51
|
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
|
52
|
Praest P, Liaci AM, Förster F, Wiertz EJ. New insights into the structure of the MHC class I peptide-loading complex and mechanisms of TAP inhibition by viral immune evasion proteins. Mol Immunol 2019; 113:103-114. [DOI: 10.1016/j.molimm.2018.03.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/16/2018] [Accepted: 03/22/2018] [Indexed: 01/08/2023]
|
53
|
Li Q, Cohen JI. Epstein-Barr Virus and the Human Leukocyte Antigen Complex. CURRENT CLINICAL MICROBIOLOGY REPORTS 2019; 6:175-181. [PMID: 33094090 DOI: 10.1007/s40588-019-00120-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Purpose While most adults are infected Epstein-Barr virus (EBV), 3-5% remain uninfected. The human leukocyte antigen (HLA) complex, which controls many pathogens, may influence infection and disease associated with EBV. Recent Findings Numerous EBV proteins and miRNAs down-regulate HLA class I and II expression on the cell surface. HLA class II functions as a receptor for EBV entry into B cells. Specific HLA class II alleles correlate with the susceptibility of B cells to EBV infection in vitro and with EBV seropositivity or seronegativity of humans. HLA class I polymorphisms correlate with development and severity of EBV infectious mononucleosis and with the risk of several virus-associated malignancies including nasopharyngeal carcinoma, Hodgkin lymphoma, and post-transplant lymphoproliferative disease. Significance These findings indicate that while EBV has evolved to use MHC class II as a receptor for virus entry, polymorphisms in MHC class II and class I influence virus infection and disease.
Collapse
Affiliation(s)
- Qingxue Li
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Jeffrey I Cohen
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
54
|
Zhao MH, Sun L, Li P, Liu L, Luo B, Wang XF. Sequence analysis of Epstein–Barr virus (EBV) BNLF2a gene in malignant hematopathy of Northern China. Future Virol 2019. [DOI: 10.2217/fvl-2018-0129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BNLF2a is an early Epstein–Barr virus gene whose protein plays an immune escaping role by inhibiting the transporter associated with antigen processing. To explore the association between BNLF2a gene variations and EBV tumorigenesis, the BNLF2a gene of 259 EBV-positive samples (171 lymphohematopoietic disease samples and 88 throat washings from healthy donors) from northern China were sequenced. On the basis of phylogenetic tree and mutation characteristics of BNLF2a, all specimens were divided into two major genotypes: BNLF2a-A and BNLF2a-B. BNLF2a-A type, similar to the prototype B95-8, was the major subtype in all subpopulations. Healthy donors carried less BNLF2a-A and more BNLF2a-B than donors with lymphohematopoietic disease. The conservation of the BNLF2a gene may be crucial to its function.
Collapse
Affiliation(s)
- Meng-He Zhao
- Department of Pathogenic Biology, Qingdao University Medical College, 38 Dengzhou Road, Qingdao, 266021, PR China
| | - Lingling Sun
- Department of Pathology, Affiliated Hospital of Qingdao University Medical College, 59 Haier Road, Qingdao, 266003, PR China
| | - Ping Li
- Department of Blood Transfusion, Affiliated Hospital of Qingdao University Medical College, 16 Jiangsu Road, Qingdao, 266003, PR China
| | - Lei Liu
- Department of Laboratory, Qingdao commercial staff hospital, 6 Haipo Road, Qingdao, 266011, Shandong Province, China
| | - Bing Luo
- Department of Pathogenic Biology, Qingdao University Medical College, 38 Dengzhou Road, Qingdao, 266021, PR China
| | - Xiao-Feng Wang
- Department of Pathogenic Biology, Qingdao University Medical College, 38 Dengzhou Road, Qingdao, 266021, PR China
| |
Collapse
|
55
|
Frank T, Niemann I, Reichel A, Stamminger T. Emerging roles of cytomegalovirus-encoded G protein-coupled receptors during lytic and latent infection. Med Microbiol Immunol 2019; 208:447-456. [PMID: 30900091 DOI: 10.1007/s00430-019-00595-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/09/2019] [Indexed: 12/28/2022]
Abstract
Cytomegaloviruses (CMVs) have developed multiple diverse strategies to ensure their replicative success and to evade immune recognition. Given the fact that G protein-coupled receptors (GPCRs) are key regulators of numerous cellular processes and modify a variety of signaling pathways, it is not surprising that CMVs and other herpesviruses have hijacked mammalian GPCRs during their coevolution. Human cytomegalovirus (HCMV) encodes for four viral GPCR homologues (vGPCRs), termed US27, US28, UL33, and UL78. Although HCMV-encoded GPCRs were first described in 1990, the pivotal functions of these viral receptor proteins were detected only recently. Here, we summarize seminal knowledge on the functions of herpesviral vGPCRs with a focus on novel roles of cytomegalovirus-encoded vGPCRs for viral spread and the regulation of latency.
Collapse
Affiliation(s)
- Theresa Frank
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ina Niemann
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Reichel
- Institute for Virology, Ulm University Medical Center, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Thomas Stamminger
- Institute for Virology, Ulm University Medical Center, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
56
|
Abstract
The human persistent and oncogenic Epstein-Barr virus (EBV) was one of the first viruses that were described to express viral microRNAs (miRNAs). These have been proposed to modulate many host and viral functions, but their predominant role in vivo has remained unclear. We compared recombinant EBVs expressing or lacking miRNAs during in vivo infection of mice with reconstituted human immune system components and found that miRNA-deficient EBV replicates to lower viral titers with decreased frequencies of proliferating EBV-infected B cells. In response, activated cytotoxic EBV-specific T cells expand to lower frequencies than during infection with miRNA-expressing EBV. However, when we depleted CD8+ T cells the miRNA-deficient virus reached similar viral loads as wild-type EBV, increasing by more than 200-fold in the spleens of infected animals. Furthermore, CD8+ T cell depletion resulted in lymphoma formation in the majority of animals after miRNA-deficient EBV infection, while no tumors emerged when CD8+ T cells were present. Thus, miRNAs mainly serve the purpose of immune evasion from T cells in vivo and could become a therapeutic target to render EBV-associated malignancies more immunogenic.IMPORTANCE Epstein-Barr virus (EBV) infects the majority of the human population and usually persists asymptomatically within its host. Nevertheless, EBV is the causative agent for infectious mononucleosis (IM) and for lymphoproliferative disorders, including Burkitt and Hodgkin lymphomas. The immune system of the infected host is thought to prevent tumor formation in healthy virus carriers. EBV was one of the first viruses described to express miRNAs, and many host and viral targets were identified for these in vitro However, their role during EBV infection in vivo remained unclear. This work is the first to describe that EBV miRNAs mainly increase viremia and virus-associated lymphomas through dampening antigen recognition by adaptive immune responses in mice with reconstituted immune responses. Currently, there is no prophylactic or therapeutic treatment to restrict IM or EBV-associated malignancies; thus, targeting EBV miRNAs could promote immune responses and limit EBV-associated pathologies.
Collapse
|
57
|
Fares S, Spiess K, Olesen ETB, Zuo J, Jackson S, Kledal TN, Wills MR, Rosenkilde MM. Distinct Roles of Extracellular Domains in the Epstein-Barr Virus-Encoded BILF1 Receptor for Signaling and Major Histocompatibility Complex Class I Downregulation. mBio 2019; 10:e01707-18. [PMID: 30647152 PMCID: PMC6336419 DOI: 10.1128/mbio.01707-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/28/2018] [Indexed: 02/02/2023] Open
Abstract
The Epstein-Barr virus (EBV) BILF1 gene encodes a constitutively active G protein-coupled receptor (GPCR) that downregulates major histocompatibility complex (MHC) class I and induces signaling-dependent tumorigenesis. Different BILF1 homologs display highly conserved extracellular loops (ECLs) including the conserved cysteine residues involved in disulfide bridges present in class A GPCRs (GPCR bridge between transmembrane helix 3 [TM-3] and ECL-2) and in chemokine receptors (CKR bridge between the N terminus and ECL-3). In order to investigate the roles of the conserved residues in the receptor functions, 25 mutations were created in the extracellular domains. Luciferase reporter assays and flow cytometry were used to investigate the G protein signaling and MHC class I downregulation in HEK293 cells. We find that the cysteine residues involved in the GPCR bridge are important for both signaling and MHC class I downregulation, whereas the cysteine residues in the N terminus and ECL-3 are dispensable for signaling but important for MHC class I downregulation. Multiple conserved residues in the extracellular regions are important for the receptor-induced MHC class I downregulation, but not for signaling, indicating distinct structural requirements for these two functions. In an engineered receptor containing a binding site for Zn+2 ions in a complex with an aromatic chelator (phenanthroline or bipyridine), a ligand-driven inhibition of both the receptor signaling and MHC class I downregulation was observed. Taken together, this suggests that distinct regions in EBV-BILF1 can be pharmacologically targeted to inhibit the signaling-mediated tumorigenesis and interfere with the MHC class I downregulation.IMPORTANCE G protein-coupled receptors constitute the largest family of membrane proteins. As targets of >30% of the FDA-approved drugs, they are valuable for drug discovery. The receptor is composed of seven membrane-spanning helices and intracellular and extracellular domains. BILF1 is a receptor encoded by Epstein-Barr virus (EBV), which evades the host immune system by various strategies. BILF1 facilitates the virus immune evasion by downregulating MHC class I and is capable of inducing signaling-mediated tumorigenesis. BILF1 homologs from primate viruses show highly conserved extracellular domains. Here, we show that conserved residues in the extracellular domains of EBV-BILF1 are important for downregulating MHC class I and that the receptor signaling and immune evasion can be inhibited by drug-like small molecules. This suggests that BILF1 could be a target to inhibit the signaling-mediated tumorigenesis and interfere with the MHC class I downregulation, thereby facilitating virus recognition by the immune system.
Collapse
Affiliation(s)
- Suzan Fares
- Laboratory for Molecular and Translational Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katja Spiess
- Laboratory for Molecular and Translational Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emma T B Olesen
- Laboratory for Molecular and Translational Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sarah Jackson
- Division of Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Thomas N Kledal
- National Veterinary Institute, Technical University of Denmark, Lyngby, Denmark
| | - Mark R Wills
- Division of Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Mette M Rosenkilde
- Laboratory for Molecular and Translational Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
58
|
Davis DA, Shrestha P, Aisabor AI, Stream A, Galli V, Pise-Masison CA, Tagawa T, Ziegelbauer JM, Franchini G, Yarchoan R. Pomalidomide increases immune surface marker expression and immune recognition of oncovirus-infected cells. Oncoimmunology 2018; 8:e1546544. [PMID: 30713808 PMCID: PMC6343774 DOI: 10.1080/2162402x.2018.1546544] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/17/2018] [Accepted: 11/04/2018] [Indexed: 01/03/2023] Open
Abstract
Most chronic viruses evade T-cell and natural killer (NK) immunity through downregulation of immune surface markers. Previously we showed that Pomalidomide (Pom) increases surface expression of major histocompatibility complex class I (MHC-I) in Kaposi sarcoma-associated herpesvirus-infected latent and lytic cells and restores ICAM-1 and B7-2 in latent cells. We explored the ability of Pom to increase immune surface marker expression in cells infected by other chronic viruses, including human T-cell leukemia virus type-1 (HTLV-1), Epstein-Barr virus (EBV), human papilloma virus (HPV), Merkel cell polyoma virus (MCV), and human immunodeficiency virus type-1 (HIV-1). Pom increased MHC-1, ICAM-1, and B7-2/CD86 in immortalized T-cell lines productively infected with HTLV-1 and also significantly increased their susceptibility to NK cell-mediated cytotoxicity. Pom enhancement of MHC-I and ICAM-1 in primary cells infected with HTLV-1 was abrogated by knockout of HTLV-1 orf-1. Pom increased expression of ICAM-1, B7-2 and MHC class I polypeptide related sequence A (MICA) surface expression in the EBV-infected Daudi cells and increased their T-cell activation and susceptibility to NK cells. Moreover, Pom increased expression of certain of these surface markers on Akata, Raji, and EBV lymphoblastic cell lines. The increased expression of immune surface markers in these virus-infected lines was generally associated with a decrease in IRF4 expression. By contrast, Pom treatment of HPV, MCV and HIV-1 infected cells did not increase these immune surface markers. Pom and related drugs may be clinically beneficial for the treatment of HTLV-1 and EBV-induced tumors by rendering infected cells more susceptible to both innate and adaptive host immune responses.
Collapse
Affiliation(s)
- David A Davis
- HIV & AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Prabha Shrestha
- HIV & AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ashley I Aisabor
- HIV & AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Alexandra Stream
- HIV & AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Veronica Galli
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Cynthia A Pise-Masison
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Takanobu Tagawa
- HIV & AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Joseph M Ziegelbauer
- HIV & AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Genoveffa Franchini
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Robert Yarchoan
- HIV & AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
59
|
Cellular-based immunotherapy in Epstein-Barr virus induced nasopharyngeal cancer. Oral Oncol 2018; 84:61-70. [DOI: 10.1016/j.oraloncology.2018.07.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/06/2018] [Accepted: 07/18/2018] [Indexed: 12/27/2022]
|
60
|
Forrest C, Hislop AD, Rickinson AB, Zuo J. Proteome-wide analysis of CD8+ T cell responses to EBV reveals differences between primary and persistent infection. PLoS Pathog 2018; 14:e1007110. [PMID: 30248160 PMCID: PMC6171963 DOI: 10.1371/journal.ppat.1007110] [Citation(s) in RCA: 14] [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: 05/16/2018] [Revised: 10/04/2018] [Accepted: 08/27/2018] [Indexed: 02/07/2023] Open
Abstract
Human herpesviruses are antigenically rich agents that induce strong CD8+T cell responses in primary infection yet persist for life, continually challenging T cell memory through recurrent lytic replication and potentially influencing the spectrum of antigen-specific responses. Here we describe the first lytic proteome-wide analysis of CD8+ T cell responses to a gamma1-herpesvirus, Epstein-Barr virus (EBV), and the first such proteome-wide analysis of primary versus memory CD8+ T cell responses to any human herpesvirus. Primary effector preparations were generated directly from activated CD8+ T cells in the blood of infectious mononucleosis (IM) patients by in vitro mitogenic expansion. For memory preparations, EBV-specific cells in the blood of long-term virus carriers were first re-stimulated in vitro by autologous dendritic cells loaded with a lysate of lytically-infected cells, then expanded as for IM cells. Preparations from 7 donors of each type were screened against each of 70 EBV lytic cycle proteins in combination with the donor's individual HLA class I alleles. Multiple reactivities against immediate early (IE), early (E) and late (L) lytic cycle proteins, including many hitherto unrecognised targets, were detected in both contexts. Interestingly however, the two donor cohorts showed a different balance between IE, E and L reactivities. Primary responses targeted IE and a small group of E proteins preferentially, seemingly in line with their better presentation on the infected cell surface before later-expressed viral evasins take full hold. By contrast, target choice equilibrates in virus carriage with responses to key IE and E antigens still present but with responses to a select subset of L proteins now often prominent. We infer that, for EBV at least, long-term virus carriage with its low level virus replication and lytic antigen release is associated with a re-shaping of the virus-specific response.
Collapse
Affiliation(s)
- Calum Forrest
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Andrew D. Hislop
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Alan B. Rickinson
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jianmin Zuo
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
61
|
Pontejo SM, Murphy PM, Pease JE. Chemokine Subversion by Human Herpesviruses. J Innate Immun 2018; 10:465-478. [PMID: 30165356 DOI: 10.1159/000492161] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/11/2018] [Indexed: 12/30/2022] Open
Abstract
Viruses use diverse molecular mechanisms to exploit and evade the immune response. Herpesviruses, in particular, encode functional chemokine and chemokine receptor homologs pirated from the host, as well as secreted chemokine-binding proteins with unique structures. Multiple functions have been described for herpesvirus chemokine components, including attraction of target cells, blockade of leukocyte migration, and modulation of gene expression and cell entry by the virus. Here we review current concepts about how human herpesvirus chemokines, chemokine receptors, and chemokine-binding proteins may be used to shape a proviral state in the host.
Collapse
Affiliation(s)
- Sergio M Pontejo
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - James E Pease
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United
| |
Collapse
|
62
|
Li CW, Jheng BR, Chen BS. Investigating genetic-and-epigenetic networks, and the cellular mechanisms occurring in Epstein-Barr virus-infected human B lymphocytes via big data mining and genome-wide two-sided NGS data identification. PLoS One 2018; 13:e0202537. [PMID: 30133498 PMCID: PMC6105016 DOI: 10.1371/journal.pone.0202537] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022] Open
Abstract
Epstein-Barr virus (EBV), also known as human herpesvirus 4, is prevalent in all human populations. EBV mainly infects human B lymphocytes and epithelial cells, and is therefore associated with their various malignancies. To unravel the cellular mechanisms during the infection, we constructed interspecies networks to investigate the molecular cross-talk mechanisms between human B cells and EBV at the first (0-24 hours) and second (8-72 hours) stages of EBV infection. We first constructed a candidate genome-wide interspecies genetic-and-epigenetic network (the candidate GIGEN) by big database mining. We then pruned false positives in the candidate GIGEN to obtain the real GIGENs at the first and second infection stages in the lytic phase by their corresponding next-generation sequencing data through dynamic interaction models, the system identification approach, and the system order detection method. The real GIGENs are very complex and comprise protein-protein interaction networks, gene/microRNA (miRNA)/long non-coding RNA regulation networks, and host-virus cross-talk networks. To understand the molecular cross-talk mechanisms underlying EBV infection, we extracted the core GIGENs including host-virus core networks and host-virus core pathways from the real GIGENs using the principal network projection method. According to the results, we found that the activities of epigenetics-associated human proteins or genes were initially inhibited by viral proteins and miRNAs, and human immune responses were then dysregulated by epigenetic modification. We suggested that EBV exploits viral proteins and miRNAs, such as EBNA1, BPLF1, BALF3, BVRF1 and miR-BART14, to develop its defensive mechanism to defeat multiple immune attacks by the human immune system, promotes virion production, and facilitates the transportation of viral particles by activating the human genes NRP1 and CLIC5. Ultimately, we propose a therapeutic intervention comprising thymoquinone, valpromide, and zebularine to act as inhibitors of EBV-associated malignancies.
Collapse
Affiliation(s)
- Cheng-Wei Li
- Laboratory of Control and Systems Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Bo-Ren Jheng
- Laboratory of Control and Systems Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Bor-Sen Chen
- Laboratory of Control and Systems Biology, National Tsing Hua University, Hsinchu, Taiwan
- * E-mail:
| |
Collapse
|
63
|
Hanson DJ, Hill JA, Koelle DM. Advances in the Characterization of the T-Cell Response to Human Herpesvirus-6. Front Immunol 2018; 9:1454. [PMID: 29988505 PMCID: PMC6026635 DOI: 10.3389/fimmu.2018.01454] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/12/2018] [Indexed: 12/29/2022] Open
Abstract
Human herpesvirus (HHV) 6 is thought to remain clinically latent in most individuals after primary infection and to reactivate to cause disease in persons with severe immunosuppression. In allogeneic hematopoietic stem cell transplant recipients, reactivation of HHV-6 species B is a considerable cause of morbidity and mortality. HHV-6B reactivation is the most frequent cause of infectious meningoencephalitis in this setting and has been associated with a variety of other complications such as graft rejection and acute graft versus host disease. This has inspired efforts to develop HHV-6-targeted immunotherapies. Basic knowledge of HHV-6-specific adaptive immunity is crucial for these endeavors, but remains incomplete. Many studies have focused on specific HHV-6 antigens extrapolated from research on human cytomegalovirus, a genetically related betaherpesvirus. Challenges to the study of HHV-6-specific T-cell immunity include the very low frequency of HHV-6-specific memory T cells in chronically infected humans, the large genome size of HHV-6, and the lack of an animal model. This review will focus on emerging techniques and methodological improvements that are beginning to overcome these barriers. Population-prevalent antigens are now becoming clear for the CD4+ T-cell response, while definition and ranking of CD8+ T-cell antigens and epitopes is at an earlier stage. This review will discuss current knowledge of the T-cell response to HHV-6, new research approaches, and translation to clinical practice.
Collapse
Affiliation(s)
- Derek J Hanson
- Department of Medicine, University of Washington, Seattle, WA, United States.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Joshua A Hill
- Department of Medicine, University of Washington, Seattle, WA, United States.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - David M Koelle
- Department of Medicine, University of Washington, Seattle, WA, United States.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Department of Laboratory Medicine, University of Washington, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States.,Benaroya Research Institute, Seattle, WA, United States
| |
Collapse
|
64
|
Encyclopedia of EBV-Encoded Lytic Genes: An Update. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1045:395-412. [DOI: 10.1007/978-981-10-7230-7_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
65
|
The Immunomodulatory Capacity of an Epstein-Barr Virus Abortive Lytic Cycle: Potential Contribution to Viral Tumorigenesis. Cancers (Basel) 2018; 10:cancers10040098. [PMID: 29601503 PMCID: PMC5923353 DOI: 10.3390/cancers10040098] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 12/11/2022] Open
Abstract
Epstein-Barr virus (EBV) is characterized by a bipartite life cycle in which latent and lytic stages are alternated. Latency is compatible with long-lasting persistency within the infected host, while lytic expression, preferentially found in oropharyngeal epithelial tissue, is thought to favor host-to-host viral dissemination. The clinical importance of EBV relates to its association with cancer, which we think is mainly a consequence of the latency/persistency mechanisms. However, studies in murine models of tumorigenesis/lymphomagenesis indicate that the lytic cycle also contributes to cancer formation. Indeed, EBV lytic expression is often observed in established cell lines and tumor biopsies. Within the lytic cycle EBV expresses a handful of immunomodulatory (BCRF1, BARF1, BNLF2A, BGLF5 & BILF1) and anti-apoptotic (BHRF1 & BALF1) proteins. In this review, we discuss the evidence supporting an abortive lytic cycle in which these lytic genes are expressed, and how the immunomodulatory mechanisms of EBV and related herpesviruses Kaposi Sarcoma herpesvirus (KSHV) and human cytomegalovirus (HCMV) result in paracrine signals that feed tumor cells. An abortive lytic cycle would reconcile the need of lytic expression for viral tumorigenesis without relaying in a complete cycle that would induce cell lysis to release the newly formed infective viral particles.
Collapse
|
66
|
Almohammed R, Osborn K, Ramasubramanyan S, Perez-Fernandez IBN, Godfrey A, Mancini EJ, Sinclair AJ. Mechanism of activation of the BNLF2a immune evasion gene of Epstein-Barr virus by Zta. J Gen Virol 2018; 99:805-817. [PMID: 29580369 PMCID: PMC6096924 DOI: 10.1099/jgv.0.001056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The human gamma herpes virus Epstein–Barr virus (EBV) exploits multiple routes to evade the cellular immune response. During the EBV lytic replication cycle, viral proteins are expressed that provide excellent targets for recognition by cytotoxic T cells. This is countered by the viral BNLF2a gene. In B cells during latency, where BNLF2a is not expressed, we show that its regulatory region is embedded in repressive chromatin. The expression of BNLF2a mirrors the expression of a viral lytic cycle transcriptional regulator, Zta (BZLF1, EB1, ZEBRA), in B cells and we propose that Zta plays a role in up-regulating BNLF2a. In cells undergoing EBV lytic replication, we identified two distinct regions of interaction of Zta with the chromatin-associated BNLF2a promoter. We identify five potential Zta-response elements (ZREs) in the promoter that are highly conserved between virus isolates. Zta binds to these elements in vitro and activates the expression of the BNLF2a promoter in both epithelial and B cells. We also found redundancy amongst the ZREs. The EBV genome undergoes a biphasic DNA methylation cycle during its infection cycle. One of the ZREs contains an integral CpG motif. We show that this can be DNA methylated during EBV latency and that both Zta binding and promoter activation are enhanced by its methylation. In summary, we find that the BNLF2a promoter is directly targeted by Zta and that DNA methylation within the proximal ZRE aids activation. The implications for regulation of this key viral gene during the reactivation of EBV from latency are discussed.
Collapse
Affiliation(s)
- Rajaei Almohammed
- School of Life Sciences, University of Sussex, Brighton, East Sussex, UK.,Present address: Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Kay Osborn
- School of Life Sciences, University of Sussex, Brighton, East Sussex, UK
| | - Sharada Ramasubramanyan
- School of Life Sciences, University of Sussex, Brighton, East Sussex, UK.,Present address: RS Mehta Jain Department of Biochemistry and Cell Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | | | - Anja Godfrey
- School of Life Sciences, University of Sussex, Brighton, East Sussex, UK
| | - Erika J Mancini
- School of Life Sciences, University of Sussex, Brighton, East Sussex, UK
| | - Alison J Sinclair
- School of Life Sciences, University of Sussex, Brighton, East Sussex, UK
| |
Collapse
|
67
|
The Immune Response to Epstein Barr Virus and Implications for Posttransplant Lymphoproliferative Disorder. Transplantation 2017; 101:2009-2016. [PMID: 28376031 DOI: 10.1097/tp.0000000000001767] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Posttransplant lymphoproliferative disorder (PTLD) is a serious complication in organ transplant recipients and is most often associated with the Epstein Barr virus (EBV). EBV is a common gammaherpes virus with tropism for B lymphocytes and infection in immunocompetent individuals is typically asymptomatic and benign. However, infection in immunocompromised or immunosuppressed individuals can result in malignant B cell lymphoproliferations, such as PTLD. EBV+ PTLD can arise after primary EBV infection, or because of reactivation of a prior infection, and represents a leading malignancy in the transplant population. The incidence of EBV+ PTLD is variable depending on the organ transplanted and whether the recipient has preexisting immunity to EBV but can be as high as 20%. It is generally accepted that impaired immune function due to immunosuppression is a primary cause of EBV+ PTLD. In this overview, we review the EBV life cycle and discuss our current understanding of the immune response to EBV in healthy, immunocompetent individuals, in transplant recipients, and in PTLD patients. We review the strategies that EBV uses to subvert and evade host immunity and discuss the implications for the development of EBV+ PTLD.
Collapse
|
68
|
Characterization of the subcellular localization of Epstein-Barr virus encoded proteins in live cells. Oncotarget 2017; 8:70006-70034. [PMID: 29050259 PMCID: PMC5642534 DOI: 10.18632/oncotarget.19549] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/29/2017] [Indexed: 12/27/2022] Open
Abstract
Epstein-Barr virus (EBV) is the pathogenic factor of numerous human tumors, yet certain of its encoded proteins have not been studied. As a first step for functional identification, we presented the construction of a library of expression constructs for most of the EBV encoded proteins and an explicit subcellular localization map of 81 proteins encoded by EBV in mammalian cells. Viral open reading frames were fused with enhanced yellow fluorescent protein (EYFP) tag in eukaryotic expression plasmid then expressed in COS-7 live cells, and protein localizations were observed by fluorescence microscopy. As results, 34.57% (28 proteins) of all proteins showed pan-nuclear or subnuclear localization, 39.51% (32 proteins) exhibitted pan-cytoplasmic or subcytoplasmic localization, and 25.93% (21 proteins) were found in both the nucleus and cytoplasm. Interestingly, most envelope proteins presented pan-cytoplasmic or membranous localization, and most capsid proteins displayed enriched or complete localization in the nucleus, indicating that the subcellular localization of specific proteins are associated with their roles during viral replication. Taken together, the subcellular localization map of EBV proteins in live cells may lay the foundation for further illustrating the functions of EBV-encoded genes in human diseases especially in its relevant tumors.
Collapse
|
69
|
Hu J, Li H, Luo X, Li Y, Bode A, Cao Y. The role of oxidative stress in EBV lytic reactivation, radioresistance and the potential preventive and therapeutic implications. Int J Cancer 2017; 141:1722-1729. [PMID: 28571118 DOI: 10.1002/ijc.30816] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/26/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Jianmin Hu
- Key Laboratory of Cancer Carcinogenesis and Invasion, Chinese Ministry of Education; Xiangya Hospital, Central South University; Changsha China
- Cancer Research Institute, Xiangya School of Medicine, Central South University; Changsha China
- Key Laboratory of Carcinogenesis; Chinese Ministry of Health; Changsha China
| | - Hongde Li
- Key Laboratory of Cancer Carcinogenesis and Invasion, Chinese Ministry of Education; Xiangya Hospital, Central South University; Changsha China
- Cancer Research Institute, Xiangya School of Medicine, Central South University; Changsha China
- Key Laboratory of Carcinogenesis; Chinese Ministry of Health; Changsha China
| | - Xiangjian Luo
- Key Laboratory of Cancer Carcinogenesis and Invasion, Chinese Ministry of Education; Xiangya Hospital, Central South University; Changsha China
- Cancer Research Institute, Xiangya School of Medicine, Central South University; Changsha China
- Key Laboratory of Carcinogenesis; Chinese Ministry of Health; Changsha China
| | - Yueshuo Li
- Key Laboratory of Cancer Carcinogenesis and Invasion, Chinese Ministry of Education; Xiangya Hospital, Central South University; Changsha China
- Cancer Research Institute, Xiangya School of Medicine, Central South University; Changsha China
- Key Laboratory of Carcinogenesis; Chinese Ministry of Health; Changsha China
| | - Ann Bode
- The Hormel Institute, University of Minnesota; Austin MN 55912
| | - Ya Cao
- Key Laboratory of Cancer Carcinogenesis and Invasion, Chinese Ministry of Education; Xiangya Hospital, Central South University; Changsha China
- Cancer Research Institute, Xiangya School of Medicine, Central South University; Changsha China
- Key Laboratory of Carcinogenesis; Chinese Ministry of Health; Changsha China
| |
Collapse
|
70
|
EBV based cancer prevention and therapy in nasopharyngeal carcinoma. NPJ Precis Oncol 2017; 1:10. [PMID: 29872698 PMCID: PMC5871899 DOI: 10.1038/s41698-017-0018-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 12/13/2022] Open
Abstract
Epstein-Barr virus is an important cancer causing virus. Nasopharyngeal carcinoma is an infection-related cancer strongly driven by Epstein-Barr virus. In this cancer model, we identified the major host targets of latent membrane protein 1 which is a driving oncogene encoded by Epstein-Barr virus in latency infection. latent membrane protein 1 activates several oncogenic signaling axes causing multiple malignant phenotypes and therapeutic resistance. Also, Epstein-Barr virus up-regulates DNA methyltransferase 1 and mediates onco-epigenetic effects in the carcinogenesis. The collaborating pathways activated by latent membrane protein 1 constructs an oncogenic signaling network, which makes latent membrane protein 1 an important potential target for effective treatment or preventive intervention. In Epstein-Barr virus lytic phase, the plasma level of Epstein-Barr virus DNA is considered as a distinguishing marker for nasopharyngeal carcinoma in subjects from healthy high-risk populations and is also a novel prognostic marker in Epstein-Barr virus-positive nasopharyngeal carcinoma. Now the early detection and screening of the lytic proteins and Epstein-Barr virus DNA have been applied to clinical and high-risk population. The knowledge generated regarding Epstein-Barr virus can be used in Epstein-Barr virus based precision cancer prevention and therapy in the near future.
Collapse
|
71
|
Achinko D, Dormer A, Narayanan M, Norman E, Abbas M. Regulatory patterns of differentially expressed genes in Ebola and related viruses are critical for viral screening and diagnosis. F1000Res 2017. [DOI: 10.12688/f1000research.10597.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background Viral detection techniques and applications are a critical first step to pathogen detection within a given population, especially during outbreaks. Common viral tests currently used are direct specimen examination, indirect examination and serological tests. Serological tests have gained intense interest because they are rapidly performed with patient blood samples for quick diagnosis and treatment. The diagnostic techniques developed around serology are often expensive, require expertise to use and cannot be afforded by developing countries with recurrent viral outbreaks. Therefore exploiting the huge amount of viral data available in various databases is critical to develop affordable and easy-to-use diagnostic tools. Methods This study obtained viral sample data from Gene Expression Omnibus database with focus on use of viral glycoprotein for host penetration. Gene relative mean across 34 obtained viral samples were extracted into data tables and used with edgeR statistical software in R version 3.3.1. Results Three clusters previously known to be LCK specific (Ebola virus relative viral cluster, EBOVC), CD209 specific (Mean differentiation cluster, MDC) and both LCK and CD209 specific (Kurtosis group cluster, KGC), expressed unique patterns of four proteins of interest (CD209, LCK, IL-2 and MYB). Differential expression analysis showed two cluster patterns on heatmaps, with differentially expressed proteins down-regulated in MDC but up-regulated in KGC and EBOVC for all pairwise cluster comparative analyses performed. Heatmaps showed two distinct immune related patterns, identifying MDC as B-lymphotropic while KGC and EBOVC as T-lymphotropic. Identified pathways were dominantly involved with homeostasis of immune cells and viral cell surface receptors involved in protein kinase activities. Conclusions Regulatory proteomic variants identified in clusters suggest transcription repression of HLA class I alleles. This study identified viral expression patterns with screening and therapeutic applications. Given that the viral pathogenetic pathway for Ebola has not been clearly identified yet, assembling its components is vital for vaccine development.
Collapse
|
72
|
Novel Chemokine-Based Immunotoxins for Potent and Selective Targeting of Cytomegalovirus Infected Cells. J Immunol Res 2017; 2017:4069260. [PMID: 28251165 PMCID: PMC5303859 DOI: 10.1155/2017/4069260] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/27/2016] [Indexed: 11/17/2022] Open
Abstract
Immunotoxins as antiviral therapeutics are largely unexplored but have promising prospective due to their high selectivity potential and their unparalleled efficiency. One recent example targeted the virus-encoded G protein-coupled receptor US28 as a strategy for specific and efficient treatment of human cytomegalovirus (HCMV) infections. US28 is expressed on virus-infected cells and scavenge chemokines by rapid internalization. The chemokine-based fusion-toxin protein (FTP) consisted of a variant (F49A) of CX3CL1 specifically targeting US28 linked to the catalytic domain of Pseudomonas exotoxin A (PE). Here, we systematically seek to improve F49A-FTP by modifications in its three structural domains; we generated variants with (1) altered chemokine sequence (K14A, F49L, and F49E), (2) shortened and elongated linker region, and (3) modified toxin domain. Only F49L-FTP displayed higher selectivity in its binding to US28 versus CX3CR1, the endogenous receptor for CX3CL1, but this was not matched by a more selective killing of US28-expressing cells. A longer linker and different toxin variants decreased US28 affinity and selective killing. Thereby, F49A-FTP represents the best candidate for HCMV treatment. Many viruses encode internalizing receptors suggesting that not only HCMV but also, for instance, Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus may be targeted by FTPs.
Collapse
|
73
|
van der Werff ten Bosch J, van den Akker M. Genetic predisposition and hematopoietic malignancies in children: Primary immunodeficiency. Eur J Med Genet 2016; 59:647-653. [DOI: 10.1016/j.ejmg.2016.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 02/18/2016] [Accepted: 03/08/2016] [Indexed: 01/24/2023]
|
74
|
Abstract
It is more than 50 years since the Epstein-Barr virus (EBV), the first human tumour virus, was discovered. EBV has subsequently been found to be associated with a diverse range of tumours of both lymphoid and epithelial origin. Progress in the molecular analysis of EBV has revealed fundamental mechanisms of more general relevance to the oncogenic process. This Timeline article highlights key milestones in the 50-year history of EBV and discusses how this virus provides a paradigm for exploiting insights at the molecular level in the diagnosis, treatment and prevention of cancer.
Collapse
Affiliation(s)
- Lawrence S Young
- Warwick Medical School, The University of Warwick, Coventry CV4 7AL, UK
| | - Lee Fah Yap
- Department of Oral and Craniofacial Sciences and Oral Cancer Research Coordinating Centre, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Paul G Murray
- Institute of Cancer and Genomic Medicine, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
75
|
Kwon EK, Min CK, Kim Y, Lee JW, Aigerim A, Schmidt S, Nam HJ, Han SK, Kim K, Cha JS, Kim H, Kim S, Cho HS, Choi MS, Cho NH. Constitutive activation of T cells by γ2-herpesviral GPCR through the interaction with cellular CXCR4. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:1-11. [PMID: 27751885 DOI: 10.1016/j.bbamcr.2016.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/14/2016] [Accepted: 10/11/2016] [Indexed: 12/28/2022]
Abstract
Members of the herpesviral family use multiple strategies to hijack infected host cells and exploit cellular signaling for their pathogenesis and latent infection. Among the most intriguing weapons in the arsenal of pathogenic herpesviruses are the constitutively active virally-encoded G protein-coupled receptors (vGPCRs). Even though vGPCRs contribute to viral pathogenesis such as immune evasion and proliferative disorders, the molecular details of how vGPCRs continuously activate cellular signaling are largely unknown. Here, we report that the vGPCR of Herpesvirus saimiri (HVS), an oncogenic γ2-herpesvirus, constitutively activates T cells via a heteromeric interaction with cellular CXCR4. Constitutive T cell activation also occurs with expression of the vGPCR of Kaposi's sarcoma-associated herpesvirus (KSHV), but not the vGPCR of Epstein-Barr virus. Expression of HVS vGPCR down-regulated the surface expression of CXCR4 but did not induce the degradation of the chemokine receptor, suggesting that vGPCR/CXCR4 signaling continues in cytosolic compartments. The physical association of vGPCR with CXCR4 was demonstrated by proximity ligation assay as well as immunoprecipitation. Interestingly, the constitutive activation of T cells by HVS vGPCR is independent of proximal T cell receptor (TCR) signaling molecules, such as TCRβ, Lck, and ZAP70, whereas CXCR4 silencing by shRNA abolished T cell activation by vGPCRs of HVS and KSHV. Furthermore, previously identified inactive vGPCR mutants failed to interact with CXCR4. These findings on the positive cooperativity of vGPCR with cellular CXCR4 in T cell activation extend our current understanding of the molecular mechanisms of vGPCR function and highlight the importance of heteromerization for GPCR activity.
Collapse
Affiliation(s)
- Eun-Kyung Kwon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Chan-Ki Min
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Yuri Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jae-Won Lee
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Abdimadiyeva Aigerim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sebastian Schmidt
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hyun-Jun Nam
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Seong Kyu Han
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Kuglae Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jeong Seok Cha
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Hoyoung Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Sanguk Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Hyun-Soo Cho
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Myung-Sik Choi
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Endemic Disease, Seoul National University Medical Research Center and Bundang Hospital, Seoul 03080, Republic of Korea.
| |
Collapse
|
76
|
Epstein-Barr virus microRNAs reduce immune surveillance by virus-specific CD8+ T cells. Proc Natl Acad Sci U S A 2016; 113:E6467-E6475. [PMID: 27698133 DOI: 10.1073/pnas.1605884113] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Infection with Epstein-Barr virus (EBV) affects most humans worldwide and persists life-long in the presence of robust virus-specific T-cell responses. In both immunocompromised and some immunocompetent people, EBV causes several cancers and lymphoproliferative diseases. EBV transforms B cells in vitro and encodes at least 44 microRNAs (miRNAs), most of which are expressed in EBV-transformed B cells, but their functions are largely unknown. Recently, we showed that EBV miRNAs inhibit CD4+ T-cell responses to infected B cells by targeting IL-12, MHC class II, and lysosomal proteases. Here we investigated whether EBV miRNAs also counteract surveillance by CD8+ T cells. We have found that EBV miRNAs strongly inhibit recognition and killing of infected B cells by EBV-specific CD8+ T cells through multiple mechanisms. EBV miRNAs directly target the peptide transporter subunit TAP2 and reduce levels of the TAP1 subunit, MHC class I molecules, and EBNA1, a protein expressed in most forms of EBV latency and a target of EBV-specific CD8+ T cells. Moreover, miRNA-mediated down-regulation of the cytokine IL-12 decreases the recognition of infected cells by EBV-specific CD8+ T cells. Thus, EBV miRNAs use multiple, distinct pathways, allowing the virus to evade surveillance not only by CD4+ but also by antiviral CD8+ T cells.
Collapse
|
77
|
Tagawa T, Albanese M, Bouvet M, Moosmann A, Mautner J, Heissmeyer V, Zielinski C, Lutter D, Hoser J, Hastreiter M, Hayes M, Sugden B, Hammerschmidt W. Epstein-Barr viral miRNAs inhibit antiviral CD4+ T cell responses targeting IL-12 and peptide processing. J Exp Med 2016; 213:2065-80. [PMID: 27621419 PMCID: PMC5030804 DOI: 10.1084/jem.20160248] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 08/01/2016] [Indexed: 12/14/2022] Open
Abstract
EBV reduces the activation of cytotoxic CD4+ effector T cells by inducing a state of reduced immunogenicity in infected B cells. EBV-derived miRNAs suppress release of proinflammatory cytokines, interfere with peptide processing and presentation on HLA class II, repress differentiation of naive CD4+ T cells to Th1 cells, and ultimately avoid killing of infected B cells. Epstein-Barr virus (EBV) is a tumor virus that establishes lifelong infection in most of humanity, despite eliciting strong and stable virus-specific immune responses. EBV encodes at least 44 miRNAs, most of them with unknown function. Here, we show that multiple EBV miRNAs modulate immune recognition of recently infected primary B cells, EBV's natural target cells. EBV miRNAs collectively and specifically suppress release of proinflammatory cytokines such as IL-12, repress differentiation of naive CD4+ T cells to Th1 cells, interfere with peptide processing and presentation on HLA class II, and thus reduce activation of cytotoxic EBV-specific CD4+ effector T cells and killing of infected B cells. Our findings identify a previously unknown viral strategy of immune evasion. By rapidly expressing multiple miRNAs, which are themselves nonimmunogenic, EBV counteracts recognition by CD4+ T cells and establishes a program of reduced immunogenicity in recently infected B cells, allowing the virus to express viral proteins required for establishment of life-long infection.
Collapse
Affiliation(s)
- Takanobu Tagawa
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Partner site Munich, Germany, D-81377 Munich, Germany German Centre for Infection Research (DZIF), Partner site Munich, Germany, D-81377 Munich, Germany
| | - Manuel Albanese
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Partner site Munich, Germany, D-81377 Munich, Germany German Centre for Infection Research (DZIF), Partner site Munich, Germany, D-81377 Munich, Germany
| | - Mickaël Bouvet
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Partner site Munich, Germany, D-81377 Munich, Germany German Centre for Infection Research (DZIF), Partner site Munich, Germany, D-81377 Munich, Germany
| | - Andreas Moosmann
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Partner site Munich, Germany, D-81377 Munich, Germany German Centre for Infection Research (DZIF), Partner site Munich, Germany, D-81377 Munich, Germany
| | - Josef Mautner
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Partner site Munich, Germany, D-81377 Munich, Germany German Centre for Infection Research (DZIF), Partner site Munich, Germany, D-81377 Munich, Germany Children's Hospital, Technical University Munich, D-80337 Munich, Germany
| | - Vigo Heissmeyer
- Research Unit Molecular Immune Regulation, Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health Munich, University of Munich, D-80539 Munich, Germany Institute for Immunology, University of Munich, D-80539 Munich, Germany
| | - Christina Zielinski
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, D-80337 Munich, Germany
| | - Dominik Lutter
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Munich, Germany
| | - Jonathan Hoser
- Institute of Bioinformatics and System Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Munich, Germany
| | - Maximilian Hastreiter
- Institute of Bioinformatics and System Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Munich, Germany
| | - Mitch Hayes
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Bill Sugden
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Wolfgang Hammerschmidt
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Partner site Munich, Germany, D-81377 Munich, Germany German Centre for Infection Research (DZIF), Partner site Munich, Germany, D-81377 Munich, Germany
| |
Collapse
|
78
|
Wei F, Zhu Q, Ding L, Liang Q, Cai Q. Manipulation of the host cell membrane by human γ-herpesviruses EBV and KSHV for pathogenesis. Virol Sin 2016; 31:395-405. [PMID: 27624182 DOI: 10.1007/s12250-016-3817-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/29/2016] [Indexed: 11/27/2022] Open
Abstract
The cell membrane regulates many physiological processes including cellular communication, homing and metabolism. It is therefore not surprising that the composition of the host cell membrane is manipulated by intracellular pathogens. Among these, the human oncogenic herpesviruses Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) exploit the host cell membrane to avoid immune surveillance and promote viral replication. Accumulating evidence has shown that both EBV and KSHV directly encode several similar membrane-associated proteins, including receptors and receptor-specific ligands (cytokines and chemokines), to increase virus fitness in spite of host antiviral immune responses. These proteins are expressed individually at different phases of the EBV/KSHV life cycle and employ various mechanisms to manipulate the host cell membrane. In recent decades, much effort has been made to address how these membrane-based signals contribute to viral tumorigenesis. In this review, we summarize and highlight the recent understanding of how EBV and KSHV similarly manipulate host cell membrane signals, particularly how remodeling of the cell membrane allows EBV and KSHV to avoid host antiviral immune responses and favors their latent and lytic infection.
Collapse
Affiliation(s)
- Fang Wei
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qing Zhu
- Key Laboratory of Medical Molecular Virology (Ministries of Education and Health), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Ling Ding
- Key Laboratory of Medical Molecular Virology (Ministries of Education and Health), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qing Liang
- Key Laboratory of Medical Molecular Virology (Ministries of Education and Health), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qiliang Cai
- Key Laboratory of Medical Molecular Virology (Ministries of Education and Health), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
79
|
Gram AM, Oosenbrug T, Lindenbergh MFS, Büll C, Comvalius A, Dickson KJI, Wiegant J, Vrolijk H, Lebbink RJ, Wolterbeek R, Adema GJ, Griffioen M, Heemskerk MHM, Tscharke DC, Hutt-Fletcher LM, Wiertz EJHJ, Hoeben RC, Ressing ME. The Epstein-Barr Virus Glycoprotein gp150 Forms an Immune-Evasive Glycan Shield at the Surface of Infected Cells. PLoS Pathog 2016; 12:e1005550. [PMID: 27077376 PMCID: PMC4831753 DOI: 10.1371/journal.ppat.1005550] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 03/14/2016] [Indexed: 12/15/2022] Open
Abstract
Cell-mediated immunity plays a key role in host control of viral infection. This is exemplified by life-threatening reactivations of e.g. herpesviruses in individuals with impaired T-cell and/or iNKT cell responses. To allow lifelong persistence and virus production in the face of primed immunity, herpesviruses exploit immune evasion strategies. These include a reduction in viral antigen expression during latency and a number of escape mechanisms that target antigen presentation pathways. Given the plethora of foreign antigens expressed in virus-producing cells, herpesviruses are conceivably most vulnerable to elimination by cell-mediated immunity during the replicative phase of infection. Here, we show that a prototypic herpesvirus, Epstein-Barr virus (EBV), encodes a novel, broadly acting immunoevasin, gp150, that is expressed during the late phase of viral replication. In particular, EBV gp150 inhibits antigen presentation by HLA class I, HLA class II, and the non-classical, lipid-presenting CD1d molecules. The mechanism of gp150-mediated T-cell escape does not depend on degradation of the antigen-presenting molecules nor does it require gp150’s cytoplasmic tail. Through its abundant glycosylation, gp150 creates a shield that impedes surface presentation of antigen. This is an unprecedented immune evasion mechanism for herpesviruses. In view of its likely broader target range, gp150 could additionally have an impact beyond escape of T cell activation. Importantly, B cells infected with a gp150-null mutant EBV displayed rescued levels of surface antigen presentation by HLA class I, HLA class II, and CD1d, supporting an important role for iNKT cells next to classical T cells in fighting EBV infection. At the same time, our results indicate that EBV gp150 prolongs the timespan for producing viral offspring at the most vulnerable stage of the viral life cycle. The human herpesvirus Epstein-Barr virus (EBV) is an important human pathogen involved in infectious mononucleosis and several malignant tumors, including lymphomas in the immunosuppressed. Upon primary infection, a balance between virus and host is established, to which EBV’s capacity to dodge T cell-mediated attack contributes. Here we identify the late protein EBV gp150 as a novel immunoevasin, frustrating antigen presentation by HLA class I, class II, and CD1d molecules. EBV gp150’s many sialoglycans create a shield impeding surface detection of presented antigen. Interestingly, exploiting glycan shielding as a mechanism to mask surface exposed proteins on infected cells could permit EBV to additionally modulate other aspects of host antiviral defense. B cells producing wild-type EBV escaped immune recognition more efficiently than those infected with a gp150-null virus, pointing towards a role for gp150 in natural infection. Our results reveal a novel, broadly active strategy by which a herpesvirus glycoprotein, EBV gp150, blocks antigen presentation to T cells through glycan shielding, a new paradigm in herpesvirus immune evasion.
Collapse
Affiliation(s)
- Anna M. Gram
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Timo Oosenbrug
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Christian Büll
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Anouskha Comvalius
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kathryn J. I. Dickson
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Joop Wiegant
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Vrolijk
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Jan Lebbink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ron Wolterbeek
- Department of Medical Statistics & Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Gosse J. Adema
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - David C. Tscharke
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Lindsey M. Hutt-Fletcher
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | | | - Rob C. Hoeben
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maaike E. Ressing
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
80
|
Wang J, Gui L, Chen ZY, Zhang QY. Mutations in the C-terminal region affect subcellular localization of crucian carp herpesvirus (CaHV) GPCR. Virus Genes 2016; 52:484-94. [PMID: 27059239 PMCID: PMC4923094 DOI: 10.1007/s11262-016-1325-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/18/2016] [Indexed: 11/25/2022]
Abstract
G protein-coupled receptors (GPCRs) are known as seven transmembrane domain receptors and consequently can mediate diverse biological functions via regulation of their subcellular localization. Crucian carp herpesvirus (CaHV) was recently isolated from infected fish with acute gill hemorrhage. CaHV GPCR of 349 amino acids (aa) was identified based on amino acid identity. A series of variants with truncation/deletion/substitution mutation in the C-terminal (aa 315–349) were constructed and expressed in fathead minnow (FHM) cells. The roles of three key C-terminal regions in subcellular localization of CaHV GPCR were determined. Lysine-315 (K-315) directed the aggregation of the protein preferentially at the nuclear side. Predicted N-myristoylation site (GGGWTR, aa 335–340) was responsible for punctate distribution in periplasm or throughout the cytoplasm. Predicted phosphorylation site (SSR, aa 327–329) and GGGWTR together determined the punctate distribution in cytoplasm. Detection of organelles localization by specific markers showed that the protein retaining K-315 colocalized with the Golgi apparatus. These experiments provided first evidence that different mutations of CaHV GPCR C-terminals have different affects on the subcellular localization of fish herpesvirus-encoded GPCRs. The study provided valuable information and new insights into the precise interactions between herpesvirus and fish cells, and could also provide useful targets for antiviral agents in aquaculture.
Collapse
Affiliation(s)
- Jun Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of Chinese Academy of Sciences, Wuhan, 430072, China
| | - Lang Gui
- Fisheries and Life Science, Shanghai Ocean University, Shanghai, 200120, China
| | - Zong-Yan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of Chinese Academy of Sciences, Wuhan, 430072, China
| | - Qi-Ya Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
81
|
Schuren AB, Costa AI, Wiertz EJ. Recent advances in viral evasion of the MHC Class I processing pathway. Curr Opin Immunol 2016; 40:43-50. [PMID: 27065088 DOI: 10.1016/j.coi.2016.02.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 02/12/2016] [Accepted: 02/22/2016] [Indexed: 11/17/2022]
Abstract
T-cell mediated adaptive immunity against viruses relies on recognition of virus-derived peptides by CD4(+) and CD8(+) T cells. Detection of pathogen-derived peptide-MHC-I complexes triggers CD8(+) T cells to eliminate the infected cells. Viruses have evolved several mechanisms to avoid recognition, many of which target the MHC-I antigen-processing pathway. While many immune evasion strategies have been described in the context of herpesvirus infections, it is becoming clear that this 'disguise' ability is more widespread. Here, we address recent findings in viral evasion of the MHC-I antigen presentation pathway and the impact on CD8(+) T cell responses.
Collapse
Affiliation(s)
- Anouk Bc Schuren
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ana I Costa
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Emmanuel Jhj Wiertz
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
82
|
Zhang J, Feng H, Xu S, Feng P. Hijacking GPCRs by viral pathogens and tumor. Biochem Pharmacol 2016; 114:69-81. [PMID: 27060663 DOI: 10.1016/j.bcp.2016.03.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 03/25/2016] [Indexed: 01/11/2023]
Abstract
G protein-coupled receptors (GPCRs) constitute the largest family of molecules that transduce signals across the plasma membrane. Herpesviruses are successful pathogens that evolved diverse mechanisms to benefit their infection. Several human herpesviruses express GPCRs to exploit cellular signaling cascades during infection. These viral GPCRs demonstrate distinct biochemical and biophysical properties that result in the activation of a broad spectrum of signaling pathways. In immune-deficient individuals, human herpesvirus infection and the expression of their GPCRs are implicated in virus-associated diseases and pathologies. Emerging studies also uncover diverse mutations in components, particularly GPCRs and small G proteins, of GPCR signaling pathways that render the constitutive activation of proliferative and survival signal, which contributes to the oncogenesis of various human cancers. Hijacking GPCR-mediated signaling is a signature shared by diseases associated with constitutively active viral GPCRs and cellular mutations activating GPCR signaling, exposing key molecules that can be targeted for anti-viral and anti-tumor therapy.
Collapse
Affiliation(s)
- Junjie Zhang
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA 90033, United States.
| | - Hao Feng
- Key Laboratory of Protein Chemistry and Fish Developmental Biology of Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Simin Xu
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA 90033, United States
| | - Pinghui Feng
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA 90033, United States.
| |
Collapse
|
83
|
Induction of the Lytic Cycle Sensitizes Epstein-Barr Virus-Infected B Cells to NK Cell Killing That Is Counteracted by Virus-Mediated NK Cell Evasion Mechanisms in the Late Lytic Cycle. J Virol 2015; 90:947-58. [PMID: 26537677 DOI: 10.1128/jvi.01932-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/26/2015] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Epstein-Barr Virus (EBV) persists for the lifetime of the infected host despite eliciting strong immune responses. This persistence requires a fine balance between the host immune system and EBV immune evasion. Accumulating evidence suggests an important role for natural killer (NK) cells in this balance. NK cells can kill EBV-infected cells undergoing lytic replication in vitro, and studies in both humans and mice with reconstituted human immune systems have shown that NK cells can limit EBV replication and prevent infectious mononucleosis. We now show that NK cells, via NKG2D and DNAM-1 interactions, recognize and kill EBV-infected cells undergoing lytic replication and that expression of a single EBV lytic gene, BZLF1, is sufficient to trigger sensitization to NK cell killing. We also present evidence suggesting the possibility of the existence of an as-yet-unidentified DNAM-1 ligand which may be particularly important for killing lytically infected normal B cells. Furthermore, while cells entering the lytic cycle become sensitized to NK cell killing, we observed that cells in the late lytic cycle are highly resistant. We identified expression of the vBcl-2 protein, BHRF1, as one effective mechanism by which EBV mediates this protection. Thus, contrary to the view expressed in some reports, EBV has evolved the ability to evade NK cell responses. IMPORTANCE This report extends our understanding of the interaction between EBV and host innate responses. It provides the first evidence that the susceptibility to NK cell lysis of EBV-infected B cells undergoing lytic replication is dependent upon the phase of the lytic cycle. Induction of the lytic cycle is associated with acquired sensitization to NK cell killing, while progress through the late lytic cycle is associated with acquired resistance to killing. We provide mechanistic explanations for this novel observation, indicating important roles for the BZLF1 immediate early transactivator, the BHRF1 vBcl-2 homologue, and a novel ligand for the DNAM-1 NK cell receptor.
Collapse
|
84
|
Han L, He H, Li F, Cui X, Xie D, Liu Y, Zheng X, Bai H, Wang S, Bo X. Inferring Infection Patterns Based on a Connectivity Map of Host Transcriptional Responses. Sci Rep 2015; 5:15820. [PMID: 26508266 PMCID: PMC4623713 DOI: 10.1038/srep15820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 10/01/2015] [Indexed: 12/25/2022] Open
Abstract
Host responses to infections represent an important pathogenicity determiner, and delineation of host responses can elucidate pathogenesis processes and inform the development of anti-infection therapies. Low cost, high throughput, easy quantitation, and rich descriptions have made gene expression profiling generated by DNA microarrays an optimal approach for describing host transcriptional responses (HTRs). However, efforts to characterize the landscape of HTRs to diverse pathogens are far from offering a comprehensive view. Here, we developed an HTR Connectivity Map based on systematic assessment of pairwise similarities of HTRs to 50 clinically important human pathogens using 1353 gene-expression profiles generated from >60 human cells/tissues. These 50 pathogens were further partitioned into eight robust “HTR communities” (i.e., groups with more consensus internal HTR similarities). These communities showed enrichment in specific infection attributes and differential gene expression patterns. Using query signatures of HTRs to external pathogens, we demonstrated four distinct modes of HTR associations among different pathogens types/class, and validated the reliability of the HTR community divisions for differentiating and categorizing pathogens from a host-oriented perspective. These findings provide a first-generation HTR Connectivity Map of 50 diverse pathogens, and demonstrate the potential for using annotated HTR community to detect functional associations among infectious pathogens.
Collapse
Affiliation(s)
- Lu Han
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,Department of Traditional Chinese Medicine and Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Haochen He
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Fei Li
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiuliang Cui
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, 200433, China
| | - Dafei Xie
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yang Liu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiaofei Zheng
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hui Bai
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,Department of Pharmacy, No.451 hospital of People's Liberation Army, Xi'an, 710065, China
| | - Shengqi Wang
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiaochen Bo
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| |
Collapse
|
85
|
The Missing Link in Epstein-Barr Virus Immune Evasion: the BDLF3 Gene Induces Ubiquitination and Downregulation of Major Histocompatibility Complex Class I (MHC-I) and MHC-II. J Virol 2015; 90:356-67. [PMID: 26468525 DOI: 10.1128/jvi.02183-15] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/09/2015] [Indexed: 01/26/2023] Open
Abstract
UNLABELLED The ability of Epstein-Barr virus (EBV) to spread and persist in human populations relies on a balance between host immune responses and EBV immune evasion. CD8(+) cells specific for EBV late lytic cycle antigens show poor recognition of target cells compared to immediate early and early antigen-specific CD8(+) cells. This phenomenon is due in part to the early EBV protein BILF1, whose immunosuppressive activity increases with lytic cycle progression. However, published data suggest the existence of a hitherto unidentified immune evasion protein further enhancing protection against late EBV antigen-specific CD8(+) cells. We have now identified the late lytic BDLF3 gene as the missing link accounting for efficient evasion during the late lytic cycle. Interestingly, BDLF3 also contributes to evasion of CD4(+) cell responses to EBV. We report that BDLF3 downregulates expression of surface major histocompatibility complex (MHC) class I and class II molecules in the absence of any effect upon other surface molecules screened, including CD54 (ICAM-1) and CD71 (transferrin receptor). BDLF3 both enhanced internalization of surface MHC molecules and reduced the rate of their appearance at the cell surface. The reduced expression of surface MHC molecules correlated with functional protection against CD8(+) and CD4(+) T cell recognition. The molecular mechanism was identified as BDLF3-induced ubiquitination of MHC molecules and their subsequent downregulation in a proteasome-dependent manner. IMPORTANCE Immune evasion is a necessary feature of viruses that establish lifelong persistent infections in the face of strong immune responses. EBV is an important human pathogen whose immune evasion mechanisms are only partly understood. Of the EBV immune evasion mechanisms identified to date, none could explain why CD8(+) T cell responses to late lytic cycle genes are so infrequent and, when present, recognize lytically infected target cells so poorly relative to CD8(+) T cells specific for early lytic cycle antigens. The present work identifies an additional immune evasion protein, BDLF3, that is expressed late in the lytic cycle and impairs CD8(+) T cell recognition by targeting cell surface MHC class I molecules for ubiquitination and proteasome-dependent downregulation. Interestingly, BDLF3 also targets MHC class II molecules to impair CD4(+) T cell recognition. BDLF3 is therefore a rare example of a viral protein that impairs both the MHC class I and class II antigen-presenting pathways.
Collapse
|
86
|
Abstract
Glycoproteins are critical to virus entry, to spread within and between hosts and can modify the behavior of cells. Many viruses carry only a few, most found in the virion envelope. EBV makes more than 12, providing flexibility in how it colonizes its human host. Some are dedicated to getting the virus through the cell membrane and on toward the nucleus of the cell, some help guide the virus back out and on to the next cell in the same or a new host. Yet others undermine host defenses helping the virus persist for a lifetime, maintaining a presence that is mostly tolerated and serves to perpetuate EBV as one of the most common infections of man.
Collapse
Affiliation(s)
- Lindsey M Hutt-Fletcher
- Department of Microbiology & Immunology, Feist-Weiller Cancer Center and Center for Molecular & Tumor Virology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, USA; Tel.: +1 318 675 4948
| |
Collapse
|
87
|
Latent Membrane Protein LMP2A Impairs Recognition of EBV-Infected Cells by CD8+ T Cells. PLoS Pathog 2015; 11:e1004906. [PMID: 26067064 PMCID: PMC4465838 DOI: 10.1371/journal.ppat.1004906] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 04/22/2015] [Indexed: 01/04/2023] Open
Abstract
The common pathogen Epstein-Barr virus (EBV) transforms normal human B cells and can cause cancer. Latent membrane protein 2A (LMP2A) of EBV supports activation and proliferation of infected B cells and is expressed in many types of EBV-associated cancer. It is not clear how latent EBV infection and cancer escape elimination by host immunity, and it is unknown whether LMP2A can influence the interaction of EBV-infected cells with the immune system. We infected primary B cells with EBV deleted for LMP2A, and established lymphoblastoid cell lines (LCLs). We found that CD8+ T cell clones showed higher reactivity against LMP2A-deficient LCLs compared to LCLs infected with complete EBV. We identified several potential mediators of this immunomodulatory effect. In the absence of LMP2A, expression of some EBV latent antigens was elevated, and cell surface expression of MHC class I was marginally increased. LMP2A-deficient LCLs produced lower amounts of IL-10, although this did not directly affect CD8+ T cell recognition. Deletion of LMP2A led to several changes in the cell surface immunophenotype of LCLs. Specifically, the agonistic NKG2D ligands MICA and ULBP4 were increased. Blocking experiments showed that NKG2D activation contributed to LCL recognition by CD8+ T cell clones. Our results demonstrate that LMP2A reduces the reactivity of CD8+ T cells against EBV-infected cells, and we identify several relevant mechanisms. Epstein-Barr virus (EBV) is carried by most humans. It can cause several types of cancer. In healthy infected people, EBV persists for life in a "latent" state in white blood cells called B cells. For infected persons to remain healthy, it is crucial that they harbor CD8-positive "killer" T cells that recognize and destroy precancerous EBV-infected cells. However, this protection is imperfect, because the virus is not eliminated from the body, and the danger of EBV-associated cancer remains. How does the virus counteract CD8+ T cell control? Here we study the effects of latent membrane protein 2A (LMP2A), which is an important viral molecule because it is present in several types of EBV-associated cancers, and in latently infected cells in healthy people. We show that LMP2A counteracts the recognition of EBV-infected B cells by antiviral killer cells. We found a number of mechanisms that are relevant to this effect. Notably, LMP2A disturbs expression of molecules on B cells that interact with NKG2D, a molecule on the surface of CD8+ T cells that aids their activation. In this way, LMP2A weakens important immune responses against EBV. Similar mechanisms may operate in different types of LMP2A-expressing cancers caused by EBV.
Collapse
|
88
|
Liu S, Wang X, Shu J, Zhao Z, Sun Z, Luo B. Sequence analysis of EBV immune evasion gene BNLF2a in EBV associated tumors and healthy individuals from nasopharyngeal carcinoma endemic and non-endemic regions of China. J Med Virol 2015; 87:1946-52. [PMID: 25959517 DOI: 10.1002/jmv.24254] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2015] [Indexed: 11/07/2022]
Abstract
BNLF2a is an Epstein-Barr virus (EBV) immune evasion gene. Its protein is located in the endoplasmic reticulum (ER) membrane, and can inhibit the antigen transporting function of TAP, thereby perturbing the immune response to EBV in lytic and prelatent phase. In order to explore whether the polymorphism of BNLF2a gene has a role in different types of EBV associated tumors, we conducted complete sequencing of the gene BNLF2a in 408 cases of EBV positive tumors (76 lymphomas, 45 gastric carcinomas, and 85 nasopharyngeal carcinomas in northern China and 27 lymphomas, 30 gastric carcinomas, and 57 nasopharyngeal carcinomas in southern China) and throat washings from healthy individuals (39 in northern China and 49 in southern China). Two main variant types of BNLF2a were identified. Type BNLF2a-A, which was similar to B95-8, was dominant in all sub-populations (66.7-100%) in this study. Type BNLF2a-B was characterized by the mutations at position 8 and 40. The variation patterns of BNLF2a were significantly different between samples from northern and southern China (P < 0.05), and between the tumors and healthy donor samples from the northern China (P < 0.0167). Type BNLF2a-B was more frequent in healthy donors of northern China (33.3%), and the proportion of this type was higher in the northern than in the southern NPCs. These data demonstrate that the BNLF2a gene is highly conserved, and its polymorphism is geographically restricted. Type BNLF2a-B is more prevalent in northern China and may be less tumor transformative.
Collapse
Affiliation(s)
- Song Liu
- Department of Medical Microbiology, Qingdao University Medical College, Qingdao, China
| | - Xiaofeng Wang
- Department of Medical Microbiology, Qingdao University Medical College, Qingdao, China
| | - Jun Shu
- Department of Medical Microbiology, Qingdao University Medical College, Qingdao, China
| | - Zhenzhen Zhao
- Department of Medical Microbiology, Qingdao University Medical College, Qingdao, China
| | - Zhifu Sun
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Bing Luo
- Department of Medical Microbiology, Qingdao University Medical College, Qingdao, China
| |
Collapse
|
89
|
Verweij MC, Horst D, Griffin BD, Luteijn RD, Davison AJ, Ressing ME, Wiertz EJHJ. Viral inhibition of the transporter associated with antigen processing (TAP): a striking example of functional convergent evolution. PLoS Pathog 2015; 11:e1004743. [PMID: 25880312 PMCID: PMC4399834 DOI: 10.1371/journal.ppat.1004743] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Herpesviruses are large DNA viruses that are highly abundant within their host populations. Even in the presence of a healthy immune system, these viruses manage to cause lifelong infections. This persistence is partially mediated by the virus entering latency, a phase of infection characterized by limited viral protein expression. Moreover, herpesviruses have devoted a significant part of their coding capacity to immune evasion strategies. It is believed that the close coexistence of herpesviruses and their hosts has resulted in the evolution of viral proteins that specifically attack multiple arms of the host immune system. Cytotoxic T lymphocytes (CTLs) play an important role in antiviral immunity. CTLs recognize their target through viral peptides presented in the context of MHC molecules at the cell surface. Every herpesvirus studied to date encodes multiple immune evasion molecules that effectively interfere with specific steps of the MHC class I antigen presentation pathway. The transporter associated with antigen processing (TAP) plays a key role in the loading of viral peptides onto MHC class I molecules. This is reflected by the numerous ways herpesviruses have developed to block TAP function. In this review, we describe the characteristics and mechanisms of action of all known virus-encoded TAP inhibitors. Orthologs of these proteins encoded by related viruses are identified, and the conservation of TAP inhibition is discussed. A phylogenetic analysis of members of the family Herpesviridae is included to study the origin of these molecules. In addition, we discuss the characteristics of the first TAP inhibitor identified outside the herpesvirus family, namely, in cowpox virus. The strategies of TAP inhibition employed by viruses are very distinct and are likely to have been acquired independently during evolution. These findings and the recent discovery of a non-herpesvirus TAP inhibitor represent a striking example of functional convergent evolution.
Collapse
Affiliation(s)
- Marieke C. Verweij
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Daniëlle Horst
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bryan D. Griffin
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rutger D. Luteijn
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Andrew J. Davison
- MRC—University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Maaike E. Ressing
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Emmanuel J. H. J. Wiertz
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
90
|
van de Weijer ML, Luteijn RD, Wiertz EJHJ. Viral immune evasion: Lessons in MHC class I antigen presentation. Semin Immunol 2015; 27:125-37. [PMID: 25887630 DOI: 10.1016/j.smim.2015.03.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 03/13/2015] [Indexed: 12/19/2022]
Abstract
The MHC class I antigen presentation pathway enables cells infected with intracellular pathogens to signal the presence of the invader to the immune system. Cytotoxic T lymphocytes are able to eliminate the infected cells through recognition of pathogen-derived peptides presented by MHC class I molecules at the cell surface. In the course of evolution, many viruses have acquired inhibitors that target essential stages of the MHC class I antigen presentation pathway. Studies on these immune evasion proteins reveal fascinating strategies used by viruses to elude the immune system. Viral immunoevasins also constitute great research tools that facilitate functional studies on the MHC class I antigen presentation pathway, allowing the investigation of less well understood routes, such as TAP-independent antigen presentation and cross-presentation of exogenous proteins. Viral immunoevasins have also helped to unravel more general cellular processes. For instance, basic principles of ER-associated protein degradation via the ubiquitin-proteasome pathway have been resolved using virus-induced degradation of MHC class I as a model. This review highlights how viral immunoevasins have increased our understanding of MHC class I-restricted antigen presentation.
Collapse
Affiliation(s)
| | - Rutger D Luteijn
- Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Emmanuel J H J Wiertz
- Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands.
| |
Collapse
|
91
|
van Gent M, Gram AM, Boer IGJ, Geerdink RJ, Lindenbergh MFS, Lebbink RJ, Wiertz EJ, Ressing ME. Silencing the shutoff protein of Epstein–Barr virus in productively infected B cells points to (innate) targets for immune evasion. J Gen Virol 2015; 96:858-865. [DOI: 10.1099/jgv.0.000021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Michiel van Gent
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anna M. Gram
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ingrid G. J. Boer
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ruben J. Geerdink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Robert Jan Lebbink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Emmanuel J. Wiertz
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maaike E. Ressing
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
92
|
de Munnik SM, Smit MJ, Leurs R, Vischer HF. Modulation of cellular signaling by herpesvirus-encoded G protein-coupled receptors. Front Pharmacol 2015; 6:40. [PMID: 25805993 PMCID: PMC4353375 DOI: 10.3389/fphar.2015.00040] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/12/2015] [Indexed: 12/22/2022] Open
Abstract
Human herpesviruses (HHVs) are widespread infectious pathogens that have been associated with proliferative and inflammatory diseases. During viral evolution, HHVs have pirated genes encoding viral G protein-coupled receptors (vGPCRs), which are expressed on infected host cells. These vGPCRs show highest homology to human chemokine receptors, which play a key role in the immune system. Importantly, vGPCRs have acquired unique properties such as constitutive activity and the ability to bind a broad range of human chemokines. This allows vGPCRs to hijack human proteins and modulate cellular signaling for the benefit of the virus, ultimately resulting in immune evasion and viral dissemination to establish a widespread and lifelong infection. Knowledge on the mechanisms by which herpesviruses reprogram cellular signaling might provide insight in the contribution of vGPCRs to viral survival and herpesvirus-associated pathologies.
Collapse
Affiliation(s)
- Sabrina M de Munnik
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| | - Martine J Smit
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| | - Rob Leurs
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| | - Henry F Vischer
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| |
Collapse
|
93
|
Viruses and human cancers: a long road of discovery of molecular paradigms. Clin Microbiol Rev 2015; 27:463-81. [PMID: 24982317 DOI: 10.1128/cmr.00124-13] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
About a fifth of all human cancers worldwide are caused by infectious agents. In 12% of cancers, seven different viruses have been causally linked to human oncogenesis: Epstein-Barr virus, hepatitis B virus, human papillomavirus, human T-cell lymphotropic virus, hepatitis C virus, Kaposi's sarcoma herpesvirus, and Merkel cell polyomavirus. Here, we review the many molecular mechanisms of oncogenesis that have been discovered over the decades of study of these viruses. We discuss how viruses can act at different stages in the complex multistep process of carcinogenesis. Early events include their involvement in mutagenic events associated with tumor initiation such as viral integration and insertional mutagenesis as well as viral promotion of DNA damage. Also involved in tumor progression is the dysregulation of cellular processes by viral proteins, and we describe how this has been investigated by studies in cell culture and in experimental animals and by molecular cellular approaches. Also important are the molecular mechanisms whereby viruses interact with the immune system and the immune evasion strategies that have evolved.
Collapse
|
94
|
Epstein-Barr virus LMP2A suppresses MHC class II expression by regulating the B-cell transcription factors E47 and PU.1. Blood 2015; 125:2228-38. [PMID: 25631773 DOI: 10.1182/blood-2014-08-594689] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 01/25/2015] [Indexed: 02/07/2023] Open
Abstract
Oncogenic Epstein-Barr virus (EBV) uses various approaches to escape host immune responses and persist in B cells. Such persistent infections may provide the opportunity for this virus to initiate tumor formation. Using EBV-immortalized lymphoblastoid cell lines (LCLs) as a model, we found that the expression of major histocompatibility complex (MHC) class II and CD74 in B cells is repressed after EBV infection. Class II transactivator (CIITA) is the master regulator of MHC class II-related genes. As expected, CIITA was downregulated in LCLs. We showed that downregulation of CIITA is caused by EBV latent membrane protein 2A (LMP2A) and driven by the CIITA-PIII promoter. Furthermore, we demonstrated that LMP2A-mediated E47 and PU.1 reduction resulted in CIITA suppression. Mechanistically, the LMP2A immunoreceptor tyrosine-based activation motif was critical for the repression of E47 and PU.1 promoter activity via Syk, Src, and the phosphatidylinositol 3-kinase/Akt pathway. Elimination of LMP2A in LCLs using a shLMP2A approach showed that the expression levels of E47, PU.1, CIITA, MHC class II, and CD74 are reversed. These data indicated that the LMP2A may reduce MHC class II expression through interference with the E47/PU.1-CIITA pathway. Finally, we demonstrated that MHC class II may be detected in tonsils and EBV-negative Hodgkin disease but not in EBV-associated posttransplant lymphoproliferative disease and Hodgkin disease.
Collapse
|
95
|
Hatton OL, Harris-Arnold A, Schaffert S, Krams SM, Martinez OM. The interplay between Epstein-Barr virus and B lymphocytes: implications for infection, immunity, and disease. Immunol Res 2015; 58:268-76. [PMID: 24619311 DOI: 10.1007/s12026-014-8496-1] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human B cells are the primary targets of Epstein-Barr virus (EBV) infection. In most cases, EBV infection is asymptomatic because of a highly effective host immune response, but some individuals develop self-limiting infectious mononucleosis, while others develop EBV-associated lymphoid or epithelial malignancies. The viral and immune factors that determine the outcome of infection are not understood. The EBV life cycle includes a lytic phase, culminating in the production of new viral particles, and a latent phase, during which the virus remains largely silent for the lifetime of the host in memory B cells. Thus, in healthy individuals, there is a tightly orchestrated interplay between EBV and the host that allows the virus to persist. To promote viral persistence, EBV has evolved a variety of strategies to modulate the host immune response including inhibition of immune cell function, blunting of apoptotic pathways, and interfering with antigen processing and presentation pathways. In this article, we focus on mechanisms by which dysregulation of the host B cell and immune modulation by the virus can contribute to development of EBV+ B cell lymphomas.
Collapse
Affiliation(s)
- Olivia L Hatton
- Program in Immunology and Department of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | | | | | | |
Collapse
|
96
|
Ressing ME, van Gent M, Gram AM, Hooykaas MJG, Piersma SJ, Wiertz EJHJ. Immune Evasion by Epstein-Barr Virus. Curr Top Microbiol Immunol 2015; 391:355-81. [PMID: 26428381 DOI: 10.1007/978-3-319-22834-1_12] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epstein-Bar virus (EBV) is widespread within the human population with over 90% of adults being infected. In response to primary EBV infection, the host mounts an antiviral immune response comprising both innate and adaptive effector functions. Although the immune system can control EBV infection to a large extent, the virus is not cleared. Instead, EBV establishes a latent infection in B lymphocytes characterized by limited viral gene expression. For the production of new viral progeny, EBV reactivates from these latently infected cells. During the productive phase of infection, a repertoire of over 80 EBV gene products is expressed, presenting a vast number of viral antigens to the primed immune system. In particular the EBV-specific CD4+ and CD8+ memory T lymphocytes can respond within hours, potentially destroying the virus-producing cells before viral replication is completed and viral particles have been released. Preceding the adaptive immune response, potent innate immune mechanisms provide a first line of defense during primary and recurrent infections. In spite of this broad range of antiviral immune effector mechanisms, EBV persists for life and continues to replicate. Studies performed over the past decades have revealed a wide array of viral gene products interfering with both innate and adaptive immunity. These include EBV-encoded proteins as well as small noncoding RNAs with immune-evasive properties. The current review presents an overview of the evasion strategies that are employed by EBV to facilitate immune escape during latency and productive infection. These evasion mechanisms may also compromise the elimination of EBV-transformed cells, and thus contribute to malignancies associated with EBV infection.
Collapse
Affiliation(s)
- Maaike E Ressing
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michiel van Gent
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anna M Gram
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marjolein J G Hooykaas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sytse J Piersma
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Emmanuel J H J Wiertz
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
97
|
Arfelt KN, Fares S, Rosenkilde MM. EBV, the Human Host, and the 7TM Receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 129:395-427. [DOI: 10.1016/bs.pmbts.2014.10.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
98
|
Identification and functional comparison of seven-transmembrane G-protein-coupled BILF1 receptors in recently discovered nonhuman primate lymphocryptoviruses. J Virol 2014; 89:2253-67. [PMID: 25505061 DOI: 10.1128/jvi.02716-14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Coevolution of herpesviruses with their respective host has resulted in a delicate balance between virus-encoded immune evasion mechanisms and host antiviral immunity. BILF1 encoded by human Epstein-Barr virus (EBV) is a 7-transmembrane (7TM) G-protein-coupled receptor (GPCR) with multiple immunomodulatory functions, including attenuation of PKR phosphorylation, activation of G-protein signaling, and downregulation of major histocompatibility complex (MHC) class I surface expression. In this study, we explored the evolutionary and functional relationships between BILF1 receptor family members from EBV and 12 previously uncharacterized nonhuman primate (NHP) lymphocryptoviruses (LCVs). Phylogenetic analysis defined 3 BILF1 clades, corresponding to LCVs of New World monkeys (clade A) or Old World monkeys and great apes (clades B and C). Common functional properties were suggested by a high degree of sequence conservation in functionally important regions of the BILF1 molecules. A subset of BILF1 receptors from EBV and LCVs from NHPs (chimpanzee, orangutan, marmoset, and siamang) were selected for multifunctional analysis. All receptors exhibited constitutive signaling activity via G protein Gαi and induced activation of the NF-κB transcription factor. In contrast, only 3 of 5 were able to activate NFAT (nuclear factor of activated T cells); chimpanzee and orangutan BILF1 molecules were unable to activate NFAT. Similarly, although all receptors were internalized, BILF1 from the chimpanzee and orangutan displayed an altered cellular localization pattern with predominant cell surface expression. This study shows how biochemical characterization of functionally important orthologous viral proteins can be used to complement phylogenetic analysis to provide further insight into diverse microbial evolutionary relationships and immune evasion function. IMPORTANCE Epstein-Barr virus (EBV), known as an oncovirus, is the only human herpesvirus in the genus Lymphocryptovirus (LCV). EBV uses multiple strategies to hijack infected host cells, establish persistent infection in B cells, and evade antiviral immune responses. As part of EBV's immune evasion strategy, the virus encodes a multifunctional 7-transmembrane (7TM) G-protein-coupled receptor (GPCR), EBV BILF1. In addition to multiple immune evasion-associated functions, EBV BILF1 has transforming properties, which are linked to its high constitutive activity. We identified BILF1 receptor orthologues in 12 previously uncharacterized LCVs from nonhuman primates (NHPs) of Old and New World origin. As 7TM receptors are excellent drug targets, our unique insight into the molecular mechanism of action of the BILF1 family and into the evolution of primate LCVs may enable validation of EBV BILF1 as a drug target for EBV-mediated diseases, as well as facilitating the design of drugs targeting EBV BILF1.
Collapse
|
99
|
Cytomegalovirus immune evasion by perturbation of endosomal trafficking. Cell Mol Immunol 2014; 12:154-69. [PMID: 25263490 PMCID: PMC4654299 DOI: 10.1038/cmi.2014.85] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/15/2014] [Accepted: 08/16/2014] [Indexed: 12/30/2022] Open
Abstract
Cytomegaloviruses (CMVs), members of the herpesvirus family, have evolved a variety of mechanisms to evade the immune response to survive in infected hosts and to establish latent infection. They effectively hide infected cells from the effector mechanisms of adaptive immunity by eliminating cellular proteins (major histocompatibility Class I and Class II molecules) from the cell surface that display viral antigens to CD8 and CD4 T lymphocytes. CMVs also successfully escape recognition and elimination of infected cells by natural killer (NK) cells, effector cells of innate immunity, either by mimicking NK cell inhibitory ligands or by downregulating NK cell-activating ligands. To accomplish these immunoevasion functions, CMVs encode several proteins that function in the biosynthetic pathway by inhibiting the assembly and trafficking of cellular proteins that participate in immune recognition and thereby, block their appearance at the cell surface. However, elimination of these proteins from the cell surface can also be achieved by perturbation of their endosomal route and subsequent relocation from the cell surface into intracellular compartments. Namely, the physiological route of every cellular protein, including immune recognition molecules, is characterized by specific features that determine its residence time at the cell surface. In this review, we summarize the current understanding of endocytic trafficking of immune recognition molecules and perturbations of the endosomal system during infection with CMVs and other members of the herpesvirus family that contribute to their immune evasion mechanisms.
Collapse
|
100
|
Quinn LL, Zuo J, Abbott RJM, Shannon-Lowe C, Tierney RJ, Hislop AD, Rowe M. Cooperation between Epstein-Barr virus immune evasion proteins spreads protection from CD8+ T cell recognition across all three phases of the lytic cycle. PLoS Pathog 2014; 10:e1004322. [PMID: 25144360 PMCID: PMC4140850 DOI: 10.1371/journal.ppat.1004322] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 07/08/2014] [Indexed: 12/20/2022] Open
Abstract
CD8+ T cell responses to Epstein-Barr virus (EBV) lytic cycle expressed antigens display a hierarchy of immunodominance, in which responses to epitopes of immediate-early (IE) and some early (E) antigens are more frequently observed than responses to epitopes of late (L) expressed antigens. It has been proposed that this hierarchy, which correlates with the phase-specific efficiency of antigen presentation, may be due to the influence of viral immune-evasion genes. At least three EBV-encoded genes, BNLF2a, BGLF5 and BILF1, have the potential to inhibit processing and presentation of CD8+ T cell epitopes. Here we examined the relative contribution of these genes to modulation of CD8+ T cell recognition of EBV lytic antigens expressed at different phases of the replication cycle in EBV-transformed B-cells (LCLs) which spontaneously reactivate lytic cycle. Selective shRNA-mediated knockdown of BNLF2a expression led to more efficient recognition of immediate-early (IE)- and early (E)-derived epitopes by CD8+ T cells, while knock down of BILF1 increased recognition of epitopes from E and late (L)-expressed antigens. Contrary to what might have been predicted from previous ectopic expression studies in EBV-negative model cell lines, the shRNA-mediated inhibition of BGLF5 expression in LCLs showed only modest, if any, increase in recognition of epitopes expressed in any phase of lytic cycle. These data indicate that whilst BNLF2a interferes with antigen presentation with diminishing efficiency as lytic cycle progresses (IE>E>>L), interference by BILF1 increases with progression through lytic cycle (IE
Collapse
Affiliation(s)
- Laura L. Quinn
- School of Cancer Sciences and Centre for Human Virology, University of Birmingham College of Medical and Dental Sciences, Edgbaston, Birmingham, United Kingdom
| | - Jianmin Zuo
- School of Cancer Sciences and Centre for Human Virology, University of Birmingham College of Medical and Dental Sciences, Edgbaston, Birmingham, United Kingdom
| | - Rachel J. M. Abbott
- School of Cancer Sciences and Centre for Human Virology, University of Birmingham College of Medical and Dental Sciences, Edgbaston, Birmingham, United Kingdom
| | - Claire Shannon-Lowe
- School of Cancer Sciences and Centre for Human Virology, University of Birmingham College of Medical and Dental Sciences, Edgbaston, Birmingham, United Kingdom
| | - Rosemary J. Tierney
- School of Cancer Sciences and Centre for Human Virology, University of Birmingham College of Medical and Dental Sciences, Edgbaston, Birmingham, United Kingdom
| | - Andrew D. Hislop
- School of Cancer Sciences and Centre for Human Virology, University of Birmingham College of Medical and Dental Sciences, Edgbaston, Birmingham, United Kingdom
| | - Martin Rowe
- School of Cancer Sciences and Centre for Human Virology, University of Birmingham College of Medical and Dental Sciences, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|