofovir to Eliminate EBV-Positive Burkitt Lymphoma. J Pharmacol Exp Ther 2023; 387:288-298. [PMID: 37875309 DOI: 10.1124/jpet.123.001583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
Epstein-Barr virus (EBV) is a type of human γ-herpesvirus, and its reactivation plays an important role in the development of EBV-driven Burkitt lymphoma (BL). Despite intensive chemotherapy, the prognosis of relapsed/refractory BL patients remains unfavorable, and a definitive method to completely eliminate latent EBV infection is lacking. Previous studies have demonstrated that histone deacetylase (HDAC) inhibitors can induce the transition of EBV from latency to the lytic phase. The lytic activation of EBV can be inhibited by tenofovir, a potent inhibitor of DNA replication. Herein, we explored the antitumor effect and EBV clearance potential of a novel HDAC inhibitor called chidamide, combined with tenofovir, in the treatment of EBV-positive BL. In the study, chidamide exhibited inhibitory activity against HDAC. Moreover, chidamide inhibited BL cell proliferation, arrested cell cycle progression, and induced BL cell apoptosis primarily by regulating the MAPK pathways. Additionally, chidamide promoted the transcription of lytic genes, including BZLF1, BMRF1, and BMLF1 Compared with chidamide alone, the addition of tenofovir further induced growth arrest and apoptosis in EBV-positive BL cells and inhibited the transcriptions of EBV lytic genes induced by chidamide alone. Furthermore, our in vivo data demonstrated that the combination of chidamide and tenofovir had superior tumor-suppressive effects in a mouse model of BL cell tumors. The aforementioned findings confirm the synergistic effect of chidamide combined with tenofovir in inducing growth inhibition and apoptosis in EBV-positive BL cells and provide an effective strategy for eliminating EBV and EBV-associated malignancies. SIGNIFICANCE STATEMENT: High levels of Epstein-Barr virus (EBV)-DNA have consistently been associated with unfavorable progression-free survival and overall survival in EBV-associated lymphomas. Therefore, identifying novel strategies to effectively eradicate tumor cells and eliminate EBV is crucial for lymphoma patients. This study confirmed, for the first time, the synergistic effect of chidamide combined with tenofovir in the treatment of Burkitt lymphoma and the eradication of EBV virus.
Collapse
Affiliation(s)
- Linyan Xu
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Meng Zhang
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dongyun Tu
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ziyi Lu
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tianyi Lu
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dongshen Ma
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yi Zhou
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shuo Zhang
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuhan Ma
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dongmei Yan
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiangmin Wang
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei Sang
- 1Blood Diseases Institute (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Key Laboratory of Bone Marrow Stem Cell (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.), Xuzhou Medical University, Xuzhou, China; and Departments of Hematology (L.X., M.Z., D.T., Z.L., T.L., Y.Z., S.Z., Y.M., D.Y., X.W., W.S.) and Pathology (D.M.), the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
6
|
Hedström AK. Risk factors for multiple sclerosis in the context of Epstein-Barr virus infection. Front Immunol 2023; 14:1212676. [PMID: 37554326 PMCID: PMC10406387 DOI: 10.3389/fimmu.2023.1212676] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/26/2023] [Indexed: 08/10/2023] Open
Abstract
Compelling evidence indicates that Epstein Barr virus (EBV) infection is a prerequisite for multiple sclerosis (MS). The disease may arise from a complex interplay between latent EBV infection, genetic predisposition, and various environmental and lifestyle factors that negatively affect immune control of the infection. Evidence of gene-environment interactions and epigenetic modifications triggered by environmental factors in genetically susceptible individuals supports this view. This review gives a short introduction to EBV and host immunity and discusses evidence indicating EBV as a prerequisite for MS. The role of genetic and environmental risk factors, and their interactions, in MS pathogenesis is reviewed and put in the context of EBV infection. Finally, possible preventive measures are discussed based on the findings presented.
Collapse
Affiliation(s)
- Anna Karin Hedström
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
7
|
Andersen O, Ernberg I, Hedström AK. Treatment Options for Epstein-Barr Virus-Related Disorders of the Central Nervous System. Infect Drug Resist 2023; 16:4599-4620. [PMID: 37465179 PMCID: PMC10351589 DOI: 10.2147/idr.s375624] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/28/2023] [Indexed: 07/20/2023] Open
Abstract
Epstein-Barr virus (EBV), a causative agent for several types of lymphomas and mucosal cancers, is a human lymphotropic herpesvirus with the capacity to establish lifelong latent infection. More than 90% of the human population worldwide is infected. The primary infection is usually asymptomatic in childhood, whereas infectious mononucleosis (IM) is common when the infection occurs in adolescence. Primary EBV infection, with or without IM, or reactivation of latent infection in immunocompromised individuals have been associated with a wide range of neurologic conditions, such as encephalitis, meningitis, acute disseminated encephalomyelitis, and cerebellitis. EBV is also involved in malignant lymphomas in the brain. An increasing number of reports on EBV-related disorders of the central nervous system (CNS) including the convincing association with multiple sclerosis (MS) have put in focus EBV-related conditions beyond its established link to malignancies. In this review, we present the clinical manifestations of EBV-related CNS-disorders, put them in the context of known EBV biology and focus on available treatment options and future therapeutic approaches.
Collapse
Affiliation(s)
- Oluf Andersen
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingemar Ernberg
- Department of Microbiology, Tumor and Cell Biology, Biomedicum Q8C, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Anna Karin Hedström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
8
|
Dunn LEM, Lu F, Su C, Lieberman PM, Baines JD. Reactivation of Epstein-Barr Virus from Latency Involves Increased RNA Polymerase Activity at CTCF Binding Sites on the Viral Genome. J Virol 2023; 97:e0189422. [PMID: 36744959 PMCID: PMC9972995 DOI: 10.1128/jvi.01894-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/11/2023] [Indexed: 02/07/2023] Open
Abstract
The ability of Epstein-Barr virus (EBV) to switch between latent and lytic infection is key to its long-term persistence, yet the molecular mechanisms behind this switch remain unclear. To investigate transcriptional events during the latent-to-lytic switch, we utilized Precision nuclear Run On followed by deep Sequencing (PRO-Seq) to map cellular RNA polymerase (Pol) activity to single-nucleotide resolution on the host and EBV genome in three different models of EBV latency and reactivation. In latently infected Mutu-I Burkitt lymphoma (BL) cells, Pol activity was enriched at the Qp promoter, the EBER region, and the BHLF1/LF3 transcripts. Upon reactivation with phorbol ester and sodium butyrate, early-phase Pol activity occurred bidirectionally at CTCF sites within the LMP-2A, EBER-1, and RPMS1 loci. PRO-Seq analysis of Akata cells reactivated from latency with anti-IgG and a lymphoblastoid cell line (LCL) reactivated with small molecule C60 showed a similar pattern of early bidirectional transcription initiating around CTCF binding sites, although the specific CTCF sites and viral genes were different for each latency model. The functional importance of CTCF binding, transcription, and reactivation was confirmed using an EBV mutant lacking the LMP-2A CTCF binding site. This virus was unable to reactivate and had disrupted Pol activity at multiple CTCF binding sites relative to the wild-type (WT) virus. Overall, these data suggest that CTCF regulates the viral early transcripts during reactivation from latency. These activities likely help maintain the accessibility of the viral genome to initiate productive replication. IMPORTANCE The ability of EBV to switch between latent and lytic infection is key to its long-term persistence in memory B cells, and its ability to persist in proliferating cells is strongly linked to oncogenesis. During latency, most viral genes are epigenetically silenced, and the virus must overcome this repression to reactivate lytic replication. Reactivation occurs once the immediate early (IE) EBV lytic genes are expressed. However, the molecular mechanisms behind the switch from the latent transcriptional program to begin transcription of the IE genes remain unknown. In this study, we mapped RNA Pol positioning and activity during latency and reactivation. Unexpectedly, Pol activity accumulated at distinct regions characteristic of transcription initiation on the EBV genome previously shown to be associated with CTCF. We propose that CTCF binding at these regions retains Pol to maintain a stable latent chromosome conformation and a rapid response to various reactivation signals.
Collapse
Affiliation(s)
- Laura E. M. Dunn
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
- Department of Pathobiology, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Fang Lu
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Chenhe Su
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | - Joel D. Baines
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
- Department of Pathobiology, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
9
|
Liao Y, Zhang JB, Lu LX, Jia YJ, Zheng MQ, Debelius JW, He YQ, Wang TM, Deng CM, Tong XT, Xue WQ, Cao LJ, Wu ZY, Yang DW, Zheng XH, Li XZ, Wu YX, Feng L, Ye W, Mu J, Jia WH. Oral Microbiota Alteration and Roles in Epstein-Barr Virus Reactivation in Nasopharyngeal Carcinoma. Microbiol Spectr 2023; 11:e0344822. [PMID: 36645283 PMCID: PMC9927204 DOI: 10.1128/spectrum.03448-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/16/2022] [Indexed: 01/17/2023] Open
Abstract
Microbiota has recently emerged as a critical factor associated with multiple malignancies. Nasopharyngeal carcinoma (NPC) is highly associated with Epstein-Barr virus (EBV); the oncovirus resides and is transmitted in the oral cavity. However, the alternation of oral microbiota in NPC patients and its potential link to EBV reactivation and host cell response under the simultaneous existence of EBV and specific bacteria is largely unknown. Here, oral microbiota profiles of 303 NPC patients and controls with detailed clinical information, including serum EBV anti-virus capsid antigen (VCA) IgA level, were conducted. A distinct microbial community with lower diversity and imbalanced composition in NPC patients was observed. Notably, among enriched bacteria in patients, Streptococcus sanguinis was associated with anti-VCA IgA, an indicator of NPC risk and EBV reactivation. By measuring the concentration of its metabolite, hydrogen peroxide (H2O2), in the saliva of clinical patients, we found the detection rate of H2O2 was 2-fold increased compared to healthy controls. Further coculture assay of EBV-positive Akata cells with bacteria in vitro showed that S. sanguinis induced EBV lytic activation by its metabolite, H2O2. Host and EBV whole genome-wide transcriptome sequencing and EBV methylation assays showed that H2O2 triggered the host cell signaling pathways, notably tumor necrosis factor alpha (TNF-α) via NF-κB, and induced the demethylation of the global EBV genome and the expression of EBV lytic-associated genes, which could result in an increase of virus particle release and potential favorable events toward tumorigenesis. In brief, our study identified a characterized oral microbial profile in NPC patients and established a robust link between specific oral microbial alteration and switch of latency to lytic EBV infection status in the oral cavity, which provides novel insights into EBV's productive cycle and might help to further clarify the etiology of NPC. IMPORTANCE EBV is classified as the group I human carcinogen and is associated with multiple cancers, including NPC. The interplays between the microbiota and oncovirus in cancer development remain largely unknown. In this study, we investigate the interactions between resident microbes and EBV coexistence in the oral cavity of NPC patients. We identify a distinct oral microbial feature for NPC patients. Among NPC-enriched bacteria, we illustrated that a specific species, S. sanguinis, associated with elevated anti-IgA VCA in patients, induced EBV lytic activation by its by-product, H2O2, and activated the TNF-α/NF-κB pathway of EBV-positive B cells in vitro, together with increased detection rate of H2O2 in patients' oral cavities, which strengthened the evidence of bacteria-virus-host interaction in physiological circumstances. The effects of imbalanced microbiota on the EBV latent-to-lytic switch in the oral cavity might create the likelihood of EBV infection in epithelial cells at the nasopharynx and help malignant transition and cancer development.
Collapse
Affiliation(s)
- Ying Liao
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Jiang-Bo Zhang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Li-Xia Lu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yi-Jing Jia
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Mei-Qi Zheng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Justine W. Debelius
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yong-Qiao He
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Tong-Min Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Chang-Mi Deng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Xia-Ting Tong
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Wen-Qiong Xue
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Lian-Jing Cao
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Zi-Yi Wu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Da-Wei Yang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Hui Zheng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Xi-Zhao Li
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Yan-Xia Wu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Lin Feng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Wei-Hua Jia
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
10
|
Yang T, You C, Meng S, Lai Z, Ai W, Zhang J. EBV Infection and Its Regulated Metabolic Reprogramming in Nasopharyngeal Tumorigenesis. Front Cell Infect Microbiol 2022; 12:935205. [PMID: 35846746 PMCID: PMC9283984 DOI: 10.3389/fcimb.2022.935205] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/31/2022] [Indexed: 01/05/2023] Open
Abstract
Viral oncogenes may drive cellular metabolic reprogramming to modulate the normal epithelia cell malignant transformation. Understanding the viral oncogene-mediated signaling transduction dysregulation that involves in metabolic reprogramming may provide new therapeutic targets for virus-associated cancer treatment. Latent EBV infection and expression of viral oncogenes, including latent membrane proteins 1 and 2 (LMP1/2), and EBV-encoded BamH I-A rightward transcripts (BART) microRNAs (miR-BARTs), have been demonstrated to play fundamental roles in altering host cell metabolism to support nasopharyngeal carcinoma (NPC) pathogenesis. Yet, how do EBV infection and its encoded oncogenes facilitated the metabolic shifting and their roles in NPC carcinogenesis remains unclear. In this review, we will focus on delineating how EBV infection and its encoded oncoproteins altered the metabolic reprograming of infected cells to support their malignances. Furthermore, based on the understanding of the host's metabolic signaling alterations induced by EBV, we will provide a new perspective on the interplay between EBV infection and these metabolic pathways and offering a potential therapeutic intervention strategy in the treatment of EBV-associated malignant diseases.
Collapse
Affiliation(s)
- Tingting Yang
- Department of Pharmacy, Shenzhen University General Hospital, Shenzhen, China
| | - Chanping You
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Shuhui Meng
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen People’s Hospital, Shenzhen, China
| | - Zhengquan Lai
- Department of Pharmacy, Shenzhen University General Hospital, Shenzhen, China
| | - Weipeng Ai
- Department of Pharmacy, Shenzhen University General Hospital, Shenzhen, China
| | - Jun Zhang
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
11
|
Wyżewski Z, Mielcarska MB, Gregorczyk-Zboroch KP, Myszka A. Virus-Mediated Inhibition of Apoptosis in the Context of EBV-Associated Diseases: Molecular Mechanisms and Therapeutic Perspectives. Int J Mol Sci 2022; 23:ijms23137265. [PMID: 35806271 PMCID: PMC9266970 DOI: 10.3390/ijms23137265] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Epstein-Barr virus (EBV), the representative of the Herpesviridae family, is a pathogen extensively distributed in the human population. One of its most characteristic features is the capability to establish latent infection in the host. The infected cells serve as a sanctuary for the dormant virus, and therefore their desensitization to apoptotic stimuli is part of the viral strategy for long-term survival. For this reason, EBV encodes a set of anti-apoptotic products. They may increase the viability of infected cells and enhance their resistance to chemotherapy, thereby contributing to the development of EBV-associated diseases, including Burkitt’s lymphoma (BL), Hodgkin’s lymphoma (HL), gastric cancer (GC), nasopharyngeal carcinoma (NPC) and several other malignancies. In this paper, we have described the molecular mechanism of anti-apoptotic actions of a set of EBV proteins. Moreover, we have reviewed the pro-survival role of non-coding viral transcripts: EBV-encoded small RNAs (EBERs) and microRNAs (miRNAs), in EBV-carrying malignant cells. The influence of EBV on the expression, activity and/or intracellular distribution of B-cell lymphoma 2 (Bcl-2) protein family members, has been presented. Finally, we have also discussed therapeutic perspectives of targeting viral anti-apoptotic products or their molecular partners.
Collapse
Affiliation(s)
- Zbigniew Wyżewski
- Institute of Biological Sciences, Cardinal Stefan Wyszyński University, Dewajtis 5, 01-815 Warsaw, Poland;
- Correspondence: ; Tel.: +48-728-208-338
| | - Matylda Barbara Mielcarska
- Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Nowoursynowska 166, 02-787 Warsaw, Poland; (M.B.M.); (K.P.G.-Z.)
| | | | - Anna Myszka
- Institute of Biological Sciences, Cardinal Stefan Wyszyński University, Dewajtis 5, 01-815 Warsaw, Poland;
| |
Collapse
|
12
|
Wang Y, Qiao X, Li Y, Yang Q, Wang L, Liu X, Wang H, Shen H. Role of the receptor for activated C kinase 1 during viral infection. Arch Virol 2022; 167:1915-1924. [PMID: 35763066 DOI: 10.1007/s00705-022-05484-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 03/30/2022] [Indexed: 11/29/2022]
Abstract
Viruses can survive only in living cells, where they depend on the host's enzymatic system for survival and reproduction. Virus-host interactions are complex. On the one hand, hosts express host-restricted factors to protect the host cells from viral infections. On the other hand, viruses recruit certain host factors to facilitate their survival and transmission. The identification of host factors critical to viral infection is essential for comprehending the pathogenesis of contagion and developing novel antiviral therapies that specifically target the host. Receptor for activated C kinase 1 (RACK1), an evolutionarily conserved host factor that exists in various eukaryotic organisms, is a promising target for antiviral therapy. This review primarily summarizes the roles of RACK1 in regulating different viral life stages, particularly entry, replication, translation, and release.
Collapse
Affiliation(s)
- Yan Wang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Xiaorong Qiao
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yuhan Li
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Qingru Yang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Lulu Wang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Xiaolan Liu
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Hua Wang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Hongxing Shen
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
13
|
The roles of DNA methylation on the promotor of the Epstein–Barr virus (EBV) gene and the genome in patients with EBV-associated diseases. Appl Microbiol Biotechnol 2022; 106:4413-4426. [PMID: 35763069 PMCID: PMC9259528 DOI: 10.1007/s00253-022-12029-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022]
Abstract
Abstract Epstein–Barr virus (EBV) is an oncogenic virus that is closely associated with several malignant and lymphoproliferative diseases. Studies have shown that the typical characteristic of EBV-associated diseases is aberrant methylation of viral DNA and the host genome. EBV gene methylation helps EBV escape from immune monitoring and persist in host cells. EBV controls viral gene promoter methylation by hijacking host epigenetic machinery to regulate the expression of viral genes. EBV proteins also interact with host epigenetic regulatory factors to mediate the methylation of the host’s important tumour suppressor gene promoters, thereby participating in the occurrence of tumorigenesis. Since epigenetic modifications, including DNA methylation, are reversible in nature, drugs that target DNA methylation can be developed for epigenetic therapy against EBV-associated tumours. Various methylation modes in the host and EBV genomes may also be of diagnostic and prognostic value. This review summarizes the regulatory roles of DNA methylation on the promotor of EBV gene and host genome in EBV-associated diseases, proposes the application prospect of DNA methylation in early clinical diagnosis and treatment, and provides insight into methylation-based strategies against EBV-associated diseases. Key points • Methylation of both the host and EBV genomes plays an important role in EBV-associateddiseases. • The functions of methylation of the host and EBV genomes in the occurrence and development of EBV-associated diseases are diverse. • Methylation may be a therapeutic target or biomarker in EBV-associated diseases.
Collapse
|
14
|
Sato Y, Yaguchi M, Okuno Y, Ishimaru H, Sagou K, Ozaki S, Suzuki T, Inagaki T, Umeda M, Watanabe T, Fujimuro M, Murata T, Kimura H. Epstein-Barr virus tegument protein BGLF2 in exosomes released from virus-producing cells facilitates de novo infection. Cell Commun Signal 2022; 20:95. [PMID: 35729616 PMCID: PMC9210680 DOI: 10.1186/s12964-022-00902-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/22/2022] [Indexed: 11/23/2022] Open
Abstract
Background Viruses must adapt to the environment of their host cells to establish infection and persist. Diverse mammalian cells, including virus-infected cells, release extracellular vesicles such as exosomes containing proteins and miRNAs, and use these vesicles to mediate intercellular communication. However, the roles of exosomes in viral infection remain unclear. Results We screened viral proteins to identify those responsible for the exosome-mediated enhancement of Epstein–Barr virus (EBV) infection. We identified BGLF2 protein encapsulated in exosomes, which were released by EBV-infected cells. BGLF2 protein is a tegument protein that exists in the space between the envelope and nucleocapsid, and it is released into the cytoplasm shortly after infection. BGLF2 protein-containing exosomes enhanced viral gene expression and repressed innate immunity, thereby supporting the EBV infection. Conclusions The EBV tegument protein BGLF2 is encapsulated in exosomes and released by infected cells to facilitate the establishment of EBV infection. These findings suggest that tegument proteins support viral infection not only between the envelope and nucleocapsid, as well as in extraviral particles such as exosomes. Graphical abstract ![]()
Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00902-7.
Collapse
Affiliation(s)
- Yoshitaka Sato
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan. .,PRESTO, Japan Science and Technology Agency (JST), Kawaguchi, Japan.
| | - Masahiro Yaguchi
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yusuke Okuno
- Department of Virology, Graduate School of Medicine, Nagoya City University, Nagoya, Japan
| | - Hanako Ishimaru
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan.,Division of Clinical Virology, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ken Sagou
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Somi Ozaki
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Suzuki
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoki Inagaki
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Dermatology, School of Medicine, University of California Davis (UC Davis), Sacramento, CA, USA
| | - Miki Umeda
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahiro Watanabe
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Fujimuro
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Takayuki Murata
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Virology and Parasitology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hiroshi Kimura
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
15
|
Yanagi Y, Watanabe T, Hara Y, Sato Y, Kimura H, Murata T. EBV Exploits RNA m6A Modification to Promote Cell Survival and Progeny Virus Production During Lytic Cycle. Front Microbiol 2022; 13:870816. [PMID: 35783391 PMCID: PMC9240777 DOI: 10.3389/fmicb.2022.870816] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/09/2022] [Indexed: 12/14/2022] Open
Abstract
N6-methyladenosine (m6A) mediates various biological processes by affecting RNA stability, splicing, and translational efficiency. The roles of m6A modification in Epstein-Barr virus (EBV) infection in the lytic phase are unclear. Here, knockout of the m6A methyltransferase, N6-methyladenosine methyltransferase-like 3 (METTL3), or inhibition of methylation by DAA or UZH1a decreased the expression of viral lytic proteins and reduced progeny virion production. Interestingly, cell growth and viability were decreased by induction of the lytic cycle in METTL3-knockout or inhibitor-treated cells. Apoptosis was induced in those conditions possibly because of a decreased level of the anti-apoptotic viral protein, BHRF1. Therefore, m6A shows potential as a target of lytic induction therapy for EBV-associated cancers, including Burkitt lymphoma.
Collapse
Affiliation(s)
- Yusuke Yanagi
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahiro Watanabe
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuya Hara
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshitaka Sato
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kimura
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takayuki Murata
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Toyoake, Japan
- *Correspondence: Takayuki Murata
| |
Collapse
|
16
|
Chen S, Zheng J, Zhang B, Tang X, Cun Y, Wu T, Xu Y, Ma T, Cheng J, Yu Z, Wang H. Identification and characterization of virus-encoded circular RNAs in host cells. Microb Genom 2022; 8. [PMID: 35731570 PMCID: PMC9455708 DOI: 10.1099/mgen.0.000848] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Emerging evidence has identified viral circular RNAs (circRNAs) in human cells infected by viruses, interfering with the immune system and inducing diseases including human cancer. However, the biogenesis and regulatory mechanisms of virus-encoded circRNAs in host cells remain unknown. In this study, we used the circRNA detection tool CIRI2 to systematically determine the virus-encoded circRNAs in virus-infected cancer cell lines and cancer patients, by analysing RNA-Seq datasets derived from RNase R-treated samples. Based on the thousands of viral circRNAs we identified, the biological characteristics and potential roles of viral circRNAs in regulating host cell function were determined. In addition, we developed a Viral-circRNA Database (http://www.hywanglab.cn/vcRNAdb/), which is open to all users to search, browse and download information on circRNAs encoded by viruses upon infection.
Collapse
Affiliation(s)
- Shuting Chen
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, PR China
| | - Jie Zheng
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, PR China
| | - Bingyue Zhang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, PR China
| | - Xinyue Tang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, PR China
| | - Yewei Cun
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, PR China
| | - Tao Wu
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, PR China
| | - Yue Xu
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, PR China
| | - Ting Ma
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, PR China
| | - Jingxin Cheng
- Department of Obstetrics and Gynecology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai 200120, PR China
| | - Zuoren Yu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai 200120, PR China
| | - Haiyun Wang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, PR China
| |
Collapse
|
17
|
The Central Role of the Ubiquitin-Proteasome System in EBV-Mediated Oncogenesis. Cancers (Basel) 2022; 14:cancers14030611. [PMID: 35158879 PMCID: PMC8833352 DOI: 10.3390/cancers14030611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Epstein–Barr virus (EBV) is the first discovered human tumor virus, which contributes to the oncogenesis of many human cancers. The ubiquitin–proteasome system is a key player during EBV-mediated oncogenesis and has been developed as a crucial therapeutic target for treatment. In this review, we briefly describe how EBV antigens can modulate the ubiquitin–proteasome system for targeted protein degradation and how they are regulated in the EBV life cycle to mediate oncogenesis. Additionally, the developed proteasome inhibitors are discussed for the treatment of EBV-associated cancers. Abstract Deregulation of the ubiquitin–proteasome system (UPS) plays a critical role in the development of numerous human cancers. Epstein–Barr virus (EBV), the first known human tumor virus, has evolved distinct molecular mechanisms to manipulate the ubiquitin–proteasome system, facilitate its successful infection, and drive opportunistic cancers. The interactions of EBV antigens with the ubiquitin–proteasome system can lead to oncogenesis through the targeting of cellular factors involved in proliferation. Recent studies highlight the central role of the ubiquitin–proteasome system in EBV infection. This review will summarize the versatile strategies in EBV-mediated oncogenesis that contribute to the development of specific therapeutic approaches to treat EBV-associated malignancies.
Collapse
|
18
|
Epstein-Barr virus miR-BHRF1-3 targets the BZLF1 3'UTR and regulates the lytic cycle. J Virol 2021; 96:e0149521. [PMID: 34878852 DOI: 10.1128/jvi.01495-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Suppression of lytic viral gene expression is a key aspect of the Epstein-Barr virus (EBV) life cycle to facilitate the establishment of latent infection. Molecular mechanisms regulating transitions between EBV lytic replication and latency are not fully understood. Here, we investigated the impact of viral microRNAs on the EBV lytic cycle. Through functional assays, we found that miR-BHRF1-3 attenuates EBV lytic gene expression following reactivation. To understand the miRNA targets contributing to this activity, we performed Ago PAR-CLIP analysis on EBV-positive, reactivated Burkitt's lymphoma cells and identified multiple miR-BHRF1-3 interactions with viral transcripts. Using luciferase reporter assays, we confirmed a miRNA interaction site within the 3'UTR of BZLF1 which encodes the essential immediate early (IE) transactivator Zta. Comparison of >850 published EBV genomes identified sequence polymorphisms within the miR-BHRF1-3 locus that deleteriously affect miRNA expression and function. Molecular interactions between the homologous viral miRNA, miR-rL1-17, and IE transcripts encoded by rhesus lymphocryptovirus were further identified. Our data demonstrate that regulation of IE gene expression by a BHRF1 miRNA is conserved amongst lymphocryptoviruses, and further reveal virally-encoded genetic elements that orchestrate viral antigen expression during the lytic cycle. Importance Epstein-Barr virus infection is predominantly latent in healthy individuals, while periodic cycles of reactivation are thought to facilitate persistent lifelong infection. Lytic infection has been linked to development of certain EBV-associated diseases. Here, we demonstrate that EBV miR-BHRF1-3 can suppress lytic replication by directly inhibiting Zta expression. Moreover, we identify nucleotide variants that impact the function of miR-BHRF1-3, which may contribute to specific EBV pathologies.
Collapse
|
19
|
Molecular Basis of Epstein-Barr Virus Latency Establishment and Lytic Reactivation. Viruses 2021; 13:v13122344. [PMID: 34960613 PMCID: PMC8706188 DOI: 10.3390/v13122344] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022] Open
Abstract
Epstein–Barr virus (EBV) is a causative agent of infectious mononucleosis and several types of cancer. Like other herpesviruses, it establishes an asymptomatic, life-long latent infection, with occasional reactivation and shedding of progeny viruses. During latency, EBV expresses a small number of viral genes, and exists as an episome in the host–cell nucleus. Expression patterns of latency genes are dependent on the cell type, time after infection, and milieu of the cell (e.g., germinal center or peripheral blood). Upon lytic induction, expression of the viral immediate-early genes, BZLF1 and BRLF1, are induced, followed by early gene expression, viral DNA replication, late gene expression, and maturation and egress of progeny virions. Furthermore, EBV reactivation involves more than just progeny production. The EBV life cycle is regulated by signal transduction, transcription factors, promoter sequences, epigenetics, and the 3D structure of the genome. In this article, the molecular basis of EBV latency establishment and reactivation is summarized.
Collapse
|
20
|
Cai J, Zhang B, Li Y, Zhu W, Akihisa T, Li W, Kikuchi T, Liu W, Feng F, Zhang J. Prophylactic and Therapeutic EBV Vaccines: Major Scientific Obstacles, Historical Progress, and Future Direction. Vaccines (Basel) 2021; 9:vaccines9111290. [PMID: 34835222 PMCID: PMC8623587 DOI: 10.3390/vaccines9111290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 11/29/2022] Open
Abstract
The Epstein-Barr virus (EBV) infects more than 95% of adults worldwide and is associated with various malignant tumors and immune diseases, imparting a huge disease burden on the human population. Available EBV vaccines are imminent. Prophylactic vaccines can effectively prevent the spread of infection, whereas therapeutic vaccines mainly stimulate cell-mediated immunity and kill infected cells, thus curbing the development of malignant tumors. Nevertheless, there are still no approved EBV vaccines after decades of effort. The complexity of the EBV life cycle, the lack of appropriate animal models, and the limited reports on adjuvant selection and immune responses are gravely impeding progress in EBV vaccines. The soluble gp350 vaccine could reduce the incidence of infectious mononucleosis (IM), which seemed to offer hope, but could not prevent EBV infection. Continuous research and vaccine trials provide deep insights into the structural biology of viruses, the designs for immunogenicity, and the evolving vaccine platforms. Moreover, the new vaccine candidates are expected to achieve further success via combined immunization to elicit both a dual protection of B cells and epithelial cells, and sustainable immunization against infected cells at several phases of infection.
Collapse
Affiliation(s)
- Jing Cai
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (J.C.); (B.Z.); (Y.L.); (T.A.); (F.F.)
| | - Bodou Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (J.C.); (B.Z.); (Y.L.); (T.A.); (F.F.)
| | - Yuqi Li
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (J.C.); (B.Z.); (Y.L.); (T.A.); (F.F.)
| | - Wanfang Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (W.Z.); (W.L.)
| | - Toshihiro Akihisa
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (J.C.); (B.Z.); (Y.L.); (T.A.); (F.F.)
- Research Institute for Science and Technology, Tokyo University of Science, Chiba 278-8510, Japan
| | - Wei Li
- Faculty of Pharmaceutical Sciences, Toho University, Chiba 274-8510, Japan; (W.L.); (T.K.)
| | - Takashi Kikuchi
- Faculty of Pharmaceutical Sciences, Toho University, Chiba 274-8510, Japan; (W.L.); (T.K.)
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (W.Z.); (W.L.)
| | - Feng Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (J.C.); (B.Z.); (Y.L.); (T.A.); (F.F.)
- Jiangsu Food and Pharmaceutical Science College, Huaian 223003, China
| | - Jie Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (J.C.); (B.Z.); (Y.L.); (T.A.); (F.F.)
- Jiangsu Food and Pharmaceutical Science College, Huaian 223003, China
- Correspondence:
| |
Collapse
|
21
|
Bauer M, Jasinski-Bergner S, Mandelboim O, Wickenhauser C, Seliger B. Epstein-Barr Virus-Associated Malignancies and Immune Escape: The Role of the Tumor Microenvironment and Tumor Cell Evasion Strategies. Cancers (Basel) 2021; 13:cancers13205189. [PMID: 34680337 PMCID: PMC8533749 DOI: 10.3390/cancers13205189] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/06/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The Epstein–Barr virus, also termed human herpes virus 4, is a human pathogenic double-stranded DNA virus. It is highly prevalent and has been linked to the development of 1–2% of cancers worldwide. EBV-associated malignancies encompass various structural and epigenetic alterations. In addition, EBV-encoded gene products and microRNAs interfere with innate and adaptive immunity and modulate the tumor microenvironment. This review provides an overview of the characteristic features of EBV with a focus on the intrinsic and extrinsic immune evasion strategies, which contribute to EBV-associated malignancies. Abstract The detailed mechanisms of Epstein–Barr virus (EBV) infection in the initiation and progression of EBV-associated malignancies are not yet completely understood. During the last years, new insights into the mechanisms of malignant transformation of EBV-infected cells including somatic mutations and epigenetic modifications, their impact on the microenvironment and resulting unique immune signatures related to immune system functional status and immune escape strategies have been reported. In this context, there exists increasing evidence that EBV-infected tumor cells can influence the tumor microenvironment to their own benefit by establishing an immune-suppressive surrounding. The identified mechanisms include EBV gene integration and latent expression of EBV-infection-triggered cytokines by tumor and/or bystander cells, e.g., cancer-associated fibroblasts with effects on the composition and spatial distribution of the immune cell subpopulations next to the infected cells, stroma constituents and extracellular vesicles. This review summarizes (i) the typical stages of the viral life cycle and EBV-associated transformation, (ii) strategies to detect EBV genome and activity and to differentiate various latency types, (iii) the role of the tumor microenvironment in EBV-associated malignancies, (iv) the different immune escape mechanisms and (v) their clinical relevance. This gained information will enhance the development of therapies against EBV-mediated diseases to improve patient outcome.
Collapse
Affiliation(s)
- Marcus Bauer
- Department of Pathology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 14, 06112 Halle (Saale), Germany; (M.B.); (C.W.)
| | - Simon Jasinski-Bergner
- Department of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112 Halle (Saale), Germany;
| | - Ofer Mandelboim
- Department of Immunology, Faculty of Medicine, The Hebrew University of Jerusalem, En Kerem, P.O. Box 12271, Jerusalem 91120, Israel;
| | - Claudia Wickenhauser
- Department of Pathology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 14, 06112 Halle (Saale), Germany; (M.B.); (C.W.)
| | - Barbara Seliger
- Department of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112 Halle (Saale), Germany;
- Fraunhofer Institute for Cell Therapy and Immunology, Perlickstr. 1, 04103 Leipzig, Germany
- Correspondence: ; Tel.: +49-(345)-557-1357
| |
Collapse
|
22
|
Stress-Induced Epstein-Barr Virus Reactivation. Biomolecules 2021; 11:biom11091380. [PMID: 34572593 PMCID: PMC8470332 DOI: 10.3390/biom11091380] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022] Open
Abstract
Epstein-Barr virus (EBV) is typically found in a latent, asymptomatic state in immunocompetent individuals. Perturbations of the host immune system can stimulate viral reactivation. Furthermore, there are a myriad of EBV-associated illnesses including various cancers, post-transplant lymphoproliferative disease, and autoimmune conditions. A thorough understanding of this virus, and the interplay between stress and the immune system, is essential to establish effective treatment. This review will provide a summary of the interaction between both psychological and cellular stressors resulting in EBV reactivation. It will examine mechanisms by which EBV establishes and maintains latency and will conclude with a brief overview of treatments targeting EBV.
Collapse
|
23
|
Münz C. Immune Escape by Non-coding RNAs of the Epstein Barr Virus. Front Microbiol 2021; 12:657387. [PMID: 34234755 PMCID: PMC8257079 DOI: 10.3389/fmicb.2021.657387] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/28/2021] [Indexed: 01/20/2023] Open
Abstract
Epstein Barr virus (EBV) is one of the most successful pathogens of humans, persistently colonizing more than 95% of the adult human population. At the same time EBV encodes oncogenes that can readily transform human B cells in culture and threaten healthy virus carriers with lymphomagenesis. Cytotoxic lymphocytes have been identified in experimental models and by primary immunodeficiencies as the main protective immune compartments controlling EBV. EBV has reached a stalemate with these cytotoxic T and innate lymphocytes to ensure persistence in most infected humans. Recent evidence suggests that the non-coding RNAs of the virus contribute to viral immune escape to prevent immune eradication. This knowledge might be used in the future to attenuate EBV for vaccine development against this human tumor virus that was discovered more than 55 years ago.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Chen WJ, Xu WN, Wang HY, Chen XX, Li XQ, Xie SH, Lin DF, Cao SM. Plasma Epstein-Barr virus DNA and risk of nasopharyngeal carcinoma in a prospective seropositive population. BMC Cancer 2021; 21:651. [PMID: 34074258 PMCID: PMC8168313 DOI: 10.1186/s12885-021-08408-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Plasma Epstein-Barr virus (EBV) DNA is considered a biomarker for nasopharyngeal carcinoma (NPC). However, its long-term role in NPC development is unclear. MATERIALS AND METHODS A total of 1363 participants seropositive for EBV VCA-IgA and EBNA1-IgA in a community-based NPC screening program in southern China were tested for plasma EBV DNA levels by real-time qPCR between 2008 and 2015. New NPC cases were confirmed by active follow-up approach and linkage to local cancer registry through the end of 2016. Cox proportional hazards regression analysis was performed to calculate the hazard ratios (HRs) for NPC risk with plasma EBV DNA. RESULTS Thirty patients were newly diagnosed during a median 7.5 years follow-up. NPC incidence increased with the plasma EBV DNA load ranging from 281.46 to 10,074.47 per 100,000 person-years in participants with undetectable and ≥ 1000 copies/ml levels; the corresponding cumulative incidence rates were 1.73 and 50%. Furthermore, plasma EBV DNA loads conferred an independent risk for NPC development after adjustment for other risk factors, with HRs of 7.63 for > 3-999 copies/ml and 39.79 for ≥1000 copies/ml. However, the HRs decreased gradually after excluding NPC cases detected in the first 2 to 3 years and became statistically nonsignificant by excluding cases detected during the first 4 years. CONCLUSION Elevated plasma EBV DNA can predict NPC risk over 3 years. Monitoring plasma EBV DNA can be used as a complementary approach to EBV serological antibody-based screening for NPC.
Collapse
Affiliation(s)
- Wen-Jie Chen
- Department of Cancer Prevention, Cancer Prevention Center, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wen-Na Xu
- Department of Medicine Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hai-Yun Wang
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Xia Chen
- Department of Cancer Prevention, Cancer Prevention Center, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xue-Qi Li
- Department of Cancer Prevention, Cancer Prevention Center, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Shang-Hang Xie
- Department of Cancer Prevention, Cancer Prevention Center, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dong-Feng Lin
- Department of Cancer Prevention, Cancer Prevention Center, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Su-Mei Cao
- Department of Cancer Prevention, Cancer Prevention Center, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China.
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
25
|
Abstract
Antigen recognition by the B cell receptor (BCR) is a physiological trigger for reactivation of Epstein-Barr virus (EBV) and can be recapitulated in vitro by cross-linking of surface immunoglobulins. Previously, we identified a subset of EBV microRNAs (miRNAs) that attenuate BCR signal transduction and subsequently dampen lytic reactivation in B cells. The roles of host miRNAs in the EBV lytic cycle are not completely understood. Here, we profiled the small RNAs in reactivated Burkitt lymphoma cells and identified several miRNAs, such as miR-141, that are induced upon BCR cross-linking. Notably, EBV encodes a viral miRNA, miR-BART9, with sequence homology to miR-141. To better understand the functions of these two miRNAs, we examined their molecular targets and experimentally validated multiple candidates commonly regulated by both miRNAs. Targets included B cell transcription factors and known regulators of EBV immediate-early genes, leading us to hypothesize that these miRNAs modulate kinetics of the lytic cascade in B cells. Through functional assays, we identified roles for miR-141 and EBV miR-BART9 and one specific target, FOXO3, in progression of the lytic cycle. Our data support a model whereby EBV exploits BCR-responsive miR-141 and further mimics activity of this miRNA family via a viral miRNA to promote productive lytic replication. IMPORTANCE EBV is a human pathogen associated with several malignancies. A key aspect of lifelong virus persistence is the ability to switch between latent and lytic replication modes. The mechanisms governing latency, reactivation, and progression of the lytic cycle are only partly understood. This study reveals that specific miRNAs can act to support the EBV lytic phase following BCR-mediated reactivation triggers. Furthermore, this study identifies a role for FOXO3, commonly suppressed by both host and viral miRNAs, in modulating progression of the EBV lytic cycle.
Collapse
|
26
|
Long C, Xu QB, Ding L, Yang L, Ji W, Gao F, Ji Y. Triptolide inhibits human telomerase reverse transcriptase by downregulating translation factors SP1 and c-Myc in Epstein-Barr virus-positive B lymphocytes. Oncol Lett 2021; 21:280. [PMID: 33732356 PMCID: PMC7905526 DOI: 10.3892/ol.2021.12541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/17/2020] [Indexed: 11/06/2022] Open
Abstract
Epstein-Barr virus (EBV) mainly causes infectious mononucleosis and is associated with several neoplasms, including Burkitt's lymphoma, nasopharyngeal carcinoma and lymphoproliferative disease. Human telomerase reverse transcriptase (hTERT) regulates enzymatic activity of telomerase and is closely associated with tumorigenesis and senescence evasion. Triptolide (TP) is a diterpenoid triepoxide, with a broad-spectrum anticancer and immunosuppressive bioactivity profile. The present study investigated whether TP inhibited hTERT expression and suppressed its activity. The mRNA and protein levels of hTERT were examined by reverse transcription-quantitative PCR and western blotting. The activity of hTERT promoter was determined by dual-luciferase reporter assay. Cell Counting Kit-8 assays were performed to analyze cell proliferation. The present study used EBV-positive B lymphoma cells as a model system, and the results demonstrated that TP significantly decreased hTERT transcription and protein expression. Mechanistically, TP attenuated the hTERT promoter activity by downregulating the expression levels of specificityprotein 1 and c-Myc transcription factors. Consistently, inhibition of hTERT via shRNA transfection efficiently enhanced the suppression of cell proliferation by TP. Furthermore, TP increased virus latent replication and promoted the lytic cycle of EBV in EBV-positive B lymphoma cells, increasing the number of lytic cells and inhibiting the viability of tumor cells. Taken together, the results of the present study revealed a molecular mechanism of the pharmacological inhibition of tumor cell proliferation by TP, encouraging the translation of TP-based therapeutics in EBV-positive B lymphoma treatment.
Collapse
Affiliation(s)
- Cong Long
- Clinical Laboratory, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Qiu-Bo Xu
- Clinical Laboratory, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Li Ding
- Clinical Laboratory, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Liu Yang
- Clinical Laboratory, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Wei Ji
- Clinical Laboratory, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Feng Gao
- Department of General Surgery, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Yong Ji
- Department of General Surgery, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| |
Collapse
|
27
|
Bayda N, Tilloy V, Chaunavel A, Bahri R, Halabi MA, Feuillard J, Jaccard A, Ranger-Rogez S. Comprehensive Epstein-Barr Virus Transcriptome by RNA-Sequencing in Angioimmunoblastic T Cell Lymphoma (AITL) and Other Lymphomas. Cancers (Basel) 2021; 13:610. [PMID: 33557089 PMCID: PMC7913808 DOI: 10.3390/cancers13040610] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/24/2021] [Accepted: 01/28/2021] [Indexed: 12/16/2022] Open
Abstract
The Epstein-Barr virus (EBV) is associated with angioimmunoblastic T cell lymphoma (AITL) in more than 80% of cases. Few studies have focused on this association and it is not clear now what role the virus plays in this pathology. We used next-generation sequencing (NGS) to study EBV transcriptome in 14 AITLs compared to 21 other lymphoma samples and 11 cell lines including 4 lymphoblastoid cell lines (LCLs). Viral transcripts were recovered using capture probes and sequencing was performed on Illumina. Bam-HI A rightward transcripts (BARTs) were the most latency transcripts expressed in AITLs, suggesting they may play a role in this pathology. Thus, BARTs, already described as highly expressed in carcinoma cells, are also very present in AITLs and other lymphomas. They were poorly expressed in cell lines other than LCLs. AITLs showed a latency IIc, with BNLF2a gene expression. For most AITLs, BCRF1, which encodes a homologous protein of human interleukin 10, vIL-10, was in addition expressed. This co-expression can contribute to immune escape and survival of infected cells. Considering these results, it can be assumed that EBV plays a pathogenic role in AITLs.
Collapse
Affiliation(s)
- Nader Bayda
- Microbiology Department, UMR CNRS 7276, INSERM U1262, Faculty of Pharmacy, 87025 Limoges, France; (N.B.); (R.B.); (M.A.H.)
- Department of Infectious Disease Control, Faculty of Public Health, Jinan University, Tripoli 1300, Lebanon
| | - Valentin Tilloy
- National Reference Center for Herpesviruses, Bioinformatics, Centre de Biologie Recherche et Santé, 87000 Limoges, France;
| | - Alain Chaunavel
- Pathology Department, Centre de Biologie Recherche et Santé, 87000 Limoges, France;
| | - Racha Bahri
- Microbiology Department, UMR CNRS 7276, INSERM U1262, Faculty of Pharmacy, 87025 Limoges, France; (N.B.); (R.B.); (M.A.H.)
| | - Mohamad Adnan Halabi
- Microbiology Department, UMR CNRS 7276, INSERM U1262, Faculty of Pharmacy, 87025 Limoges, France; (N.B.); (R.B.); (M.A.H.)
| | - Jean Feuillard
- Biological Hematology Department, UMR CNRS 7276, INSERM U1262, Centre de Biologie Recherche et Santé, 87000 Limoges, France;
| | - Arnaud Jaccard
- Clinical Hematology Department, UMR CNRS 7276, INSERM U1262, University Hospital Dupuytren, 87042 Limoges, France;
| | - Sylvie Ranger-Rogez
- Microbiology Department, UMR CNRS 7276, INSERM U1262, Faculty of Pharmacy, 87025 Limoges, France; (N.B.); (R.B.); (M.A.H.)
- Virology Department, UMR CNRS 7276, INSERM U1262, Centre de Biologie Recherche et Santé, 87000 Limoges, France
| |
Collapse
|
28
|
Houen G, Trier NH, Frederiksen JL. Epstein-Barr Virus and Multiple Sclerosis. Front Immunol 2020; 11:587078. [PMID: 33391262 PMCID: PMC7773893 DOI: 10.3389/fimmu.2020.587078] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a neurologic disease affecting myelinated nerves in the central nervous system (CNS). The disease often debuts as a clinically isolated syndrome, e.g., optic neuritis (ON), which later develops into relapsing-remitting (RR) MS, with temporal attacks or primary progressive (PP) MS. Characteristic features of MS are inflammatory foci in the CNS and intrathecal synthesis of immunoglobulins (Igs), measured as an IgG index, oligoclonal bands (OCBs), or specific antibody indexes. Major predisposing factors for MS are certain tissue types (e.g., HLA DRB1*15:01), vitamin D deficiency, smoking, obesity, and infection with Epstein-Barr virus (EBV). Many of the clinical signs of MS described above can be explained by chronic/recurrent EBV infection and current models of EBV involvement suggest that RRMS may be caused by repeated entry of EBV-transformed B cells to the CNS in connection with attacks, while PPMS may be caused by more chronic activity of EBV-transformed B cells in the CNS. In line with the model of EBV's role in MS, new treatments based on monoclonal antibodies (MAbs) targeting B cells have shown good efficacy in clinical trials both for RRMS and PPMS, while MAbs inhibiting B cell mobilization and entry to the CNS have shown efficacy in RRMS. Thus, these agents, which are now first line therapy in many patients, may be hypothesized to function by counteracting a chronic EBV infection.
Collapse
Affiliation(s)
- Gunnar Houen
- Institute of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Rigshospitalet, Glostrup, Denmark
| | | | - Jette Lautrup Frederiksen
- Department of Neurology, Rigshospitalet, Glostrup, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
Abstract
Epstein-Barr virus (EBV) infects 95% of adults worldwide and causes infectious mononucleosis. EBV is associated with endemic Burkitt lymphoma, Hodgkin lymphoma, posttransplant lymphomas, nasopharyngeal and gastric carcinomas. In these cancers and in most infected B-cells, EBV maintains a state of latency, where nearly 80 lytic cycle antigens are epigenetically suppressed. To gain insights into host epigenetic factors necessary for EBV latency, we recently performed a human genome-wide CRISPR screen that identified the chromatin assembly factor CAF1 as a putative Burkitt latency maintenance factor. CAF1 loads histones H3 and H4 onto newly synthesized host DNA, though its roles in EBV genome chromatin assembly are uncharacterized. Here, we found that CAF1 depletion triggered lytic reactivation and virion secretion from Burkitt cells, despite also strongly inducing interferon-stimulated genes. CAF1 perturbation diminished occupancy of histones 3.1 and 3.3 and of repressive histone 3 lysine 9 and 27 trimethyl (H3K9me3 and H3K27me3) marks at multiple viral genome lytic cycle regulatory elements. Suggestive of an early role in establishment of latency, EBV strongly upregulated CAF1 expression in newly infected primary human B-cells prior to the first mitosis, and histone 3.1 and 3.3 were loaded on the EBV genome by this time point. Knockout of CAF1 subunit CHAF1B impaired establishment of latency in newly EBV-infected Burkitt cells. A nonredundant latency maintenance role was also identified for the DNA synthesis-independent histone 3.3 loader histone regulatory homologue A (HIRA). Since EBV latency also requires histone chaperones alpha thalassemia/mental retardation syndrome X-linked chromatin remodeler (ATRX) and death domain-associated protein (DAXX), EBV coopts multiple host histone pathways to maintain latency, and these are potential targets for lytic induction therapeutic approaches.IMPORTANCE Epstein-Barr virus (EBV) was discovered as the first human tumor virus in endemic Burkitt lymphoma, the most common childhood cancer in sub-Saharan Africa. In Burkitt lymphoma and in 200,000 EBV-associated cancers per year, epigenetic mechanisms maintain viral latency, during which lytic cycle factors are silenced. This property complicated EBV's discovery and facilitates tumor immunoevasion. DNA methylation and chromatin-based mechanisms contribute to lytic gene silencing. Here, we identified histone chaperones CAF1 and HIRA, which have key roles in host DNA replication-dependent and replication-independent pathways, respectively, as important for EBV latency. EBV strongly upregulates CAF1 in newly infected B-cells, where viral genomes acquire histone 3.1 and 3.3 variants prior to the first mitosis. Since histone chaperones ATRX and DAXX also function in maintenance of EBV latency, our results suggest that EBV coopts multiple histone pathways to reprogram viral genomes and highlight targets for lytic induction therapeutic strategies.
Collapse
|
30
|
Targeted Therapies for Epstein-Barr Virus-Associated Lymphomas. Cancers (Basel) 2020; 12:cancers12092565. [PMID: 32916819 PMCID: PMC7564798 DOI: 10.3390/cancers12092565] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Epstein-Barr virus (EBV) is the first-discovered and important human tumor virus. It infects more than 90% of human population and induces various lymphomas. Development of specific targeted therapies is very critical for treatment of EBV-induced lymphomas, but it remains a great challenge. In this review, we introduced the current progress of EBV-specific therapies and the promising approaches that can be developed as novel targeted therapies, which involve protective or therapeutic strategies to target these lymphomas on different levels. This work will provide new insights into the development of new targeted therapies against EBV-associated lymphomas. Abstract The Epstein-Barr virus (EBV) is the first human tumor virus identified that can transform quiescent B lymphocytes into lymphoblastoid cell lines (LCLs) in vitro. EBV can establish asymptomatic life-long persistence and is associated with multiple human malignancies, including non-Hodgkin lymphoma and Hodgkin lymphoma, as well as infectious mononucleosis. Although EBV-associated lymphomagenesis has been investigated for over 50 years, viral-mediated transformation is not completely understood, and the development of EBV-specific therapeutic strategies to treat the associated cancers is still a major challenge. However, the rapid development of several novel therapies offers exciting possibilities to target EBV-induced lymphomas. This review highlights targeted therapies with potential for treating EBV-associated lymphomas, including small molecule inhibitors, immunotherapy, cell therapy, preventative and therapeutic vaccines, and other potent approaches, which are novel strategies for controlling, preventing, and treating these viral-induced malignances.
Collapse
|
31
|
Identification and Cloning of a New Western Epstein-Barr Virus Strain That Efficiently Replicates in Primary B Cells. J Virol 2020; 94:JVI.01918-19. [PMID: 32102884 DOI: 10.1128/jvi.01918-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/14/2020] [Indexed: 11/20/2022] Open
Abstract
The Epstein-Barr virus (EBV) causes human cancers, and epidemiological studies have shown that lytic replication is a risk factor for some of these tumors. This fits with the observation that EBV M81, which was isolated from a Chinese patient with nasopharyngeal carcinoma, induces potent virus production and increases the risk of genetic instability in infected B cells. To find out whether this property extends to viruses found in other parts of the world, we investigated 22 viruses isolated from Western patients. While one-third of the viruses hardly replicated, the remaining viruses showed variable levels of replication, with three isolates replicating at levels close to that of M81 in B cells. We cloned one strongly replicating virus into a bacterial artificial chromosome (BAC); the resulting recombinant virus (MSHJ) retained the properties of its nonrecombinant counterpart and showed similarities to M81, undergoing lytic replication in vitro and in vivo after 3 weeks of latency. In contrast, B cells infected with the nonreplicating Western B95-8 virus showed early but abortive replication accompanied by cytoplasmic BZLF1 expression. Sequencing confirmed that rMSHJ is a Western virus, being genetically much closer to B95-8 than to M81. Spontaneous replication in rM81- and rMSHJ-infected B cells was dependent on phosphorylated Btk and was inhibited by exposure to ibrutinib, opening the way to clinical intervention in patients with abnormal EBV replication. As rMSHJ contains the complete EBV genome and induces lytic replication in infected B cells, it is ideal to perform genetic analyses of all viral functions in Western strains and their associated diseases.IMPORTANCE The Epstein-Barr virus (EBV) infects the majority of the world population but causes different diseases in different countries. Evidence that lytic replication, the process that leads to new virus progeny, is linked to cancer development is accumulating. Indeed, viruses such as M81 that were isolated from Far Eastern nasopharyngeal carcinomas replicate strongly in B cells. We show here that some viruses isolated from Western patients, including the MSHJ strain, share this property. Moreover, replication of both M81 and of MSHJ was sensitive to ibrutinib, a commonly used drug, thereby opening an opportunity for therapeutic intervention. Sequencing of MSHJ showed that this virus is quite distant from M81 and is much closer to nonreplicating Western viruses. We conclude that Western EBV strains are heterogeneous, with some viruses being able to replicate more strongly and therefore being potentially more pathogenic than others, and that the virus sequence information alone cannot predict this property.
Collapse
|
32
|
Abstract
The etiopathogenesis of severe periodontitis includes herpesvirus-bacteria coinfection. This article evaluates the pathogenicity of herpesviruses (cytomegalovirus and Epstein-Barr virus) and periodontopathic bacteria (Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis) and coinfection of these infectious agents in the initiation and progression of periodontitis. Cytomegalovirus and A. actinomycetemcomitans/P. gingivalis exercise synergistic pathogenicity in the development of localized ("aggressive") juvenile periodontitis. Cytomegalovirus and Epstein-Barr virus are associated with P. gingivalis in adult types of periodontitis. Periodontal herpesviruses that enter the general circulation may also contribute to disease development in various organ systems. A 2-way interaction is likely to occur between periodontal herpesviruses and periodontopathic bacteria, with herpesviruses promoting bacterial upgrowth, and bacterial factors reactivating latent herpesviruses. Bacterial-induced gingivitis may facilitate herpesvirus colonization of the periodontium, and herpesvirus infections may impede the antibacterial host defense and alter periodontal cells to predispose for bacterial adherence and invasion. Herpesvirus-bacteria synergistic interactions, are likely to comprise an important pathogenic determinant of aggressive periodontitis. However, mechanistic investigations into the molecular and cellular interaction between periodontal herpesviruses and bacteria are still scarce. Herpesvirus-bacteria coinfection studies may yield significant new discoveries of pathogenic determinants, and drug and vaccine targets to minimize or prevent periodontitis and periodontitis-related systemic diseases.
Collapse
Affiliation(s)
- Casey Chen
- Division of Periodontology, Diagnostic Sciences & Dental Hygiene, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Jørgen Slots
- Division of Periodontology, Diagnostic Sciences & Dental Hygiene, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
33
|
[Dynamic changes of cellular environment during Epstein-Barr virus productive replication]. Uirusu 2020; 70:83-90. [PMID: 33967117 DOI: 10.2222/jsv.70.83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Productive (lytic) replication of DNA viruses elicits host cell DNA damage responses, which cause both beneficial and detrimental effects on viral replication. Viruses utilize them and selectively cancel the 'noisy' downstream signaling pathways, leading to maintain high S-phase CDK activities required for viral replication. To achieve this fine tuning of cellular environment, herpesviruses encode many (>70) genes in their genome, which are expressed in a strictly regulated temporal cascade (immediate-early, early, and late). Here, I introduce and discuss how Epstein-Barr virus, an oncogenic herpesvirus, hijacks the cellular environment and adapt it for the progeny production.
Collapse
|
34
|
Pharmacologic Activation of Lytic Epstein-Barr Virus Gene Expression without Virion Production. J Virol 2019; 93:JVI.00998-19. [PMID: 31341058 DOI: 10.1128/jvi.00998-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Several therapeutic strategies targeting Epstein-Barr virus (EBV)-associated tumors involve upregulation of viral lytic gene expression. Evidence has been presented that the unfolded protein response (UPR) leads to EBV lytic gene expression. Clofoctol, an antibacterial antibiotic, has been reported to upregulate the UPR in prostate cancer cell lines and to slow their growth. We investigated the effects of clofoctol on an EBV-positive Burkitt lymphoma cell line and confirmed the upregulation of all three branches of the UPR and activation of EBV lytic gene expression. While immediate early, early, and late EBV RNAs were all upregulated, immediate early and early viral proteins but not late viral proteins were expressed. Furthermore, infectious virions were not produced. The use of clofoctol in combination with a protein kinase R-like endoplasmic reticulum kinase inhibitor led to expression of late viral proteins. The effects of clofoctol on EBV lytic protein upregulation were not limited to lymphoid tumor cell lines but also occurred in naturally infected epithelial gastric cancer and nasopharyngeal cancer cell lines. An agent that upregulates lytic viral protein expression but that does not lead to the production of infectious virions may have particular value for lytic induction strategies in the clinical setting.IMPORTANCE Epstein-Barr virus is associated with many different cancers. In these cancers the viral genome is predominantly latent; i.e., most viral genes are not expressed, most viral proteins are not synthesized, and new virions are not produced. Some strategies for treating these cancers involve activation of lytic viral gene expression. We identify an antibacterial antibiotic, clofoctol, that is an activator of EBV lytic RNA and protein expression but that does not lead to virion production.
Collapse
|
35
|
Identification of ARKL1 as a Negative Regulator of Epstein-Barr Virus Reactivation. J Virol 2019; 93:JVI.00989-19. [PMID: 31341047 DOI: 10.1128/jvi.00989-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
Epstein-Barr virus (EBV) maintains a life-long infection due to the ability to alternate between latent and lytic modes of replication. Lytic reactivation starts with derepression of the Zp promoter controlling BZLF1 gene expression, which binds and is activated by the c-Jun transcriptional activator. Here, we identified the cellular Arkadia-like 1 (ARKL1) protein as a negative regulator of Zp and EBV reactivation. Silencing of ARKL1 in the context of EBV-positive gastric carcinoma (AGS) cells, nasopharyngeal carcinoma (NPC43) cells, and B (M81) cells led to increased lytic protein expression, whereas overexpression inhibited BZLF1 expression. Similar effects of ARKL1 modulation were seen on BZLF1 transcripts as well as on Zp activity in Zp reporter assays, showing that ARKL1 repressed Zp. Proteomic profiling of ARKL1-host interactions identified c-Jun as an ARKL1 interactor, and reporter assays for Jun transcriptional activity showed that ARKL1 inhibited Jun activity. The ARKL1-Jun interaction required ARKL1 sequences that we previously showed mediated binding to the CK2 kinase regulatory subunit CK2β, suggesting that CK2β might mediate the ARKL1-Jun interaction. This model was supported by the findings that silencing of CK2β, but not the CK2α catalytic subunit, abrogated the ARKL1-Jun interaction and phenocopied ARKL1 silencing in promoting EBV reactivation. Additionally, ARKL1 was associated with Zp in reporter assays and this was increased by additional CK2β. Together, the data indicate that ARKL1 is a negative regulator of Zp and EBV reactivation that acts by inhibiting Jun activity through a CK2β-mediated interaction.IMPORTANCE Epstein-Barr virus (EBV) maintains a life-long infection due to the ability to alternate between latent and lytic modes of replication and is associated with several types of cancer. We have identified a cellular protein (ARKL1) that acts to repress the reactivation of EBV from the latent to the lytic cycle. We show that ARKL1 acts to repress transcription of the EBV lytic switch protein by inhibiting the activity of the cellular transcription factor c-Jun. This not only provides a new mechanism of regulating EBV reactivation but also identifies a novel cellular function of ARKL1 as an inhibitor of Jun-mediated transcription.
Collapse
|
36
|
Li J, Walsh A, Lam TT, Delecluse HJ, El-Guindy A. A single phosphoacceptor residue in BGLF3 is essential for transcription of Epstein-Barr virus late genes. PLoS Pathog 2019; 15:e1007980. [PMID: 31461506 PMCID: PMC6713331 DOI: 10.1371/journal.ppat.1007980] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/12/2019] [Indexed: 02/07/2023] Open
Abstract
Almost one third of herpesvirus proteins are expressed with late kinetics. Many of these late proteins serve crucial structural functions such as formation of virus particles, attachment to host cells and internalization. Recently, we and others identified a group of Epstein-Barr virus early proteins that form a pre-initiation complex (vPIC) dedicated to transcription of late genes. Currently, there is a fundamental gap in understanding the role of post-translational modifications in regulating assembly and function of the complex. Here, we used mass spectrometry to map potential phosphorylation sites in BGLF3, a core component of the vPIC module that connects the BcRF1 viral TATA box binding protein to other components of the complex. We identified threonine 42 (T42) in BGLF3 as a phosphoacceptor residue. T42 is conserved in BGLF3 orthologs encoded by other gamma herpesviruses. Abolishing phosphorylation at T42 markedly reduced expression of vPIC-dependent late genes and disrupted production of new virus particles, but had no effect on early gene expression, viral DNA replication, or expression of vPIC-independent late genes. We complemented failure of BGLF3(T42A) to activate late gene expression by ectopic expression of other components of vPIC. Only BFRF2 and BVLF1 were sufficient to suppress the defect in late gene expression associated with BGLF3(T42A). These results were corroborated by the ability of wild type BGLF3 but not BGLF3(T42A) to form a trimeric complex with BFRF2 and BVLF1. Our findings suggest that phosphorylation of BGLF3 at threonine 42 serves as a new checkpoint for subsequent formation of BFRF2:BGLF3:BVLF1; a trimeric subcomplex essential for transcription of late genes. Our findings provide evidence that post-translational modifications regulate the function of the vPIC nanomachine that initiates synthesis of late transcripts in herpesviruses.
Collapse
Affiliation(s)
- Jinlin Li
- Department of Pediatrics Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Ann Walsh
- Department of Pediatrics Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - TuKiet T. Lam
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, United States of America
- Keck MS and Proteomics Resource, Yale University, New Haven, Connecticut, United States of America
| | - Henri-Jacques Delecluse
- Department of Tumor Virology, German Cancer Research Center, Im Neuenheimer Feld, Heidelberg, Germany
| | - Ayman El-Guindy
- Department of Pediatrics Yale University School of Medicine, New Haven, Connecticut, United States of America
- Yale Cancer Center, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
37
|
Initial Characterization of the Epstein⁻Barr Virus BSRF1 Gene Product. Viruses 2019; 11:v11030285. [PMID: 30901892 PMCID: PMC6466199 DOI: 10.3390/v11030285] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/16/2022] Open
Abstract
Epstein⁻Barr virus (EBV) is a ubiquitous virus that causes infectious mononucleosis and several types of cancer, such as Burkitt lymphoma, T/NK-cell lymphoma, and nasopharyngeal carcinoma. As a herpesvirus, it encodes more than 80 genes, many of which have not been characterized. EBV BamHI S rightward reading frame 1 (BSRF1) encodes a tegument protein that, unlike its homologs herpes simplex virus unique long 51 (UL51) and human cytomegalovirus UL71, has not been extensively investigated. To examine the role of BSRF1, we prepared knockout and revertant strains using the bacterial artificial chromosome system. Unexpectedly, the disruption of the gene had little or no effect on EBV lytic replication and the transformation of primary B cells. However, the knockdown of BSRF1 in B95-8 cells decreased progeny production. An immunofluorescence assay revealed that BSRF1 localized to the Golgi apparatus in the cytoplasm, as did its homologs. BSRF1 also associated with BamHI G leftward reading frame 3.5 (BGLF3.5), BamHI B rightward reading frame 2 (BBRF2), and BamHI A leftward reading frame 1 (BALF1), and BALF1 was incorporated into the tegument fraction with BSRF1. Taken together, our results indicate that BSRF1 plays a role in secondary envelopment or virion egress in the cytoplasm, as do its homolog genes.
Collapse
|
38
|
Sau S, Ghosh SK, Liu YT, Ma CH, Jayaram M. Hitchhiking on chromosomes: A persistence strategy shared by diverse selfish DNA elements. Plasmid 2019; 102:19-28. [PMID: 30726706 DOI: 10.1016/j.plasmid.2019.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 12/12/2022]
Abstract
An underlying theme in the segregation of low-copy bacterial plasmids is the assembly of a 'segrosome' by DNA-protein and protein-protein interactions, followed by energy-driven directed movement. Analogous partitioning mechanisms drive the segregation of host chromosomes as well. Eukaryotic extra-chromosomal elements, exemplified by budding yeast plasmids and episomes of certain mammalian viruses, harbor partitioning systems that promote their physical association with chromosomes. In doing so, they indirectly take advantage of the spindle force that directs chromosome movement to opposite cell poles. Molecular-genetic, biochemical and cell biological studies have revealed several unsuspected aspects of 'chromosome hitchhiking' by the yeast 2-micron plasmid, including the ability of plasmid sisters to associate symmetrically with sister chromatids. As a result, the plasmid overcomes the 'mother bias' experienced by plasmids lacking a partitioning system, and elevates itself to near chromosome status in equal segregation. Chromosome association for stable propagation, without direct energy expenditure, may also be utilized by a small minority of bacterial plasmids-at least one case has been reported. Given the near perfect accuracy of chromosome segregation, it is not surprising that elements residing in evolutionarily distant host organisms have converged upon the common strategy of gaining passage to daughter cells as passengers on chromosomes.
Collapse
Affiliation(s)
- Soumitra Sau
- Amity Institute of Biotechnology, Amity University Kolkata, Kolkata 700135, India
| | - Santanu Kumar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Yen-Ting Liu
- Department of Molecular Biosciences, UT Austin, Austin, TX TX7 8712, USA
| | - Chien-Hui Ma
- Department of Molecular Biosciences, UT Austin, Austin, TX TX7 8712, USA
| | - Makkuni Jayaram
- Department of Molecular Biosciences, UT Austin, Austin, TX TX7 8712, USA.
| |
Collapse
|
39
|
Regulation of Epstein-Barr Virus Life Cycle and Cell Proliferation by Histone H3K27 Methyltransferase EZH2 in Akata Cells. mSphere 2018; 3:3/6/e00478-18. [PMID: 30487153 PMCID: PMC6262262 DOI: 10.1128/msphere.00478-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epigenetic modifications play a pivotal role in the expression of the genes of Epstein-Barr virus (EBV). We found that de novo EBV infection of primary B cells caused moderate induction of enhancer of zeste homolog 2 (EZH2), the major histone H3 lysine 27 (K27) methyltransferase. To investigate the role of EZH2, we knocked out the EZH2 gene in EBV-negative Akata cells by the CRISPR/Cas9 system and infected the cells with EBV, followed by selection of EBV-positive cells. During the latent state, growth of EZH2-knockout (KO) cells was significantly slower after infection compared to wild-type controls, despite similar levels of viral gene expression between cell lines. After induction of the lytic cycle by anti-IgG, KO of EZH2 caused notable induction of expression of both latent and lytic viral genes, as well as increases in both viral DNA replication and progeny production. These results demonstrate that EZH2 is crucial for the intricate epigenetic regulation of not only lytic but also latent gene expression in Akata cells.IMPORTANCE The life cycle of EBV is regulated by epigenetic modifications, such as CpG methylation and histone modifications. Here, we found that the expression of EZH2, which encodes a histone H3K27 methyltransferase, was induced by EBV infection; therefore, we generated EZH2-KO cells to investigate the role of EZH2 in EBV-infected Akata B cells. Disruption of EZH2 resulted in increased expression of EBV genes during the lytic phase and, therefore, efficient viral replication and progeny production. Our results shed light on the mechanisms underlying reactivation from an epigenetic point of view and further suggest a role for EZH2 as a form of innate immunity that restricts viral replication in infected cells.
Collapse
|
40
|
Natural Variations in BRLF1 Promoter Contribute to the Elevated Reactivation Level of Epstein-Barr Virus in Endemic Areas of Nasopharyngeal Carcinoma. EBioMedicine 2018; 37:101-109. [PMID: 30420297 PMCID: PMC6286269 DOI: 10.1016/j.ebiom.2018.10.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
Background Epstein-Barr virus (EBV) infection is a crucial risk factor for nasopharyngeal carcinoma (NPC), but the mechanism for its elevated activation level in NPC endemic areas remains unclear. This study aims to identify the EBV natural variations contributed to the different reactivation potential between NPC endemic and non-endemic areas. Methods 1030 subjects were recruited in China, including 303 healthy individuals from two NPC non-endemic areas, 483 healthy people from three endemic areas and 244 NPC patients. Among which, saliva DNA samples from 244 participants were sequenced for the EBV immediate early (IE) genes of BRLF1 and BZLF1, their promoters were included; the rest 786 subjects were used for the validation of significant variations among three different populations. Haplotype and population structure analysis were conducted. Dual-luciferase assay was used to detect the promoter activity. Results A total of 246 distinct variations were detected, 29 showed significant difference in the frequencies between healthy people from NPC endemic area and non-endemic area. Population structure analysis clustered EBV strains into 9 subgroups mostly in accordance with the geographical origin of samples. Interestingly, two EBV genotypes, Rp-V1 and Rp-V2, were identified according to the linkage relationship of the variations in BRLF1 promoter (Rp). Rp-V1 has higher frequency in NPC endemic areas than in non-endemic areas (52.38% vs 18.15%, P = 2.07 × 10−14), and was associated with higher oral EBV DNA levels (adjusted OR = 1.64, 95% CI = 1.21–2.24, P = .002), suggesting a more powerful activation ability of Rp-V1 than that of the prototype Rp-of the EBV strain; On the contrary, Rp-V2 has higher frequency in NPC non-endemic areas than in endemic areas (18.48% vs 0.38%, P = 1.17 × 10−7), might represent a reduced activation potential of EBV. Further dual-luciferase assay showed Rp-V1 has higher promoter activity while compared with Rp-V2 (P < .0001). Notably, Rp-V1 impaired the transcription repression effect of YY1 while Rp-V2 strengthened the transcription repression effect of EBF1 on Rp. In addition, significant differences of Rta 393–407 CTL epitope which may influence the recognition of Rta by CD8+ T cells were detected between healthy people from NPC endemic area and non-endemic area. Conclusions This study identified natural variations in cis-acting elements (YY1 and EBF1) of EBV Rp altering Rp transcription activities, which may contribute to the elevated EBV activation level in NPC endemic areas than non-endemic areas.
Collapse
|
41
|
Tsao SW, Tsang CM, Lo KW. Epstein-Barr virus infection and nasopharyngeal carcinoma. Philos Trans R Soc Lond B Biol Sci 2018; 372:rstb.2016.0270. [PMID: 28893937 DOI: 10.1098/rstb.2016.0270] [Citation(s) in RCA: 371] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2017] [Indexed: 12/24/2022] Open
Abstract
Epstein-Barr virus (EBV) is associated with multiple types of human cancer, including lymphoid and epithelial cancers. The closest association with EBV infection is seen in undifferentiated nasopharyngeal carcinoma (NPC), which is endemic in the southern Chinese population. A strong association between NPC risk and the HLA locus at chromosome 6p has been identified, indicating a link between the presentation of EBV antigens to host immune cells and NPC risk. EBV infection in NPC is clonal in origin, strongly suggesting that NPC develops from the clonal expansion of a single EBV-infected cell. In epithelial cells, the default program of EBV infection is lytic replication. However, latent infection is the predominant mode of EBV infection in NPC. The establishment of latent EBV infection in pre-invasive nasopharyngeal epithelium is believed to be an early stage of NPC pathogenesis. Recent genomic study of NPC has identified multiple somatic mutations in the upstream negative regulators of NF-κB signalling. Dysregulated NF-κB signalling may contribute to the establishment of latent EBV infection in NPC. Stable EBV infection and the expression of latent EBV genes are postulated to drive the transformation of pre-invasive nasopharyngeal epithelial cells to cancer cells through multiple pathways.This article is part of the themed issue 'Human oncogenic viruses'.
Collapse
Affiliation(s)
- Sai Wah Tsao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Chi Man Tsang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology and State Key Laboratory in Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| |
Collapse
|
42
|
Ramayanti O, Brinkkemper M, Verkuijlen SAWM, Ritmaleni L, Go ML, Middeldorp JM. Curcuminoids as EBV Lytic Activators for Adjuvant Treatment in EBV-Positive Carcinomas. Cancers (Basel) 2018; 10:cancers10040089. [PMID: 29565326 PMCID: PMC5923344 DOI: 10.3390/cancers10040089] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 12/24/2022] Open
Abstract
Epstein-Barr virus (EBV) persists in nasopharyngeal (NPC) and gastric carcinomas (EBVaGC) in a tightly latent form. Cytolytic virus activation (CLVA) therapy employs gemcitabine and valproic acid (GCb+VPA) to reactivate latent EBV into the lytic phase and antiviral valganciclovir to enhance cell death and prevent virus production. CLVA treatment has proven safe in phase-I/II trials with promising clinical responses in patients with recurrent NPC. However, a major challenge is to maximize EBV lytic reactivation by CLVA. Curcumin, a dietary spice used in Asian countries, is known for its antitumor property and therapeutic potential. Novel curcuminoids that were developed to increase efficacy and bioavailability may serve as oral CLVA adjuvants. We investigated the potential of curcumin and its analogs (curcuminoids) to trigger the EBV lytic cycle in EBVaGC and NPC cells. EBV-reactivating effects were measured by immunoblot and immunofluorescence using monoclonal antibodies specific for EBV lytic proteins. Two of the hit compounds (41, EF24) with high lytic inducing activity were further studied for their synergistic or antagonistic effects when combined with GCb+VPA and analyzed by cytotoxicity and mRNA profiling assays to measure the EBV reactivation. Curcuminoid as a single agent significantly induced EBV reactivation in recombinant GC and NPC lines. The drug effects were dose- and time-dependent. Micromolar concentration of curcuminoid EF24 enhanced the CLVA effect in all cell systems except SNU719, a naturally infected EBVaGC cell that carries a more tightly latent viral genome. These findings indicated that EF24 has potential as EBV lytic activator and may serve as an adjuvant in CLVA treatment.
Collapse
Affiliation(s)
- Octavia Ramayanti
- Department of Pathology, VU University Medical Center, 1081HV Amsterdam, The Netherlands.
| | - Mitch Brinkkemper
- Department of Pathology, VU University Medical Center, 1081HV Amsterdam, The Netherlands.
| | | | - Leni Ritmaleni
- Laboratory of Medicinal Chemistry, Gadjah Mada University, Yogyakarta 55281, Indonesia.
| | - Mei Lin Go
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore.
| | - Jaap M Middeldorp
- Department of Pathology, VU University Medical Center, 1081HV Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Cavallari I, Scattolin G, Silic-Benussi M, Raimondi V, D'Agostino DM, Ciminale V. Mitochondrial Proteins Coded by Human Tumor Viruses. Front Microbiol 2018; 9:81. [PMID: 29467726 PMCID: PMC5808139 DOI: 10.3389/fmicb.2018.00081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/12/2018] [Indexed: 12/26/2022] Open
Abstract
Viruses must exploit the cellular biosynthetic machinery and evade cellular defense systems to complete their life cycles. Due to their crucial roles in cellular bioenergetics, apoptosis, innate immunity and redox balance, mitochondria are important functional targets of many viruses, including tumor viruses. The present review describes the interactions between mitochondria and proteins coded by the human tumor viruses human T-cell leukemia virus type 1, Epstein-Barr virus, Kaposi's sarcoma-associated herpesvirus, human hepatitis viruses B and C, and human papillomavirus, and highlights how these interactions contribute to viral replication, persistence and transformation.
Collapse
Affiliation(s)
| | - Gloria Scattolin
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padova, Italy
| | | | | | | | - Vincenzo Ciminale
- Veneto Institute of Oncology IOV-IRRCS, Padova, Italy.,Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
44
|
Carrasco-Avino G, Riquelme I, Padilla O, Villaseca M, Aguayo FR, Corvalan AH. The conundrum of the Epstein-Barr virus-associated gastric carcinoma in the Americas. Oncotarget 2017; 8:75687-75698. [PMID: 29088902 PMCID: PMC5650457 DOI: 10.18632/oncotarget.18497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/29/2017] [Indexed: 02/07/2023] Open
Abstract
Epstein-Barr virus-associated gastric carcinoma shows a higher prevalence in the Americas than Asia. We summarize all studies of Epstein Barr virus-associated gastric carcinoma in the Americas, focusing on host characteristics, environmental associations and phylogeographic diversity of Epstein-Barr virus strains. In the Americas, the prevalence of Epstein Barr virus-associated gastric carcinoma is 11.4%, more frequent in males and portray predominantly diffuse-type histology. EBERs, EBNAs, BARTs and LMP are the highest expressed genes; their variations in healthy individuals may explain the phylogeographic diversity of Epstein-Barr virus across the region. Gastric cancer cases harbor exclusively the western genotype (subtype D and kept Xho I site), suggesting a disrupted co-evolution between the pathogen and its host. Epstein-Barr virus-associated gastric carcinoma molecular subtype cases from The Cancer Genome Atlas display PIK3CA gene mutations, amplification of JAK2, PD-L1 and PD-L2 and CpG island methylator phenotype, leading to more extensive methylation of host and viral genomes than any other subtypes from the study. Environmental conditions include negative- and positive- associations with being firstborn child and smoking, respectively. A marginal association with H. pylori has also been reported. Lymphoepithelioma-like carcinoma is associated with Epstein Barr virus in 80%-86% of cases, most of which have been included as part of Epstein Barr virus-associated gastric carcinoma series (prevalence 1.1%-7.6%). Whether these cases represent a variant of Epstein-Barr virus-associated gastric carcinoma is discussed. We propose novel research strategies to solve the conundrum of the high prevalence of Epstein-Barr virus-associated gastric carcinoma in the Americas.
Collapse
Affiliation(s)
- Gonzalo Carrasco-Avino
- Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Catolica de Chile, Santiago, Chile
- Department of Pathology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ismael Riquelme
- Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de la Frontera, Temuco, Chile
- Department of Pathology, Universidad de la Frontera, Temuco, Chile
| | - Oslando Padilla
- Department of Public Health, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Miguel Villaseca
- Department of Pathology, Universidad de la Frontera, Temuco, Chile
| | - Francisco R. Aguayo
- Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Catolica de Chile, Santiago, Chile
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Alejandro H. Corvalan
- Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Catolica de Chile, Santiago, Chile
- UC-Center for Investigational Oncology (CITO), Pontificia Universidad Catolica de Chile, Santiago, Chile
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| |
Collapse
|
45
|
Small molecule perturbation of the CAND1-Cullin1-ubiquitin cycle stabilizes p53 and triggers Epstein-Barr virus reactivation. PLoS Pathog 2017; 13:e1006517. [PMID: 28715492 PMCID: PMC5531659 DOI: 10.1371/journal.ppat.1006517] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 07/27/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022] Open
Abstract
The chemical probe C60 efficiently triggers Epstein-Barr Virus (EBV) reactivation from latency through an unknown mechanism. Here, we identify the Cullin exchange factor CAND1 as a biochemical target of C60. We also identified CAND1 in an shRNA library screen for EBV lytic reactivation. Gene expression profiling revealed that C60 activates the p53 pathway and protein analysis revealed a strong stabilization and S15 phosphorylation of p53. C60 reduced Cullin1 association with CAND1 and led to a global accumulation of ubiquitylated substrates. C60 also stabilized the EBV immediate early protein ZTA through a Cullin-CAND1-interaction motif in the ZTA transcription activation domain. We propose that C60 perturbs the normal interaction and function of CAND1 with Cullins to promote the stabilization of substrates like ZTA and p53, leading to EBV reactivation from latency. Understanding the mechanism of action of C60 may provide new approaches for treatment of EBV associated tumors, as well as new tools to stabilize p53.
Collapse
|
46
|
Ersing I, Nobre L, Wang LW, Soday L, Ma Y, Paulo JA, Narita Y, Ashbaugh CW, Jiang C, Grayson NE, Kieff E, Gygi SP, Weekes MP, Gewurz BE. A Temporal Proteomic Map of Epstein-Barr Virus Lytic Replication in B Cells. Cell Rep 2017; 19:1479-1493. [PMID: 28514666 PMCID: PMC5446956 DOI: 10.1016/j.celrep.2017.04.062] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/24/2017] [Accepted: 04/20/2017] [Indexed: 01/10/2023] Open
Abstract
Epstein-Barr virus (EBV) replication contributes to multiple human diseases, including infectious mononucleosis, nasopharyngeal carcinoma, B cell lymphomas, and oral hairy leukoplakia. We performed systematic quantitative analyses of temporal changes in host and EBV proteins during lytic replication to gain insights into virus-host interactions, using conditional Burkitt lymphoma models of type I and II EBV infection. We quantified profiles of >8,000 cellular and 69 EBV proteins, including >500 plasma membrane proteins, providing temporal views of the lytic B cell proteome and EBV virome. Our approach revealed EBV-induced remodeling of cell cycle, innate and adaptive immune pathways, including upregulation of the complement cascade and proteasomal degradation of the B cell receptor complex, conserved between EBV types I and II. Cross-comparison with proteomic analyses of human cytomegalovirus infection and of a Kaposi-sarcoma-associated herpesvirus immunoevasin identified host factors targeted by multiple herpesviruses. Our results provide an important resource for studies of EBV replication.
Collapse
Affiliation(s)
- Ina Ersing
- Division of Infectious Disease, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA; Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Luis Nobre
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Liang Wei Wang
- Division of Infectious Disease, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA; Harvard Virology Program, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Lior Soday
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Yijie Ma
- Division of Infectious Disease, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Yohei Narita
- Division of Infectious Disease, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA; Department of Immunobiology and Microbiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Camille W Ashbaugh
- Division of Infectious Disease, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Chang Jiang
- Division of Infectious Disease, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | | | - Elliott Kieff
- Division of Infectious Disease, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA; Department of Immunobiology and Microbiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK.
| | - Benjamin E Gewurz
- Division of Infectious Disease, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA; Harvard Virology Program, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
47
|
Lupey-Green LN, Moquin SA, Martin KA, McDevitt SM, Hulse M, Caruso LB, Pomerantz RT, Miranda JL, Tempera I. PARP1 restricts Epstein Barr Virus lytic reactivation by binding the BZLF1 promoter. Virology 2017; 507:220-230. [PMID: 28456021 PMCID: PMC5521201 DOI: 10.1016/j.virol.2017.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/14/2017] [Accepted: 04/06/2017] [Indexed: 12/12/2022]
Abstract
The Epstein Barr virus (EBV) genome persists in infected host cells as a chromatinized episome and is subject to chromatin-mediated regulation. Binding of the host insulator protein CTCF to the EBV genome has an established role in maintaining viral latency type, and in other herpesviruses, loss of CTCF binding at specific regions correlates with viral reactivation. Here, we demonstrate that binding of PARP1, an important cofactor of CTCF, at the BZLF1 lytic switch promoter restricts EBV reactivation. Knockdown of PARP1 in the Akata-EBV cell line significantly increases viral copy number and lytic protein expression. Interestingly, CTCF knockdown has no effect on viral reactivation, and CTCF binding across the EBV genome is largely unchanged following reactivation. Moreover, EBV reactivation attenuates PARP activity, and Zta expression alone is sufficient to decrease PARP activity. Here we demonstrate a restrictive function of PARP1 in EBV lytic reactivation.
Collapse
Affiliation(s)
- Lena N Lupey-Green
- Fels Institute for Cancer Research & Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Stephanie A Moquin
- Gladstone Institute of Virology and Immunology, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Kayla A Martin
- Fels Institute for Cancer Research & Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Shane M McDevitt
- Fels Institute for Cancer Research & Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Michael Hulse
- Fels Institute for Cancer Research & Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Lisa B Caruso
- Fels Institute for Cancer Research & Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Richard T Pomerantz
- Fels Institute for Cancer Research & Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Jj L Miranda
- Gladstone Institute of Virology and Immunology, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Italo Tempera
- Fels Institute for Cancer Research & Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| |
Collapse
|
48
|
The SWI/SNF Chromatin Regulator BRG1 Modulates the Transcriptional Regulatory Activity of the Epstein-Barr Virus DNA Polymerase Processivity Factor BMRF1. J Virol 2017; 91:JVI.02114-16. [PMID: 28228591 DOI: 10.1128/jvi.02114-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/14/2017] [Indexed: 12/14/2022] Open
Abstract
During the lytic phase of Epstein-Barr virus (EBV), binding of the transactivator Zta to the origin of lytic replication (oriLyt) and the BHLF1 transcript, forming a stable RNA-DNA hybrid, is required to initiate viral DNA replication. EBV-encoded viral DNA replication proteins form complexes to amplify viral DNA. BMRF1, the viral DNA polymerase accessory factor, is essential for lytic DNA replication and also known as a transcriptional regulator of the expression of BHLF1 and BALF2 (single-stranded DNA [ssDNA]-binding protein). In order to determine systematically how BMRF1 regulates viral transcription, a BMRF1 knockout bacmid was generated to analyze viral gene expression using a viral DNA microarray. We found that a subset of Rta-responsive late genes, including BcLF1, BLLF1, BLLF2, and BDLF3, were downregulated in cells harboring a BMRF1 knockout EBV bacmid (p2089ΔBMRF1). In reporter assays, BMRF1 appears to transactivate a subset of viral late promoters through distinct pathways. BMRF1 activates the BDLF3 promoter in an SP1-dependent manner. Notably, BMRF1 associates with the transcriptional regulator BRG1 in EBV-reactivated cells. BMRF1-mediated transactivation activities on the BcLF1 and BLLF1 promoters were attenuated by knockdown of BRG1. In BRG1-depleted EBV-reactivated cells, BcLF1 and BLLF1 transcripts were reduced in number, resulting in reduced virion secretion. BMRF1 and BRG1 bound to the adjacent upstream regions of the BcLF1 and BLLF1 promoters, and depletion of BRG1 attenuated the recruitment of BMRF1 onto both promoters, suggesting that BRG1 is involved in BMRF1-mediated regulation of these two genes. Overall, we reveal a novel pathway by which BMRF1 can regulate viral promoters through interaction with BRG1.IMPORTANCE The cascade of viral gene expression during Epstein-Barr virus (EBV) replication is exquisitely regulated by the coordination of the viral DNA replication machinery and cellular factors. Upon lytic replication, the EBV immediate early proteins Zta and Rta turn on the expression of early proteins that assemble into viral DNA replication complexes. The DNA polymerase accessory factor, BMRF1, also is known to transactivate early gene expression through its interaction with SP1 or Zta on specific promoters. Through a global analysis, we demonstrate that BMRF1 also turns on a subset of Rta-regulated, late structural gene promoters. Searching for BMRF1-interacting cellular partners revealed that the SWI/SNF chromatin modifier BRG1 contributes to BMRF1-mediated transactivation of a subset of late promoters through protein-protein interaction and viral chromatin binding. Our findings indicate that BMRF1 regulates the expression of more viral genes than thought previously through distinct viral DNA replication-independent mechanisms.
Collapse
|
49
|
Farina A, Peruzzi G, Lacconi V, Lenna S, Quarta S, Rosato E, Vestri AR, York M, Dreyfus DH, Faggioni A, Morrone S, Trojanowska M, Farina GA. Epstein-Barr virus lytic infection promotes activation of Toll-like receptor 8 innate immune response in systemic sclerosis monocytes. Arthritis Res Ther 2017; 19:39. [PMID: 28245863 PMCID: PMC5331713 DOI: 10.1186/s13075-017-1237-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/16/2017] [Indexed: 01/15/2023] Open
Abstract
Background Monocytes/macrophages are activated in several autoimmune diseases, including systemic sclerosis (scleroderma; SSc), with increased expression of interferon (IFN)-regulatory genes and inflammatory cytokines, suggesting dysregulation of the innate immune response in autoimmunity. In this study, we investigated whether the lytic form of Epstein-Barr virus (EBV) infection (infectious EBV) is present in scleroderma monocytes and contributes to their activation in SSc. Methods Monocytes were isolated from peripheral blood mononuclear cells (PBMCs) depleted of the CD19+ cell fraction, using CD14/CD16 negative-depletion. Circulating monocytes from SSc and healthy donors (HDs) were infected with EBV. Gene expression of innate immune mediators were evaluated in EBV-infected monocytes from SSc and HDs. Involvement of Toll-like receptor (TLR)8 in viral-mediated TLR8 response was investigated by comparing the TLR8 expression induced by infectious EBV to the expression stimulated by CL075/TLR8/agonist-ligand in the presence of TLR8 inhibitor in THP-1 cells. Results Infectious EBV strongly induced TLR8 expression in infected SSc and HD monocytes in vitro. Markers of activated monocytes, such as IFN-regulated genes and chemokines, were upregulated in SSc- and HD-EBV-infected monocytes. Inhibiting TLR8 expression reduced virally induced TLR8 in THP-1 infected cells, demonstrating that innate immune activation by infectious EBV is partially dependent on TLR8. Viral mRNA and proteins were detected in freshly isolated SSc monocytes. Microarray analysis substantiated the evidence of an increased IFN signature and altered level of TLR8 expression in SSc monocytes carrying infectious EBV compared to HD monocytes. Conclusion This study provides the first evidence of infectious EBV in monocytes from patients with SSc and links EBV to the activation of TLR8 and IFN innate immune response in freshly isolated SSc monocytes. This study provides the first evidence of EBV replication activating the TLR8 molecular pathway in primary monocytes. Immunogenicity of infectious EBV suggests a novel mechanism mediating monocyte inflammation in SSc, by which EBV triggers the innate immune response in infected cells. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1237-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Antonella Farina
- Rheumatology, Boston University School of Medicine, Arthritis Center, 72 E. Concord Street, E-5, Boston, MA, 02118, USA.,Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | - Valentina Lacconi
- Rheumatology, Boston University School of Medicine, Arthritis Center, 72 E. Concord Street, E-5, Boston, MA, 02118, USA
| | - Stefania Lenna
- Rheumatology, Boston University School of Medicine, Arthritis Center, 72 E. Concord Street, E-5, Boston, MA, 02118, USA
| | - Silvia Quarta
- Department of Clinical Medicine, Sapienza University, Rome, Italy
| | - Edoardo Rosato
- Department of Clinical Medicine, Sapienza University, Rome, Italy
| | | | - Michael York
- Rheumatology, Boston University School of Medicine, Arthritis Center, 72 E. Concord Street, E-5, Boston, MA, 02118, USA
| | | | - Alberto Faggioni
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Stefania Morrone
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Maria Trojanowska
- Rheumatology, Boston University School of Medicine, Arthritis Center, 72 E. Concord Street, E-5, Boston, MA, 02118, USA
| | - G Alessandra Farina
- Rheumatology, Boston University School of Medicine, Arthritis Center, 72 E. Concord Street, E-5, Boston, MA, 02118, USA.
| |
Collapse
|
50
|
Chijioke O, Landtwing V, Münz C. NK Cell Influence on the Outcome of Primary Epstein-Barr Virus Infection. Front Immunol 2016; 7:323. [PMID: 27621731 PMCID: PMC5002423 DOI: 10.3389/fimmu.2016.00323] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/15/2016] [Indexed: 01/23/2023] Open
Abstract
The herpesvirus Epstein–Barr virus (EBV) was discovered as the first human candidate tumor virus in Burkitt’s lymphoma more than 50 years ago. Despite its strong growth transforming capacity, more than 90% of the human adult population carries this virus asymptomatically under near perfect immune control. The mode of primary EBV infection is in part responsible for EBV-associated diseases, including Hodgkin’s lymphoma. It is, therefore, important to understand which circumstances lead to symptomatic primary EBV infection, called infectious mononucleosis (IM). Innate immune control of lytic viral replication by early-differentiated natural killer (NK) cells was found to attenuate IM symptoms and continuous loss of the respective NK cell subset during the first decade of life might predispose for IM during adolescence. In this review, we discuss the evidence that NK cells are involved in the immune control of EBV, mechanisms by which they might detect and control lytic EBV replication, and compare NK cell subpopulations that expand during different human herpesvirus infections.
Collapse
Affiliation(s)
- Obinna Chijioke
- Institute of Surgical Pathology, University Hospital Zürich, Zürich, Switzerland; Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Vanessa Landtwing
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Zürich , Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Zürich , Switzerland
| |
Collapse
|