1
|
Wagner KI, Mateyka LM, Jarosch S, Grass V, Weber S, Schober K, Hammel M, Burrell T, Kalali B, Poppert H, Beyer H, Schambeck S, Holdenrieder S, Strötges-Achatz A, Haselmann V, Neumaier M, Erber J, Priller A, Yazici S, Roggendorf H, Odendahl M, Tonn T, Dick A, Witter K, Mijočević H, Protzer U, Knolle PA, Pichlmair A, Crowell CS, Gerhard M, D'Ippolito E, Busch DH. Recruitment of highly cytotoxic CD8 + T cell receptors in mild SARS-CoV-2 infection. Cell Rep 2021; 38:110214. [PMID: 34968416 PMCID: PMC8677487 DOI: 10.1016/j.celrep.2021.110214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/07/2021] [Accepted: 12/13/2021] [Indexed: 01/12/2023] Open
Abstract
T cell immunity is crucial for control of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and has been studied widely on a quantitative level. However, the quality of responses, in particular of CD8+ T cells, has only been investigated marginally so far. Here, we isolate T cell receptor (TCR) repertoires specific for immunodominant SARS-CoV-2 epitopes restricted to common human Leukocyte antigen (HLA) class I molecules in convalescent individuals. SARS-CoV-2-specific CD8+ T cells are detected up to 12 months after infection. TCR repertoires are diverse, with heterogeneous functional avidity and cytotoxicity toward virus-infected cells, as demonstrated for TCR-engineered T cells. High TCR functionality correlates with gene signatures that, remarkably, could be retrieved for each epitope:HLA combination analyzed. Overall, our data demonstrate that polyclonal and highly functional CD8+ TCRs—classic features of protective immunity—are recruited upon mild SARS-CoV-2 infection, providing tools to assess the quality of and potentially restore functional CD8+ T cell immunity.
Collapse
Affiliation(s)
- Karolin I Wagner
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Laura M Mateyka
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Sebastian Jarosch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Vincent Grass
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Simone Weber
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Kilian Schober
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany; Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Monika Hammel
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Teresa Burrell
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Behnam Kalali
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Holger Poppert
- Department of Neurology, Helios Klinikum München West, 81241 Munich, Germany; Neurologische Klinik, University Hospital Rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Henriette Beyer
- Department of Neurology, Helios Klinikum München West, 81241 Munich, Germany
| | - Sophia Schambeck
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany; Department of Neurology, Helios Klinikum München West, 81241 Munich, Germany
| | - Stefan Holdenrieder
- Institute of Laboratory Medicine, Munich Biomarker Research Center, Deutsches Herzzentrum München, Technical University of Munich (TUM), 80636 Munich, Germany
| | - Andrea Strötges-Achatz
- Institute of Laboratory Medicine, Munich Biomarker Research Center, Deutsches Herzzentrum München, Technical University of Munich (TUM), 80636 Munich, Germany
| | - Verena Haselmann
- Department of Clinical Chemistry, University Medicine Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany
| | - Michael Neumaier
- Department of Clinical Chemistry, University Medicine Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany
| | - Johanna Erber
- Department of Internal Medicine II, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Alina Priller
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Sarah Yazici
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Hedwig Roggendorf
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Marcus Odendahl
- Experimental Transfusion Medicine, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, 01307 Dresden, Germany
| | - Torsten Tonn
- Experimental Transfusion Medicine, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, 01307 Dresden, Germany
| | - Andrea Dick
- Laboratory of Immunogenetics and Molecular Diagnostics, Department of Transfusion Medicine, Cellular Therapeutics and Hemostaseology, LMU University Hospital, 81377 Munich, Germany
| | - Klaus Witter
- Laboratory of Immunogenetics and Molecular Diagnostics, Department of Transfusion Medicine, Cellular Therapeutics and Hemostaseology, LMU University Hospital, 81377 Munich, Germany
| | - Hrvoje Mijočević
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Percy A Knolle
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Andreas Pichlmair
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Claudia S Crowell
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Elvira D'Ippolito
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany.
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
2
|
Successful Use of Heterologous CMV-Reactive T Lymphocyte to Treat Severe Refractory Cytomegalovirus (CMV) Infection in a Liver Transplanted Patient: Correlation of the Host Antiviral Immune Reconstitution with CMV Viral Load and CMV miRNome. Microorganisms 2021; 9:microorganisms9040684. [PMID: 33810329 PMCID: PMC8066103 DOI: 10.3390/microorganisms9040684] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
Cytomegalovirus (CMV) infection is the most significant viral infection in hosts with compromised immune systems as solid organ transplant patients. Despite significant progress being made in the prevention of CMV disease in these patients, further therapeutic strategies for CMV disease and for the CMV reactivation prevention are needed. Here, we describe the outcome of the infusion of in vitro expanded CMV-reactive T-cells, taken from a healthy CMV-seropositive donor, in a liver-transplanted recipient with a refractory recurrent CMV. In this particular case, adoptive transfer of allogenic CMV-reactive T-lymphocytes resulted in the clearance of CMV infection and resolution of the pathological manifestations of the patient. In the study we also investigated circulating miRNAs, both cellular and viral, as potential biomarkers during the course of CMV infection. The results indicate that the infusion of allogenic CMV-reactive T-cells can be an effective strategy to treat CMV infection recurrence when the generation of autologous virus specific T cell clones is not possible.
Collapse
|
3
|
Gain C, Malik S, Bhattacharjee S, Ghosh A, Robertson ES, Das BB, Saha A. Proteasomal inhibition triggers viral oncoprotein degradation via autophagy-lysosomal pathway. PLoS Pathog 2020; 16:e1008105. [PMID: 32092124 PMCID: PMC7058366 DOI: 10.1371/journal.ppat.1008105] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 03/05/2020] [Accepted: 01/28/2020] [Indexed: 12/22/2022] Open
Abstract
Epstein-Barr virus (EBV) nuclear oncoprotein EBNA3C is essential for B-cell transformation and development of several B-cell lymphomas particularly those are generated in an immuno-compromised background. EBNA3C recruits ubiquitin-proteasome machinery for deregulating multiple cellular oncoproteins and tumor suppressor proteins. Although EBNA3C is found to be ubiquitinated at its N-terminal region and interacts with 20S proteasome, the viral protein is surprisingly stable in growing B-lymphocytes. EBNA3C can also circumvent autophagy-lysosomal mediated protein degradation and subsequent antigen presentation for T-cell recognition. Recently, we have shown that EBNA3C enhances autophagy, which serve as a prerequisite for B-cell survival particularly under growth deprivation conditions. We now demonstrate that proteasomal inhibition by MG132 induces EBNA3C degradation both in EBV transformed B-lymphocytes and ectopic-expression systems. Interestingly, MG132 treatment promotes degradation of two EBNA3 family oncoproteins-EBNA3A and EBNA3C, but not the viral tumor suppressor protein EBNA3B. EBNA3C degradation induced by proteasomal inhibition is partially blocked when autophagy-lysosomal pathway is inhibited. In response to proteasomal inhibition, EBNA3C is predominantly K63-linked polyubiquitinated, colocalized with the autophagy-lysosomal fraction in the cytoplasm and participated within p62-LC3B complex, which facilitates autophagy-mediated degradation. We further show that the degradation signal is present at the first 50 residues of the N-terminal region of EBNA3C. Proteasomal inhibition reduces the colony formation ability of this important viral oncoprotein, induces apoptotic cell death and increases transcriptional activation of both latent and lytic gene expression which further promotes viral reactivation from EBV transformed B-lymphocytes. Altogether, this study offers rationale to use proteasome inhibitors as potential therapeutic strategy against multiple EBV associated B-cell lymphomas, where EBNA3C is expressed.
Collapse
Affiliation(s)
- Chandrima Gain
- Department of Life Sciences, Presidency University, West Bengal, India
| | - Samaresh Malik
- Department of Life Sciences, Presidency University, West Bengal, India
| | | | - Arijit Ghosh
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, India
| | - Erle S. Robertson
- Department of Otorhinolaryngology Head and Neck Surgery, and the Tumor Virology Program, Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Benu Brata Das
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, India
| | - Abhik Saha
- Department of Life Sciences, Presidency University, West Bengal, India
| |
Collapse
|
4
|
Dugan JP, Coleman CB, Haverkos B. Opportunities to Target the Life Cycle of Epstein-Barr Virus (EBV) in EBV-Associated Lymphoproliferative Disorders. Front Oncol 2019; 9:127. [PMID: 30931253 PMCID: PMC6428703 DOI: 10.3389/fonc.2019.00127] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/13/2019] [Indexed: 12/29/2022] Open
Abstract
Many lymphoproliferative disorders (LPDs) are considered "EBV associated" based on detection of the virus in tumor tissue. EBV drives proliferation of LPDs via expression of the viral latent genes and many pre-clinical and clinical studies have shown EBV-associated LPDs can be treated by exploiting the viral life cycle. After a brief review of EBV virology and the natural life cycle within a host we will discuss the importance of the viral gene programs expressed during specific viral phases, as well as within immunocompetent vs. immunocompromised hosts and corresponding EBV-associated LPDs. We will then review established and emerging treatment approaches for EBV-associated LPDs based on EBV gene expression programs. Patients with EBV-associated LPDs can have a poor performance status, multiple comorbidities, and/or are immunocompromised from organ transplantation, autoimmune disease, or other congenital or acquired immunodeficiency making them poor candidates to receive intensive cytotoxic chemotherapy. With the emergence of EBV-directed therapy there is hope that we can devise more effective therapies that confer milder toxicity.
Collapse
Affiliation(s)
- James P. Dugan
- Division of Hematology, University of Colorado, Aurora, CO, United States
| | - Carrie B. Coleman
- Division of Immunology, University of Colorado, Aurora, CO, United States
| | - Bradley Haverkos
- Division of Hematology, University of Colorado, Aurora, CO, United States
| |
Collapse
|
5
|
NF-κB Signaling Regulates Expression of Epstein-Barr Virus BART MicroRNAs and Long Noncoding RNAs in Nasopharyngeal Carcinoma. J Virol 2016; 90:6475-88. [PMID: 27147748 PMCID: PMC4936125 DOI: 10.1128/jvi.00613-16] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/26/2016] [Indexed: 12/17/2022] Open
Abstract
Epstein-Barr virus (EBV) expresses few viral proteins in nasopharyngeal carcinoma (NPC) but high levels of BamHI-A rightward transcripts (BARTs), which include long noncoding RNAs (lncRNAs) and BART microRNAs (miRNAs). It is hypothesized that the mechanism for regulation of BARTs may relate to EBV pathogenesis in NPC. We showed that nuclear factor-κB (NF-κB) activates the BART promoters and modulates the expression of BARTs in EBV-infected NPC cells but that introduction of mutations into the putative NF-κB binding sites abolished activation of BART promoters by NF-κB. Binding of p50 subunits to NF-κB sites in the BART promoters was confirmed in electrophoretic mobility shift assays (EMSA) and further demonstrated in vivo using chromatin immunoprecipitation (ChIP) analysis. Expression of BART miRNAs and lncRNAs correlated with NF-κB activity in EBV-infected epithelial cells, while treatment of EBV-harboring NPC C666-1 cells with aspirin (acetylsalicylic acid [ASA]) and the IκB kinase inhibitor PS-1145 inhibited NF-κB activity, resulting in downregulation of BART expression. Expression of EBV LMP1 activates BART promoters, whereas an LMP1 mutant which cannot induce NF-κB activation does not activate BART promoters, further supporting the idea that expression of BARTs is regulated by NF-κB signaling. Expression of LMP1 is tightly regulated in NPC cells, and this study confirmed that miR-BART5-5p downregulates LMP1 expression, suggesting a feedback loop between BART miRNA and LMP1-mediated NF-κB activation in the NPC setting. These findings provide new insights into the mechanism underlying the deregulation of BARTs in NPC and identify a regulatory loop through which BARTs support EBV latency in NPC.
IMPORTANCE Nasopharyngeal carcinoma (NPC) cells are ubiquitously infected with Epstein-Barr virus (EBV). Notably, EBV expresses very few viral proteins in NPC cells, presumably to avoid triggering an immune response, but high levels of EBV BART miRNAs and lncRNAs which exhibit complex functions associated with EBV pathogenesis. The mechanism for regulation of BARTs is critical for understanding NPC oncogenesis. This study provides multiple lines of evidence to show that expression of BARTs is subject to regulation by NF-κB signaling. EBV LMP1 is a potent activator of NF-κB signaling, and we demonstrate that LMP1 can upregulate expression of BARTs through NF-κB signaling and that BART miRNAs are also able to downregulate LMP1 expression. It appears that aberrant NF-κB signaling and expression of BARTs form an autoregulatory loop for maintaining EBV latency in NPC cells. Further exploration of how targeting NF-κB signaling interrupts EBV latency in NPC cells may reveal new options for NPC treatment.
Collapse
|
6
|
Abstract
EBV expresses a number of viral noncoding RNAs (ncRNAs) during latent infection, many of which have known regulatory functions and can post-transcriptionally regulate viral and/or cellular gene expression. With recent advances in RNA sequencing technologies, the list of identified EBV ncRNAs continues to grow. EBV-encoded RNAs (EBERs) , the BamHI-A rightward transcripts (BARTs) , a small nucleolar RNA (snoRNA) , and viral microRNAs (miRNAs) are all expressed during EBV infection in a variety of cell types and tumors. Recently, additional novel EBV ncRNAs have been identified. Viral miRNAs, in particular, have been under extensive investigation since their initial identification over ten years ago. High-throughput studies to capture miRNA targets have revealed a number of miRNA-regulated viral and cellular transcripts that tie into important biological networks. Functions for many EBV ncRNAs are still unknown; however, roles for many EBV miRNAs in latency and in tumorigenesis have begun to emerge. Ongoing mechanistic studies to elucidate the functions of EBV ncRNAs should unravel additional roles for ncRNAs in the viral life cycle. In this chapter, we will discuss our current knowledge of the types of ncRNAs expressed by EBV, their potential roles in viral latency, and their potential involvement in viral pathogenesis.
Collapse
|
7
|
Kamperschroer C, Gosink MM, Kumpf SW, O'Donnell LM, Tartaro KR. The genomic sequence of lymphocryptovirus from cynomolgus macaque. Virology 2015; 488:28-36. [PMID: 26590795 DOI: 10.1016/j.virol.2015.10.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/27/2014] [Accepted: 10/27/2015] [Indexed: 12/12/2022]
Abstract
Lymphocryptoviruses such as Epstein-Barr virus (EBV) cause persistent infections in human and non-human primates, and suppression of the immune system can increase the risk of lymphocryptovirus (LCV)-associated tumor development in both human and non-human primates. To enable LCV infection as a non-clinical model to study effects of therapeutics on EBV immunity, we determined the genomic DNA sequence of the LCV from cynomolgus macaque, a species commonly used for non-clinical testing. Comparison to rhesus macaque LCV and human EBV sequences indicates that LCV from the cynomolgus macaque has the same genomic arrangement and a high degree of similarity in most genes, especially with rhesus macaque LCV. Genes showing lower similarity were those encoding proteins involved in latency and/or tumor promotion or immune evasion. The genomic sequence of LCV from cynomolgus macaque should aid the development of non-clinical tools for identifying therapeutics that impact LCV immunity and carry potential lymphoma risk.
Collapse
Affiliation(s)
- Cris Kamperschroer
- Drug Safety Research and Development, Pfizer Global Research and Development, Pfizer, Inc., Groton, CT, USA.
| | - Mark M Gosink
- Drug Safety Research and Development, Pfizer Global Research and Development, Pfizer, Inc., Groton, CT, USA
| | - Steven W Kumpf
- Drug Safety Research and Development, Pfizer Global Research and Development, Pfizer, Inc., Groton, CT, USA
| | - Lynn M O'Donnell
- Drug Safety Research and Development, Pfizer Global Research and Development, Pfizer, Inc., Groton, CT, USA
| | - Karrie R Tartaro
- Drug Safety Research and Development, Pfizer Global Research and Development, Pfizer, Inc., Groton, CT, USA
| |
Collapse
|
8
|
Yamamoto T, Iwatsuki K. Diversity of Epstein-Barr virus BamHI-A rightward transcripts and their expression patterns in lytic and latent infections. J Med Microbiol 2012; 61:1445-1453. [PMID: 22700548 DOI: 10.1099/jmm.0.044727-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epstein-Barr virus (EBV) BamHI-A rightward transcripts (BARTs; also designated complementary strand transcripts or CSTs) have been demonstrated to contain several splicing forms in EBV-infected cells. To date, however, little is known about the actual full-length splicing form and its functions. In the present study, we proved that six forms of BARTs were present in EBV-positive cell lines and various tissue specimens with different EBV infection patterns. Of the BART-encoded genes, mRNA of four major splicing forms, including BARF0, RPMS1, RPMS1A and A73, were expressed in all EBV-infected cells. On the other hand, mRNA of two minor splicing forms, RK-BARF0 and RB3, was rarely detected, or if at all, at very low expression levels. Both RPMS1A and RPMS1 mRNA was transcribed at higher levels in EBV-infected cells. In particular, RPMS1 mRNA was expressed abundantly in epithelial carcinoma cells, including gastric carcinoma and nasopharyngeal carcinoma, in association with a lytic infection signal, BZLF1 mRNA. The four major splicing forms were expressed much less in B-cell lines with an integrated EBV genome than in those with episomal EBV genomes. These data indicate that at least six splicing forms can be expressed by EBV-infected cells or tissues, although the expression patterns or levels differ for different infection states such as lytic and latent infections.
Collapse
Affiliation(s)
- Takenobu Yamamoto
- Department of Dermatology, Kawasaki Hospital, Kawasaki Medical School, 2-1-80 Nakasange, Kita-Ku, Okayama 700-8505, Japan.,Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Keiji Iwatsuki
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
9
|
Marquitz AR, Raab-Traub N. The role of miRNAs and EBV BARTs in NPC. Semin Cancer Biol 2011; 22:166-72. [PMID: 22178394 DOI: 10.1016/j.semcancer.2011.12.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 11/28/2011] [Accepted: 12/02/2011] [Indexed: 01/07/2023]
Abstract
The BamHI A rightward transcripts are a set of alternatively splicing transcripts produced by Epstein-Barr Virus that are highly expressed in nasopharyngeal carcinoma. These transcripts contain several open reading frames as well as precursors for twenty-two miRNAs. Although the putative proteins corresponding to these open reading frames have not been detected, several studies have identified properties that are interesting and potentially significant with respect to cellular transformation. The miRNAs, however, are very abundant in all nasopharyngeal carcinomas and several potentially significant functions have been identified for some of the miRNAs. This article will focus on the nature of this complicated set of transcripts and the evidence that they contribute to the development of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Aron R Marquitz
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | | |
Collapse
|
10
|
Lee JM, Kim H, Noh SH, Lee WY, Kim SJ, Park JH. Expression of Epstein-Barr Virus Gene and Clonality of Infiltrated T Lymphocytes in Epstein-Barr Virus-associated Gastric Carcinoma. Immune Netw 2011; 11:50-8. [PMID: 21494374 PMCID: PMC3072675 DOI: 10.4110/in.2011.11.1.50] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 01/06/2011] [Accepted: 01/10/2011] [Indexed: 12/13/2022] Open
Abstract
Background Epstein-Barr virus associated gastric lymphoepithelioma-like carcinoma (LELC) is characterized by the intensive infiltration of lymphoid cells, the presence of EBV, and the better prognosis over typical adenocarcinoma. Thus, it was assumable that viral latent proteins may be responsible for the recruitment of a certain T cell repertoire to EBV-associated gastric carcinoma. Methods To examine above possibility, EBV gene expression in gastric carcinoma tissues and usage of TCR among the tumor infiltrating lymphocytes were analyzed. Results EBV specific DNA and EBERs RNA were detected in 4 out of 30 patients. RT-PCR analysis revealed that all 4 of EBV-positive tumor tissues expressed EBNA1 mRNA and BARTs and LMP2a was detected only one sample out of 4. However, the EBNA2 and LMP-1 transcripts were not detected in these tissues. CD8+ T cells were the predominant population of infiltrating lymphocytes in the EBV-positive gastric carcinoma. According to spectra type analysis of infiltrating T cells, 10 predominant bands were detected by TCR Vβ CDR3 specific RT-PCR from 4 EBV-positive tumor tissues. Sequence analysis of these bands revealed oligoclonal expansion of T cells. Conclusion These findings suggest that clonally expanded T cells in vivo might be a population of cytotoxic T cells reactive to EBV-associated gastric carcinoma.
Collapse
Affiliation(s)
- Jae Myun Lee
- Department of Microbiology, Brain Korea Project 21 of Medical Sciences, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 120-752, Korea
| | | | | | | | | | | |
Collapse
|
11
|
Quantitative studies of Epstein-Barr virus-encoded microRNAs provide novel insights into their regulation. J Virol 2010; 85:996-1010. [PMID: 21068248 DOI: 10.1128/jvi.01528-10] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Epstein-Barr virus (EBV) has been shown to encode at least 40 microRNAs (miRNAs), an important class of molecules that negatively regulate the expression of many genes through posttranscriptional mechanisms. Here, we have used real-time PCR assays to quantify the levels of EBV-encoded BHRF1 and BART miRNAs in latently infected cells and in cells induced into the lytic cycle. During latency, BHRF1 miRNAs were seen only in cells with detectable Cp- and/or Wp-initiated EBNA transcripts, while the BART miRNAs were expressed in all forms of latent infection. Surprisingly, levels of different BART miRNAs were found to vary up to 50-fold within a cell line. However, this variation could not be explained by differential miRNA turnover, as all EBV miRNAs appeared to be remarkably stable. Following entry into the virus lytic cycle, miR-BHRF1-2 and -1-3 were rapidly induced, coincident with the onset of lytic BHRF1 transcripts, while miR-BHRF1-1 expression was delayed until 48 h and correlated with the appearance of Cp/Wp-initiated EBNA transcripts. In contrast, levels of BART miRNAs were relatively unchanged during virus replication, despite dramatic increases in BART transcription. Finally, we show that BHRF1 and BART miRNAs were delayed relative to the induction of BHRF1 and BART transcripts in freshly infected primary B cell cultures. In summary, our data show that changes in BHRF1 and BART transcription are not necessarily reflected in altered miRNA levels, suggesting that miRNA maturation is a key step in regulating steady-state levels of EBV miRNAs.
Collapse
|
12
|
Al-Mozaini M, Bodelon G, Karstegl CE, Jin B, Al-Ahdal M, Farrell PJ. Epstein-Barr virus BART gene expression. J Gen Virol 2009; 90:307-316. [PMID: 19141439 DOI: 10.1099/vir.0.006551-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Introns from the Epstein-Barr virus (EBV) BART RNAs produce up to 20 micro RNAs (miRNAs) but the spliced exons of the BART RNAs have also been investigated as possible mRNAs, with the potential to express the RPMS1 and A73 proteins. Recombinant RPMS1 and A73 proteins were expressed in Escherichia coli and used to make new monoclonal antibodies that reacted specifically with artificially expressed RPMS1 and A73. These antibodies did not detect endogenous expression of A73 and RPMS1 proteins in a panel of EBV-infected cell lines representing the different known types of EBV infection. BART RNA could not be detected on Northern blots of cytoplasmic poly(A)(+) RNA from the C666.1 NPC cell line and BART RNA was found to be mainly in the nucleus of C666.1 cells, arguing against an mRNA role for BART RNAs. In contrast, some early lytic cycle EBV mRNAs were found to be expressed in C666.1 cells. Artificially expressed A73 protein was known to be able to bind to the cellular RACK1 protein and has now also been shown to be able to regulate calcium flux, presumably via RACK1. Overall, the results support the conclusion that the miRNAs are functionally important products of BART transcription in the cell lines studied because the A73 and RPMS1 proteins could not be detected in natural EBV infections. However, the possibility remains that A73 and RPMS1 might be expressed in some situations because of the clear potential relevance of their biochemical functions.
Collapse
Affiliation(s)
- Maha Al-Mozaini
- Molecular Virology Department, BMR King Faisal Hospital and Research Center, PO Box 3354 MBC-03, Riyadh 11211, Saudi Arabia.,Department of Virology, Imperial College London, Norfolk Place, London W2 1PG, UK
| | - Gustavo Bodelon
- Department of Virology, Imperial College London, Norfolk Place, London W2 1PG, UK
| | | | - Boquan Jin
- Department of Immunology, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Mohammed Al-Ahdal
- Molecular Virology Department, BMR King Faisal Hospital and Research Center, PO Box 3354 MBC-03, Riyadh 11211, Saudi Arabia
| | - Paul J Farrell
- Department of Virology, Imperial College London, Norfolk Place, London W2 1PG, UK
| |
Collapse
|
13
|
Epstein-Barr virus BART microRNAs are produced from a large intron prior to splicing. J Virol 2008; 82:9094-106. [PMID: 18614630 DOI: 10.1128/jvi.00785-08] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Latent Epstein-Barr virus (EBV) infection is associated with several lymphoproliferative disorders, including posttransplant lymphoma, Hodgkin's disease, and Burkitt's lymphoma, as well as nasopharyngeal carcinoma (NPC). Twenty-nine microRNAs (miRNAs) have been identified that are transcribed during latent infection from three clusters in the EBV genome. Two of the three clusters of miRNAs are made from the BamHI A rightward transcripts (BARTs), a set of alternatively spliced transcripts that are highly abundant in NPC but have not been shown to produce a detectable protein. This study indicates that while the BART miRNAs are located in the first four introns of the transcripts, processing of the pre-miRNAs from the primary transcript occurs prior to completion of the splicing reaction. Additionally, production of the BART miRNAs correlates with accumulation of a spliced mRNA in which exon 1 is joined directly to exon 3, suggesting that this form of the transcript may favor production of miRNAs. Sequence variations and processing of pre-miRNAs to the mature form also may account for various differences in miRNA abundance. Importantly, residual intronic pieces that result from processing of the pre-miRNAs were detected in the nucleus. The predicted structures of these pieces suggest there is a bias or temporal pattern to the production of the individual pre-miRNAs. These findings indicate that multiple factors contribute to the production of the BART miRNAs and to the apparent differences in abundance between the individual miRNAs of the cluster.
Collapse
|
14
|
Lin JH, Tsai CH, Chu JS, Chen JY, Takada K, Shew JY. Dysregulation of HER2/HER3 signaling axis in Epstein-Barr virus-infected breast carcinoma cells. J Virol 2007; 81:5705-13. [PMID: 17376931 PMCID: PMC1900270 DOI: 10.1128/jvi.00076-07] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The role of Epstein-Barr virus (EBV) in the pathogenesis of breast cancer has been of long-standing interest to the field. Breast epithelial cells can be infected by EBV through direct contact with EBV-bearing lymphoblastoid cells, and EBV infection has recently been shown to confer breast cancer cells an increased resistance to chemotherapeutic drugs. In this study, we established EBV-infected breast cancer MCF7 and BT474 cells and demonstrated that EBV infection promotes tumorigenic activity of breast cancer cells. Firstly, we showed that the EBV-infected MCF7-A and BT474-A cells exhibited increased anchorage-independent growth in soft agar. The increased colony formation capacity in soft agar was associated with increased expression and activation of HER2/HER3 signaling cascades, as evidenced by the findings that the treatment of HER2 antibody trastuzumab (Herceptin), phosphatidylinositol 3-kinase inhibitor, or MEK inhibitor completely abolished the tumorigenic capacity. In the EBV-infected breast cancer cells, the expression of EBV latency genes including EBNA1, EBER1, and BARF0 was detected. We next showed that BARF0 alone was sufficient to efficiently up-regulate HER2/HER3 expression and promoted tumorigenic activity in MCF7 and BT474 cells by the use of both overexpression and small interfering RNA knock-down. Collectively, we demonstrated that EBV-encoded BARF0 promotes the tumorigenic activity of breast cancer cells through activation of HER2/HER3 signaling cascades.
Collapse
Affiliation(s)
- Jiun-Han Lin
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | | | | | | | | | | |
Collapse
|
15
|
Steven N. The potential of adoptive transfer of immunity for reducing post transplant Epstein-Barr virus-associated disease. Curr Opin Infect Dis 2006; 12:585-91. [PMID: 17035825 DOI: 10.1097/00001432-199912000-00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- N Steven
- The University of Birmingham, CRC Institute for Cancer Studies, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
16
|
Dutta N, Gupta A, Mazumder DNG, Banerjee S. Down-regulation of locus-specific human lymphocyte antigen class I expression in Epstein-Barr virus-associated gastric cancer: implication for viral-induced immune evasion. Cancer 2006; 106:1685-93. [PMID: 16541432 DOI: 10.1002/cncr.21784] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND To understand whether the association between Epstein-Barr Virus (EBV) and gastric cancer (GC) has any role in loss of surface expression of human lymphocyte antigen (HLA) class I, the authors analyzed locus-specific transcriptional expression of HLA-A, HLA-B, HLA-C, and HLA-E along with other HLA-associated molecules (beta2-microglobulin [beta2M], cellular latent membrane protein [LMP], and transporter associated with antigen presentation [TAP]) in EBV-associated, primary GC (EBVaGC) and EBV-negative GC (EBVnGC) tissues. METHODS Approximately 20 EBVaGC tissues and 40 EBVnGC tissues and their corresponding normal tissues were used in the study. The presence of EBV in GC was established by EBV-encoded small RNA in situ hybridization analysis and BamHI W polymerase chain reaction (PCR) analysis. Transcriptional expression of viral LMP2A and several HLA class I genes were analyzed by reverse transcriptase (RT)-PCR. Surface expression levels of HLA class I proteins in cancer samples along with their normal counterparts also were quantified by flow cytometry. RESULTS The RT-PCR data suggested selective down-regulation of the HLA-A/HLA-B locus along with over-expression of HLA-E transcripts in EBVaGC (P < .05). This was confirmed further by the flow-cytometric studies using antibodies to HLA-ABC and HLA-E. Among the accessory molecules, LMP7 transcript was down-regulated in a number of EBVaGC tissues compared with EBVnGC. CONCLUSIONS The current results suggested that the establishment of EBV latent infection in gastric tissues allows malignant cells to avoid the immune surveillance of both cytotoxic T-lymphocytes and natural killer cells by regulating the differential expression of HLA class I molecules.
Collapse
Affiliation(s)
- Nirmal Dutta
- Biophysics Division, Saha Institute of Nuclear Physics, Kolkata, India
| | | | | | | |
Collapse
|
17
|
Bihl FK, Loggi E, Chisholm JV, Hewitt HS, Henry LM, Linde C, Suscovich TJ, Wong JT, Frahm N, Andreone P, Brander C. Simultaneous assessment of cytotoxic T lymphocyte responses against multiple viral infections by combined usage of optimal epitope matrices, anti- CD3 mAb T-cell expansion and "RecycleSpot". J Transl Med 2005; 3:20. [PMID: 15888204 PMCID: PMC1164435 DOI: 10.1186/1479-5876-3-20] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2005] [Accepted: 05/11/2005] [Indexed: 12/23/2022] Open
Abstract
The assessment of cellular anti-viral immunity is often hampered by the limited availability of adequate samples, especially when attempting simultaneous, high-resolution determination of T cell responses against multiple viral infections. Thus, the development of assay systems, which optimize cell usage, while still allowing for the detailed determination of breadth and magnitude of virus-specific cytotoxic T lymphocyte (CTL) responses, is urgently needed. This study provides an up-to-date listing of currently known, well-defined viral CTL epitopes for HIV, EBV, CMV, HCV and HBV and describes an approach that overcomes some of the above limitations through the use of peptide matrices of optimally defined viral CTL epitopes in combination with anti-CD3 in vitro T cell expansion and re-use of cells from negative ELISpot wells. The data show that, when compared to direct ex vivo cell preparations, antigen-unspecific in vitro T cell expansion maintains the breadth of detectable T cell responses and demonstrates that harvesting cells from negative ELISpot wells for re-use in subsequent ELISpot assays (RecycleSpot), further maximized the use of available cells. Furthermore when combining T cell expansion and RecycleSpot with the use of rationally designed peptide matrices, antiviral immunity against more than 400 different CTL epitopes from five different viruses can be reproducibly assessed from samples of less than 10 milliliters of blood without compromising information on the breadth and magnitude of these responses. Together, these data support an approach that facilitates the assessment of cellular immunity against multiple viral co-infections in settings where sample availability is severely limited.
Collapse
Affiliation(s)
- Florian K Bihl
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Elisabetta Loggi
- Dipartimento di Cardioangiologia ed Epatologia, Ospedale S. Orsola-Malpighi, Università degli Studi di Bologna, Italy
| | - John V Chisholm
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Hannah S Hewitt
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Leah M Henry
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Caitlyn Linde
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Todd J Suscovich
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Johnson T Wong
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Nicole Frahm
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Pietro Andreone
- Dipartimento di Cardioangiologia ed Epatologia, Ospedale S. Orsola-Malpighi, Università degli Studi di Bologna, Italy
| | - Christian Brander
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
18
|
Chen H, Huang J, Wu FY, Liao G, Hutt-Fletcher L, Hayward SD. Regulation of expression of the Epstein-Barr virus BamHI-A rightward transcripts. J Virol 2005; 79:1724-33. [PMID: 15650197 PMCID: PMC544122 DOI: 10.1128/jvi.79.3.1724-1733.2005] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2004] [Accepted: 09/09/2004] [Indexed: 11/20/2022] Open
Abstract
The Epstein-Barr virus (EBV) BamHI-A rightward transcripts, or BARTs, are a family of mRNAs expressed in all EBV latency programs, including EBV-infected B cells in healthy carriers. Despite their ubiquitous expression, the regulation and biological function of BARTs are still unclear. In this study, the BART 5' termini were characterized by using a procedure that selects capped, full-length mRNAs. Two TATA-less promoter regions, designated P1 and P2, were mapped. P1 had relatively high basal activity in both epithelial and B cells, whereas P2 exhibited higher activity in epithelial cells. Upon EBV infection of B cells, transcription from P1 was detected soon after infection, while expression from P2 was delayed. Promoter-reporter assays in transiently transfected cells revealed that P1 and P2 were differentially regulated. Interferon regulatory factor 7 (IRF7) and IRF5 negatively regulated P1 activity. c-Myc and C/EBP family members positively regulated P2. Regulation of P2 by C/EBPs was characterized by electrophoretic mobility shift assay, chromatin immunoprecipitation, and reporter assays. More-abundant BART expression in epithelial cells correlated with the relative expression of positive and negative regulators in these cells.
Collapse
Affiliation(s)
- Honglin Chen
- Department of Microbiology, The University of Hong Kong, Hong Kong
| | | | | | | | | | | |
Collapse
|
19
|
Thornburg NJ, Kusano S, Raab-Traub N. Identification of Epstein-Barr virus RK-BARF0-interacting proteins and characterization of expression pattern. J Virol 2004; 78:12848-56. [PMID: 15542637 PMCID: PMC525031 DOI: 10.1128/jvi.78.23.12848-12856.2004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Epstein-Barr virus (EBV) BamHI A transcripts are a family of transcripts that are differentially spliced and can be detected in multiple EBV-associated malignancies. Several of the transcripts may encode proteins. One transcript of interest, RK-BARF0, is proposed to encode a 279-amino-acid protein with a possible endoplasmic reticulum-targeting sequence. In this study, the properties of RK-BARF0 were examined through identification of cellular-interacting proteins through yeast two-hybrid analysis and characterization of its expression in EBV-infected cells and tumors. In addition to the interaction previously identified with cellular Notch, it was determined that RK-BARF0 also bound cellular human I-mfa domain-containing protein (HIC), epithelin, and scramblase. An interaction between RK-BARF0 and Notch or epithelin induced proteasome-dependent degradation of Notch and epithelin but not of HIC or scramblase. Low levels of endogenous Notch expression in EBV-positive cell lines may correlate with RK-BARF0 expression. However, a screen of EBV-positive cell lines and tumors with an affinity-purified alpha-RK-BARF0 antiserum did not consistently detect RK-BARF0. These data suggest that while RK-BARF0 may have important cellular functions during EBV infection, and while the phenotype of EBV-positive cells suggest its expression, RK-BARF0 levels may be too low to detect.
Collapse
Affiliation(s)
- Natalie J Thornburg
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | | | | |
Collapse
|
20
|
van Beek J, Brink AATP, Vervoort MBHJ, van Zijp MJM, Meijer CJLM, van den Brule AJC, Middeldorp JM. In vivo transcription of the Epstein–Barr virus (EBV) BamHI-A region without associated in vivo BARF0 protein expression in multiple EBV-associated disorders. J Gen Virol 2003; 84:2647-2659. [PMID: 13679598 DOI: 10.1099/vir.0.19196-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The in vivo expression of the Epstein–Barr virus (EBV) BamHI-A rightward transcripts (BARTs) as well as the putative BART-encoded BARF0 and RK-BARF0 proteins in various EBV-associated malignancies was investigated. RT-PCRs specific for the different splice variants of the BARTs and both a nucleic acid sequence-based amplification assay and an RT-PCR specific for the BARF0 ORF were used. Abundant transcription of BARTs was found in EBV-associated Hodgkin's lymphomas, Burkitt's lymphomas (BL), T-cell non-Hodgkin's lymphomas, post-transplant lymphoproliferative disorders, AIDS-related lymphomas and gastric carcinomas. Using RNA in situ hybridization (RISH), BARTs were detected within the neoplastic cells of these malignancies. BARTs encoding RK-BARF0 were not detected. The BARTs detected were shown possibly to encode the RPMS1 and BARF0 proteins, based on their splicing. However, BARTs actually harbouring the BARF0 ORF were detected only in specimens containing a relatively large number of EBV-positive cells. New monoclonal antibodies against the BARF0 protein were generated that efficiently recognized prokaryotic and eukaryotic recombinant BARF0. However, the BARF0 protein was not detected in clinical samples, nor in EBV-positive cell lines, even though these were positive for BARTs by RISH and/or BARF0 RNA in vitro analysis. Using immunoblot analysis, no antibodies against baculovirus-expressed BARF0 protein were detected in the sera of nasopharyngeal carcinoma patients, BL patients and Hodgkin's disease patients, patients with chronic EBV infection, infectious mononucleosis patients or EBV-positive healthy donors. Thus, BARTs containing the BARF0 ORF are expressed in vivo but the BARF0 protein cannot be detected and may be expressed only marginally. It is concluded that the BARF0 protein is unlikely to play a role in vivo in EBV-positive malignancies.
Collapse
Affiliation(s)
- Josine van Beek
- Department of Pathology, Vrije Universiteit Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Antoinette A T P Brink
- Department of Pathology, Vrije Universiteit Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Marcel B H J Vervoort
- Department of Pathology, Vrije Universiteit Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Mireille J M van Zijp
- Department of Pathology, Vrije Universiteit Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Chris J L M Meijer
- Department of Pathology, Vrije Universiteit Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Adriaan J C van den Brule
- Department of Pathology, Vrije Universiteit Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Jaap M Middeldorp
- Department of Pathology, Vrije Universiteit Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| |
Collapse
|
21
|
de Jesus O, Smith PR, Spender LC, Elgueta Karstegl C, Niller HH, Huang D, Farrell PJ. Updated Epstein-Barr virus (EBV) DNA sequence and analysis of a promoter for the BART (CST, BARF0) RNAs of EBV. J Gen Virol 2003; 84:1443-1450. [PMID: 12771413 DOI: 10.1099/vir.0.19054-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Two sequences required for activity of the Epstein-Barr virus BART RNA promoter in transfection assays have been identified by site-directed mutagenesis. One contains a consensus AP-1 site; the other has some similarity to Ets and Stat consensus binding sites. Candidate sequences were suggested by mapping a region of unmethylated DNA in EBV around the BART promoter followed by in vivo footprinting the promoter in the C666-1 nasopharyngeal carcinoma cell line, which expresses BART RNAs. The data are presented in the context of a revised EBV DNA sequence, known as EBV wt, that is proposed as a future standard sequence for EBV.
Collapse
Affiliation(s)
- Orlando de Jesus
- Ludwig Institute for Cancer Research, Faculty of Medicine, Imperial College, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| | - Paul R Smith
- Ludwig Institute for Cancer Research, Faculty of Medicine, Imperial College, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| | - Lindsay C Spender
- Ludwig Institute for Cancer Research, Faculty of Medicine, Imperial College, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| | - Claudio Elgueta Karstegl
- Ludwig Institute for Cancer Research, Faculty of Medicine, Imperial College, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| | - Hans Helmut Niller
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Regensburg, D-93053 Regensburg, Germany
| | - Dolly Huang
- Department of Clinical Oncology, Sir Y.K. Pao Centre for Cancer, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Paul J Farrell
- Ludwig Institute for Cancer Research, Faculty of Medicine, Imperial College, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| |
Collapse
|
22
|
Krüger S, Schroers R, Rooney CM, Gahn B, Chen SY. Identification of a naturally processed HLA-DR-restricted T-helper epitope in Epstein-Barr virus nuclear antigen type 1. J Immunother 2003; 26:212-21. [PMID: 12806275 DOI: 10.1097/00002371-200305000-00005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Epstein-Barr virus nuclear antigen type 1 (EBNA1), the only viral protein that is unequivocally expressed in all Epstein-Barr virus (EBV)-associated malignant diseases, is essential for viral DNA replication and maintenance of the viral episome in infected cells. A glycine-alanine repeat domain inhibits antigen processing through the ubiquitin-proteasome pathway for presentation on human leukocyte antigen (HLA) class I molecules. EBNA1 is not protected from the HLA class II processing pathway, and CD4+ HLA class II-restricted T cells recognize the antigen. CD4+ T-helper (Th) cells play critical roles in initiating, regulating, and maintaining immune responses against viral infections and tumors, so that inclusion of EBNA1 as a target antigen may improve immunotherapy for EBV-associated cancers. In this study, the authors used the TEPITOPE software program to predict promiscuous class II epitope candidates. After several HLA-DR-restricted peptides were identified by in vitro analysis of the T-cell response to synthetic peptides, a T-cell clone was established that was specific for one of the peptides. Functional studies were performed with this clone. The CD4+ T helper cells specific for the HLA-DR15-restricted peptide EBNA1(482) (AEGLRALLARSHVER) recognized naturally processed EBNA1 protein. This epitope was presented by several HLA-DR alleles, including DR4, DR7, and DR11. The inclusion of the promiscuous, naturally processed EBNA1(482) epitope in vaccine constructs could enhance immune responses against EBV-positive cancers.
Collapse
Affiliation(s)
- Stefan Krüger
- Center for Cell and Gene Therapy, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
23
|
Gottschalk S, Edwards OL, Sili U, Huls MH, Goltsova T, Davis AR, Heslop HE, Rooney CM. Generating CTLs against the subdominant Epstein-Barr virus LMP1 antigen for the adoptive immunotherapy of EBV-associated malignancies. Blood 2003; 101:1905-12. [PMID: 12411306 DOI: 10.1182/blood-2002-05-1514] [Citation(s) in RCA: 149] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Epstein-Barr virus (EBV)-encoded LMP1 protein is expressed in EBV-positive Hodgkin disease and is a potential target for cytotoxic T-lymphocyte (CTL) therapy. However, the LMP1-specific CTL frequency is low, and so far the generation of LMP1-specific CTLs has required T-cell cloning. The toxicity of LMP1 has prevented the use of dendritic cells (DCs) for CTL stimulation, and we reasoned that an inactive, nontoxic LMP1 mutant (DeltaLMP1) could be expressed in DCs and would enable the activation and expansion of polyclonal LMP1-specific CTLs. Recombinant adenoviral vectors expressing LMP1 or DeltaLMP1 were tested for their ability to transduce DCs. LMP1 expression was toxic within 48 hours whereas high levels of DeltaLMP1 expression were achieved with minimal toxicity. DeltaLMP1-expressing DCs were able to reactivate and expand LMP1-specific CTLs from 3 healthy EBV-seropositive donors. LMP1-specific T cells were detected by interferon-gamma (IFN-gamma) enzyme-linked immunospot assay (ELISPOT) assays using the HLA-A2-restricted LMP1 peptide, YLQQNWWTL (YLQ). YLQ-specific T cells were undetectable (less than 0.001%) in donor peripheral blood mononuclear cells (PBMCs); however, after stimulation the frequency increased to 0.5% to 3.8%. Lysis of autologous target cells by CTLs was dependent on the level of LMP1 expression. In contrast, the frequency of YLQ-specific CTLs in EBV-specific CTLs reactivated and expanded using lymphoblastoid cell lines was low and no LMP1-specific cytotoxic activity was observed. Thus, DeltaLMP1 expression in DCs is nontoxic and enables the generation of LMP1-specific CTLs for future adoptive immunotherapy protocols for patients with LMP1-positive malignancies such as EBV-positive Hodgkin disease. Targeting LMP1 in these malignancies may improve the efficacy of current adoptive immunotherapy approaches.
Collapse
MESH Headings
- Adenoviruses, Human/genetics
- Antigen Presentation
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Epitopes/immunology
- Epstein-Barr Virus Infections/therapy
- Epstein-Barr Virus Nuclear Antigens/genetics
- Genetic Vectors/genetics
- HLA-A2 Antigen/immunology
- Hodgkin Disease/therapy
- Hodgkin Disease/virology
- Humans
- Immunotherapy, Adoptive
- Interferon-gamma/metabolism
- Lymphocyte Activation
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Transduction, Genetic
- Tumor Virus Infections/therapy
- Vaccinia virus/genetics
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/immunology
- Viral Matrix Proteins/toxicity
Collapse
Affiliation(s)
- Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children's Cancer Center, Departments of Pediatrics, Medicine, and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Middeldorp JM, Brink AATP, van den Brule AJC, Meijer CJLM. Pathogenic roles for Epstein-Barr virus (EBV) gene products in EBV-associated proliferative disorders. Crit Rev Oncol Hematol 2003; 45:1-36. [PMID: 12482570 DOI: 10.1016/s1040-8428(02)00078-1] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Epstein-Barr virus (EBV) is associated with a still growing spectrum of clinical disorders, ranging from acute and chronic inflammatory diseases to lymphoid and epithelial malignancies. Based on a combination of in vitro and in vivo findings, EBV is thought to contribute in the pathogenesis of these diseases. The different EBV gene expression patterns in the various disorders, suggest different EBV-mediated pathogenic mechanisms. In the following pages, an overview of the biology of EBV-infection is given and functional aspects of EBV-proteins are discussed and their putative role in the various EBV-associated disorders is described. EBV gene expression patterns and possible pathogenic mechanisms are discussed. In addition, expression of the cellular genes upregulated by EBV in vitro is discussed, and a comparison with the in vivo situation is made.
Collapse
Affiliation(s)
- Jaap M Middeldorp
- Department of Pathology, Vrije Universiteit Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
25
|
Matsumura S, Kita H, He XS, Ansari AA, Lian ZX, Van De Water J, Yamamoto K, Tsuji T, Coppel RL, Kaplan M, Gershwin ME. Comprehensive mapping of HLA-A0201-restricted CD8 T-cell epitopes on PDC-E2 in primary biliary cirrhosis. Hepatology 2002; 36:1125-34. [PMID: 12395322 DOI: 10.1053/jhep.2002.36161] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Growing evidence has implicated the involvement of autoreactive T lymphocytes in the pathogenesis of primary biliary cirrhosis (PBC). We have recently taken advantage of motif prediction analysis of HLA-A*0201 and identified the first major histocompatibility complex (MHC) class I restricted epitope, amino acids 159 to 167 on E2 components of pyruvate dehydrogenase complexes (PDC-E2), the major mitochondrial antigens in PBC. The mechanisms involved in the selection of epitope peptide(s) that comprise the PDC-E2-specific autoreactive cytotoxic T lymphocytes (CTLs) are unknown and likely involve other epitopes on PDC-E2 restricted by MHC class I molecules. To address this issue, a comprehensive mapping of the CTL epitope repertoire on the PDC-E2 molecule that binds HLA-A*0201 was performed to provide further clues regarding the role of CTLs. We used the T2 cell line to screen 79 overlapping 15mer peptides, spanning the entire PDC-E2 molecule. Six of the 79 peptides exhibited significantly higher binding activity to HLA-A*0201 than the other 15mer peptides. Two of these 6 peptides induced CTL lines from patients with PBC. Fine mapping with N-terminus or C-terminus truncated peptides identified 10mer peptide, PDC-E2 amino acids 165 to 174, which is a novel CD8 epitope restricted by HLA-A*0201. In conclusion, using a combination of the 15mer peptide library screening with the T2 binding assay and also the induction of CTL lines with candidate peptides, we have defined a novel HLA-A*0201-restricted epitope PDC-E2 165 to 174 in patients with PBC. These data will become important in the development of altered peptide ligands to modulate disease activity.
Collapse
Affiliation(s)
- Shuji Matsumura
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California at Davis, 95616, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Gottschalk S, Heslop HE, Roon CM. Treatment of Epstein-Barr virus-associated malignancies with specific T cells. Adv Cancer Res 2002; 84:175-201. [PMID: 11883527 DOI: 10.1016/s0065-230x(02)84006-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Latent Epstein-Barr virus (EBV) infection is associated with a heterogeneous group of malignancies, including Burkitt's lymphoma, Hodgkin's disease, nasopharyngeal carcinoma, and lymphoproliferative disease (LPD). The development of adoptive immunotherapies for these malignancies is being fueled by the successful generation of allogeneic donor derived EBV-specific cytotoxic T cells (CTL) for the prevention and treatment of EBV-LPD after hematopoietic stem cell transplantation. This approach is being extended to EBV-LPD after solid organ transplantation by use of autologous and haploidentical EBV-specific CTL. For other EBV-associated malignancies, there is only limited clinical experience with EBV-specific CTL. With few exceptions, only patients with recurrent Hodgkin's disease have been treated with autologous EBV-specific CTL, and although the results have been promising, they do not include cures. Lack of CTL efficacy may reflect either down-regulation of immunodominant EBV proteins, which are major CTL targets, or the presence of inhibitory cytokines. Further improvement of EBV-specific CTL therapy for Hodgkin's disease will require improved methods to activate and expand CTL specific for the latent EBV genes expressed in Hodgkin's disease and to genetically modify the expanded CTL to render them resistant to inhibitory cytokines. If effective, such strategies could be applied not only to other EBV-associated malignancies, but also to a broad range of human tumors with defined tumor antigens and similar immune evasion strategies.
Collapse
Affiliation(s)
- Stephen Gottschalk
- Center for Cell and Gene Therapy, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | | | | |
Collapse
|
27
|
Abstract
Epstein-Barr virus (EBV) encodes a family of related transcripts, the complementary strand transcripts (CSTs) or BARTs (Bam A rightward transcripts). These are present in all types of EBV infection but are expressed to particularly high levels in nasopharyngeal carcinomas. Although convincing demonstration of protein expression from these transcripts is still subject to some debate, potential proteins encoded by them have been shown to modify Notch signalling pathways.
Collapse
MESH Headings
- Gene Expression Regulation, Viral
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/physiology
- Humans
- Membrane Proteins/metabolism
- Nasopharyngeal Neoplasms/virology
- Neoplasm Proteins
- Nucleic Acid Conformation
- Open Reading Frames/genetics
- RNA, Complementary/chemistry
- RNA, Complementary/genetics
- RNA, Viral/chemistry
- RNA, Viral/genetics
- RNA, Viral/metabolism
- Receptors, Notch
- Transcription, Genetic/genetics
- Viral Proteins/genetics
- Viral Proteins/metabolism
Collapse
Affiliation(s)
- P Smith
- Institute for Cancer Genetics and Pharmacogenomics, Department of Biology, Brunel University, Uxbridge, UB8 3PH, UK.
| |
Collapse
|
28
|
Catalina MD, Sullivan JL, Bak KR, Luzuriaga K. Differential evolution and stability of epitope-specific CD8(+) T cell responses in EBV infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4450-7. [PMID: 11591771 DOI: 10.4049/jimmunol.167.8.4450] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Murine models of lymphocytic choriomeningitis virus infection suggest that the memory CD8(+) T cell repertoire is reflective of the CD8(+) T cell repertoire generated during acute infection. Less is known regarding the evolution of CD8(+) T cell repertoires during human viral infections. We therefore examined epitope-specific CD8(+) T cell responses in a large cohort of individuals with acute through latent Epstein-Barr virus infection. Using 16 of 20 published EBV epitopes restricted by HLA-A2, HLA-A3 or HLA-B7, we showed that lytic cycle-specific CD8(+) T cell responses predominated during acute EBV infection. However, whereas HLA-A2(+)-restricted BMLF-1-specific CD8(+) T cell responses were maintained through latency, HLA-A2(+)- and HLA-B7(+)-restricted BZLF-1, as well as HLA-A3(+)-restricted BRLF-1 CD8(+) T cell responses, were generated but not readily maintained. Analyses of CD8(+) T cell responses to EBV latent cycle Ags showed delayed detection and lower frequencies of latent epitope-specific CD8(+) T cell responses during acute EBV infection, with maintenance of these responses 1 yr post-EBV infection. Early BMLF-1 and EBNA-3A epitope-specific CD8(+) T cell frequencies did not correlate with their frequencies at 1 yr postinfection. Interestingly, populations of EBV-specific CD8(+) T cells were stable during 20 mo in our long term EBV-seropositive populations, suggesting homeostasis between virus and the host immune system. This study demonstrates that CD8(+) T cell repertoires generated during persistent viral infections are not simply reflective of the initial pool of CD8(+) T cells and provides evidence that the generation of CD8(+) T cell responses to a persistent infection is a dynamic process.
Collapse
Affiliation(s)
- M D Catalina
- Department of Pediatrics and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
29
|
Khanna R, Tellam J, Duraiswamy J, Cooper L. Immunotherapeutic strategies for EBV-associated malignancies. Trends Mol Med 2001; 7:270-6. [PMID: 11378517 DOI: 10.1016/s1471-4914(01)02002-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Advances in our understanding of the role of cytotoxic T lymphocytes (CTLs) in the control of Epstein-Barr virus (EBV)-associated malignancies and the overall biology of these diseases have led to the development of novel therapeutic strategies designed to specifically target viral antigens expressed in these malignancies. Long-term success of many of these strategies is constrained by the latency phenotypes adopted by different diseases. Adoptive transfer of polyclonal virus-specific CTLs has been used successfully to reverse the outgrowth of malignancies such as post-transplant lymphoproliferative disease (PTLD). On the other hand, limited viral gene expression in other EBV-associated malignancies such as Burkitt's lymphoma, Hodgkin's disease and nasopharyngeal carcinoma limits the efficacy of immunotherapeutic strategies used for PTLD. Preclinical studies based on specific targeting of viral antigens expressed in these malignancies have provided very encouraging results and thus are likely to serve as an important platform for the treatment of human patients.
Collapse
Affiliation(s)
- R Khanna
- Tumour Immunology Laboratory, Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research, Bancroft Centre, 300 Herston Road, Herston (Qld) 4006, Brisbane, Australia.
| | | | | | | |
Collapse
|
30
|
Zhang J, Chen H, Weinmaster G, Hayward SD. Epstein-Barr virus BamHi-a rightward transcript-encoded RPMS protein interacts with the CBF1-associated corepressor CIR to negatively regulate the activity of EBNA2 and NotchIC. J Virol 2001; 75:2946-56. [PMID: 11222720 PMCID: PMC115921 DOI: 10.1128/jvi.75.6.2946-2956.2001] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2000] [Accepted: 12/22/2000] [Indexed: 11/20/2022] Open
Abstract
The Epstein-Barr virus (EBV) BamHI-A rightward transcripts (BARTs) are expressed in all EBV-associated tumors as well as in latently infected B cells in vivo and cultured B-cell lines. One of the BART family transcripts contains an open reading frame, RPMS1, that encodes a nuclear protein termed RPMS. Reverse transcription-PCR analysis revealed that BART transcripts with the splicing pattern that generates the RPMS1 open reading frame are commonly expressed in EBV-positive lymphoblastoid cell lines and are also detected in Hodgkin's disease tissues. Experiments undertaken to determine the function of RPMS revealed that RPMS interacts with both CBF1 and components of the CBF1-associated corepressor complex. RPMS interaction with CBF1 was demonstrated in a glutathione S-transferase (GST) affinity assay and by the ability of RPMS to alter the intracellular localization of a mutant CBF1. A Gal4-RPMS fusion protein mediated transcriptional repression, suggesting an additional interaction between RPMS and corepressor proteins. GST affinity assays revealed interaction between RPMS and the corepressor Sin3A and CIR. The RPMS-CIR interaction was further substantiated in mammalian two-hybrid, coimmunoprecipitation, and colocalization experiments. RPMS has been shown to interfere with NotchIC and EBNA2 activation of CBF1-containing promoters in reporter assays. Consistent with this function, immunofluorescence assays performed on cotransfected cells showed that there was colocalization of RPMS with NotchIC and with EBNA2 in intranuclear punctate speckles. The effect of RPMS on NotchIC function was further examined in a muscle cell differentiation assay where RPMS was found to partially reverse NotchIC-mediated inhibition of differentiation. The mechanism of RPMS action was examined in cotransfection and mammalian two-hybrid assays. The results revealed that RPMS blocked relief of CBF1-mediated repression and interfered with SKIP-CIR interactions. We conclude that RPMS acts as a negative regulator of EBNA2 and Notch activity through its interactions with the CBF1-associated corepressor complex.
Collapse
Affiliation(s)
- J Zhang
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, USA
| | | | | | | |
Collapse
|
31
|
Gottschalk S, Ng CY, Perez M, Smith CA, Sample C, Brenner MK, Heslop HE, Rooney CM. An Epstein-Barr virus deletion mutant associated with fatal lymphoproliferative disease unresponsive to therapy with virus-specific CTLs. Blood 2001; 97:835-43. [PMID: 11159505 DOI: 10.1182/blood.v97.4.835] [Citation(s) in RCA: 196] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
There is a growing interest in using antigen-specific T cells for the treatment of human malignancy. For example, adoptive transfer of Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes (CTLs) has been effective prophylaxis and treatment of EBV-associated lymphoproliferative disease in immunocompromised patients. For all immunotherapies, however, there has been a hypothetical concern that mutations in tumor-specific antigens may lead to tumor escape. We now demonstrate that such events may indeed occur, with lethal outcome. A patient who developed lymphoma after marrow transplantation received donor-derived, EBV-specific CTLs but died with progressive disease. The tumor cells proved substantially less sensitive to cytolysis than the EBV-transformed B-cell line used for CTL generation. The major cytolytic activity of the donor CTL was directed against 2 HLA-A11-restricted epitopes in the viral EBNA-3B antigen. Sequence analysis of this gene in the tumor virus revealed a 245-base pair deletion, which removed these 2 CTL epitopes. Hence, the viral antigen in the tumor had mutated in a way that allowed escape from CTLs. Analysis of EBV polymorphisms demonstrated that before CTL infusion, more than one virus was present, including a virus with wild-type EBNA-3B. After CTL infusion, only the virus with the EBNA-3B deletion could be detected, suggesting that the infused CTLs had selected a resistant strain in vivo. Such an occurrence, even when polyclonal CTL lines are used against genetically stable virus antigens, suggests that escape mutants may be a serious problem when CTL therapy is directed against more unstable tumor cell-derived targets.
Collapse
MESH Headings
- Adolescent
- Amino Acid Sequence
- Antineoplastic Agents, Alkylating/therapeutic use
- B-Lymphocytes/immunology
- Bone Marrow Transplantation/adverse effects
- Cell Line, Transformed/immunology
- Combined Modality Therapy
- Cyclophosphamide/therapeutic use
- Cytotoxicity, Immunologic
- Disease Progression
- Epstein-Barr Virus Infections/genetics
- Epstein-Barr Virus Infections/immunology
- Epstein-Barr Virus Infections/transmission
- Epstein-Barr Virus Infections/virology
- Epstein-Barr Virus Nuclear Antigens/genetics
- Epstein-Barr Virus Nuclear Antigens/immunology
- Fatal Outcome
- Female
- Genes, Viral
- HLA-A Antigens/immunology
- HLA-A11 Antigen
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/immunology
- Humans
- Immunodominant Epitopes/genetics
- Immunodominant Epitopes/immunology
- Immunotherapy, Adoptive
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/virology
- Molecular Sequence Data
- Polymerase Chain Reaction
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Sequence Alignment
- Sequence Deletion
- Sequence Homology, Amino Acid
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
- Tissue Donors
- Transplantation, Homologous/adverse effects
- Viral Structural Proteins/genetics
Collapse
Affiliation(s)
- S Gottschalk
- Center for Cell and Gene Therapy, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Imai S, Nishikawa J, Kuroda M, Takada K. Epstein-Barr virus infection of human epithelial cells. Curr Top Microbiol Immunol 2001; 258:161-84. [PMID: 11443861 DOI: 10.1007/978-3-642-56515-1_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- S Imai
- Department of Microbiology, Kochi Medical School, Kohasu, Okoh-cho, Nankoku, Kochi 783-8505, Japan
| | | | | | | |
Collapse
|
33
|
Kienzle N, Buck M, Silins SL, Burrows SR, Moss DJ, Winterhalter A, Brooks A, Khanna R. Differential splicing of antigen-encoding RNA reduces endogenous epitope presentation that regulates the expansion and cytotoxicity of T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:1840-6. [PMID: 10925262 DOI: 10.4049/jimmunol.165.4.1840] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The activation of CTLs is dependent on the recognition of MHC-bound peptide present on the surface of APCs. We give evidence in this study that differential splicing of Ag-encoding RNA can decrease the antigenic dose in APCs and regulate the recall of human memory CTLs. Differential splicing of RNA that encoded an immunodominant HLA-B8-restricted CTL epitope of EBV reduced the functional presentation of this epitope, and consequently the in vitro expansion and activity of CTLs, as measured by MHC/peptide-tetramer staining and cytotoxicity assays. The reduced activity of the stimulated CTLs was not only due to lower numbers of Ag-specific CTLs but, surprisingly, was also characterized by decreased cytotoxicity of the CTLs to target cells presenting limiting amounts of the peptide epitope. As indicated by TCR repertoire analysis, the reduction in CTL activity was not caused by stimulation of distinct populations of TCR clonotypes. This study demonstrates how a common eukaryotic posttranscriptional mechanism of gene regulation can modulate the endogenous presentation of Ag and ultimately contribute to the fine tuning of immunological memory cells, which are important in the fight against pathogens and tumors and in autoimmunity.
Collapse
MESH Headings
- Alternative Splicing/immunology
- Antigen Presentation/genetics
- Antigen-Presenting Cells/metabolism
- Cell Line, Transformed
- Cytotoxicity Tests, Immunologic
- Cytotoxicity, Immunologic/genetics
- Dose-Response Relationship, Immunologic
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/metabolism
- Epstein-Barr Virus Nuclear Antigens/biosynthesis
- Epstein-Barr Virus Nuclear Antigens/genetics
- Epstein-Barr Virus Nuclear Antigens/immunology
- Epstein-Barr Virus Nuclear Antigens/metabolism
- Genetic Vectors/biosynthesis
- Genetic Vectors/chemical synthesis
- Humans
- Immunodominant Epitopes/genetics
- Immunodominant Epitopes/metabolism
- Immunologic Memory/genetics
- Lymphocyte Activation/genetics
- Oligopeptides/antagonists & inhibitors
- Oligopeptides/biosynthesis
- Oligopeptides/genetics
- Oligopeptides/immunology
- RNA, Viral/genetics
- RNA, Viral/immunology
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- N Kienzle
- EBV Unit, The Queensland Institute of Medical Research and University of Queensland Joint Oncology Program, Brisbane, Queensland, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Lee SP, Chan AT, Cheung ST, Thomas WA, CroomCarter D, Dawson CW, Tsai CH, Leung SF, Johnson PJ, Huang DP. CTL control of EBV in nasopharyngeal carcinoma (NPC): EBV-specific CTL responses in the blood and tumors of NPC patients and the antigen-processing function of the tumor cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:573-82. [PMID: 10861098 DOI: 10.4049/jimmunol.165.1.573] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Undifferentiated nasopharyngeal carcinoma (NPC) is latently infected with EBV and expresses a restricted number of viral proteins. Studies in healthy virus carriers have demonstrated that at least some of these proteins can act as targets for HLA class I-restricted CTLs. Therefore we have explored the possibility of a CTL-based therapy for NPC by characterizing EBV-specific CTL responses in 10 newly diagnosed NPC cases and 21 healthy virus carriers from Southeast Asia. Using the autologous EBV-transformed lymphoblastoid cell line, virus-specific CTL were reactivated in vitro from PBMC, cloned, and screened for cytotoxicity against target cells expressing individual EBV proteins from recombinant vaccinia vectors. EBV-specific CTLs were identified in 6 of 10 patients and 14 of 21 controls and mainly targeted the EBV nuclear Ag 3 (EBNA3) family of viral latent proteins. However, in 3 of 10 patients and 11 of 21 controls, CTLs specific for the NPC-associated protein LMP2 were also detected, albeit at low frequency. EBV-specific CTLs were detected in tumor biopsy material obtained from 3 of 6 of the patients, indicating that functional CTL are present at the tumor site, but none was specific for tumor-associated viral proteins. To assess the Ag-presenting function in NPC we studied two NPC-derived cell lines (C15 and c666.1) and demonstrated that both were capable of processing and presenting endogenously synthesized protein to HLA class I-restricted CTL clones. Overall, our data provide a sound theoretical basis for therapeutic strategies that aim to boost or elicit LMP2-specific CTL responses in NPC patients.
Collapse
Affiliation(s)
- S P Lee
- CRC Institute for Cancer Studies, University of Birmingham, Edgbaston, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Smith PR, de Jesus O, Turner D, Hollyoake M, Karstegl CE, Griffin BE, Karran L, Wang Y, Hayward SD, Farrell PJ. Structure and coding content of CST (BART) family RNAs of Epstein-Barr virus. J Virol 2000; 74:3082-92. [PMID: 10708423 PMCID: PMC111807 DOI: 10.1128/jvi.74.7.3082-3092.2000] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/1999] [Accepted: 01/03/2000] [Indexed: 11/20/2022] Open
Abstract
CST (BART BARF0) family viral RNAs are expressed in several types of Epstein-Barr virus (EBV) infection, including EBV-associated cancers. Many different spliced forms of these RNAs have been described; here we have clarified the structures of some of the more abundant splicing patterns. We report the first cDNAs representing a full-length CST mRNA from a clone library and further characterize the transcription start. The relative abundance of splicing patterns and genomic analysis of the open reading frames (ORFs) suggest that, in addition to the much studied BARF0 ORF, there may be important products made from some of the upstream ORFs in the CST RNAs. Potential biological functions are identified for two of these. The product of the RPMS1 ORF is shown to be a nuclear protein that can bind to the CBF1 component of Notch signal transduction. RPMS1 can inhibit the transcription activation induced through CBF1 by NotchIC or EBNA-2. The protein product of another CST ORF, A73, is shown to be a cytoplasmic protein which can interact with the cell RACK1 protein. Since RACK1 modulates signaling from protein kinase C and Src tyrosine kinases, the results suggest a possible role for CST products in growth control, perhaps consistent with the abundant transcription of CST RNAs in cancers such as nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- P R Smith
- Virology and Cell Biology, Imperial College School of Medicine, London W2 1PG, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Benninger-Döring G, Pepperl S, Deml L, Modrow S, Wolf H, Jilg W. Frequency of CD8(+) T lymphocytes specific for lytic and latent antigens of Epstein-Barr virus in healthy virus carriers. Virology 1999; 264:289-97. [PMID: 10562493 DOI: 10.1006/viro.1999.9996] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We investigated CD8(+) T cell frequencies of five different Epstein-Barr virus-specific cytotoxic T lymphocyte epitopes located within proteins of the replicative cycle and the latent state in healthy long-term virus carriers with IFN-gamma enzyme-linked immunospot assay. Frequencies of the HLA-A3-restricted epitope RVRAYTYSK (RVR) whose minimal length was mapped in this study to amino acid position 148-156 of the immediate-early protein BRLF1 were compared with those of a further known HLA-A3-restricted epitope within EBNA3A, RLRAEAQVK (RLR). Determination of frequencies of CD8(+) T lymphocytes directed against lytic antigen epitope RVR revealed that only one of eight donors recognized this epitope. Frequency was calculated to be 65 RVR-specific CD8(+) T lymphocytes per 10(6) PBMC. None of the HLA-A3-positive donors exhibited IFN-gamma release after antigenic stimulation with the EBNA3A-specific peptide epitope RLR. Furthermore, we chose three known HLA-B8-restricted epitopes, RAKFKQLL (RAK), FLRGRAYGL (FLR), and QAKWRLQTL (QAK), of the lytic protein BZLF1 and the latent protein EBNA3A. Examination of eight HLA-B8-positive virus carriers revealed that the BZLF1-specific epitope RAK was recognized by all donors with a median frequency of 233 RAK-specific CD8(+) T lymphocytes per 10(6) PBMC. Only 50% of these donors reacted against EBNA3A-specific epitope FLR and a minority (25%) reacted against EBNA3A-specific epitope QAK.
Collapse
Affiliation(s)
- G Benninger-Döring
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Regensburg, Regensburg, D-93053, Germany.
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
The cellular localization of the Epstein-Barr virus-encoded RK-BARF0 protein was analyzed by fluorescence microscopy and immunoblotting. The recombinant RK-BARF0 protein was found to be tightly bound to nuclear structures, whereas 16- to 20-kDa RK-BARF0 derivatives, generated by differential splicing of the RK-BARF0 transcript, were present throughout the cell. Moreover, a previously generated anti-RK-BARF0 rabbit serum was found to cross-react with cellular proteins, showing that the previously identified 30- to 35-kDa membrane-associated proteins do not represent RK-BARF0.
Collapse
Affiliation(s)
- N Kienzle
- EBV Unit, The Queensland Institute of Medical Research and University of Queensland Joint Oncology Program, Brisbane, Australia.
| | | | | | | | | |
Collapse
|
38
|
Kuzushima K, Nakamura S, Nakamura T, Yamamura Y, Yokoyama N, Fujita M, Kiyono T, Tsurumi T. Increased frequency of antigen-specific CD8(+) cytotoxic T lymphocytes infiltrating an Epstein-Barr virus-associated gastric carcinoma. J Clin Invest 1999; 104:163-71. [PMID: 10411545 PMCID: PMC408473 DOI: 10.1172/jci6062] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Gastric adenocarcinomas carrying Epstein-Barr virus (EBV) are known to be accompanied by massive lymphocyte infiltration. To characterize the tumor-infiltrating lymphocytes (TILs), we isolated and cultured such cells from a surgically resected EBV-associated gastric carcinoma. They were found to be positive for CD3, CD8, T-cell receptor beta chain, and cytotoxic molecules. The isolated TILs consisted of human leukocyte antigen (HLA) class I-restricted CD8(+) cytotoxic T lymphocytes (CTLs), which killed autologous EBV-transformed cells (but not phytohemagglutinin blast cells) and recognized HLA-A24 as restriction molecules. However, the TILs did not recognize known EBV antigenic peptides presented by HLA-A24 molecules, nor HLA-A24(+) fibroblasts infected with vaccinia recombinant virus expressing each of the EBV latent proteins. EBV(+) gastric carcinomas do not express conventional target proteins of EBV-specific CTLs, and the data suggest that some cellular proteins may be involved in the strong T-cell response to EBV-associated gastric carcinoma. In addition, our data suggest that class I-restricted, antigen-specific CD8(+) CTLs are specifically expanded within EBV(+) gastric carcinoma tissue.
Collapse
Affiliation(s)
- K Kuzushima
- Laboratory of Viral Oncology, Aichi Cancer Center Research Institute, Nagoya 464-0021, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Kienzle N, Sculley TB, Greco S, Khanna R. Cutting Edge: Silencing Virus-Specific Cytotoxic T Cell-Mediated Immune Recognition by Differential Splicing: A Novel Implication of RNA Processing for Antigen Presentation. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.12.6963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
Persistent viruses have developed potent strategies to overcome host immune defenses. In particular, viral interference with Ag presentation by HLA class I molecules can effectively impair the host’s CTL function. Here we provide evidence for a novel aspect of differential splicing on endogenous processing of a latent viral transcript resulting in dominant protein isoforms from which the CTL determinant has been deleted. Consequently, virus-infected cells expressing these isoforms were poorly recognized by CTLs. Molecular analysis revealed that this splicing significantly reduced expression of the viral transcript encoding the relevant epitope to levels below the threshold required for CTL recognition. The importance of splicing was further reinforced by the observation of efficient CTL recognition of target cells expressing a truncated viral transcript that abolished differential splicing. Thus, differential splicing, which is a common mechanism of gene regulation in many pathogens, may unexpectedly interfere with immune recognition.
Collapse
Affiliation(s)
- Norbert Kienzle
- EBV Unit, Queensland Institute of Medical Research, and University of Queensland Joint Oncology Program, Brisbane, Australia
| | - Tom B. Sculley
- EBV Unit, Queensland Institute of Medical Research, and University of Queensland Joint Oncology Program, Brisbane, Australia
| | - Sonia Greco
- EBV Unit, Queensland Institute of Medical Research, and University of Queensland Joint Oncology Program, Brisbane, Australia
| | - Rajiv Khanna
- EBV Unit, Queensland Institute of Medical Research, and University of Queensland Joint Oncology Program, Brisbane, Australia
| |
Collapse
|