Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Wiley Interdiscip Rev RNA ; 6(2): 157-71, 2015.
Article in English | MEDLINE | ID: mdl-25264139

ABSTRACT

Effective T-cell surveillance of antigen-presenting cells is dependent on the expression of an array of antigenic peptides bound to major histocompatibility complex (MHC) class I (MHC-I) or class II (MHC-II) molecules. Pathogens co-evolving with their hosts exploit crucial translational regulatory mechanisms in order to evade host immune recognition and thereby sustain their infection. Evasion strategies that downregulate viral protein synthesis and thereby restrict antigen presentation to cytotoxic T-cells through the endogenous MHC-I pathway have been implicated in the pathogenesis of viral-associated malignancies. An understanding of the mechanisms by which messenger RNA (mRNA) structure modulates both viral mRNA translation and the antigen processing machinery to escape immune surveillance, will stimulate the development of alternative therapeutic strategies focused on RNA-directed drugs designed to enhance immune responses against infected cells. In this review, we discuss regulatory aspects of the MHC-I pathway and summarize current knowledge of the role attributed by mRNA structure and other translational regulatory mechanisms in immune evasion. In particular we highlight the impact of recently identified G-quadruplex structures within virally encoded transcripts as unique regulatory signals for translational control and antigen presentation.


Subject(s)
Antigen Presentation , Histocompatibility Antigens Class I/biosynthesis , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Adaptive Immunity , Animals , Humans , Immune Evasion , Immune Tolerance , Viruses/immunology
2.
PLoS Pathog ; 10(10): e1004423, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25299404

ABSTRACT

Recent studies have shown that virally encoded mRNA sequences of genome maintenance proteins from herpesviruses contain clusters of unusual structural elements, G-quadruplexes, which modulate viral protein synthesis. Destabilization of these G-quadruplexes can override the inhibitory effect on self-synthesis of these proteins. Here we show that the purine-rich repetitive mRNA sequence of Epstein-Barr virus encoded nuclear antigen 1 (EBNA1) comprising G-quadruplex structures, limits both the presentation of MHC class I-restricted CD8(+) T cell epitopes by CD11c(+) dendritic cells in draining lymph nodes and early priming of antigen-specific CD8(+) T-cells. Destabilization of the G-quadruplex structures through codon-modification significantly enhanced in vivo antigen presentation and activation of virus-specific T cells. Ex vivo imaging of draining lymph nodes by confocal microscopy revealed enhanced antigen-specific T-cell trafficking and APC-CD8(+) T-cell interactions in mice primed with viral vectors encoding a codon-modified EBNA1 protein. More importantly, these antigen-specific T cells displayed enhanced expression of the T-box transcription factor and superior polyfunctionality consistent with the qualitative impact of translation efficiency. These results provide an important insight into how viruses exploit mRNA structure to down regulate synthesis of their viral maintenance proteins and delay priming of antigen-specific T cells, thereby establishing a successful latent infection in vivo. Furthermore, targeting EBNA1 mRNA rather than protein by small molecules or antisense oligonucleotides will enhance EBNA1 synthesis and the early priming of effector T cells, to establish a more rapid immune response and prevent persistent infection.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Nuclear Antigens/genetics , RNA, Messenger/genetics , Animals , CD11c Antigen/immunology , Epitopes, T-Lymphocyte/immunology , Female , Genes, MHC Class I/immunology , Mice, Inbred C57BL , Protein Biosynthesis/genetics
3.
Nat Chem Biol ; 10(5): 358-64, 2014 May.
Article in English | MEDLINE | ID: mdl-24633353

ABSTRACT

Viruses that establish latent infections have evolved unique mechanisms to avoid host immune recognition. Maintenance proteins of these viruses regulate their synthesis to levels sufficient for maintaining persistent infection but below threshold levels for host immune detection. The mechanisms governing this finely tuned regulation of viral latency are unknown. Here we show that mRNAs encoding gammaherpesviral maintenance proteins contain within their open reading frames clusters of unusual structural elements, G-quadruplexes, which are responsible for the cis-acting regulation of viral mRNA translation. By studying the Epstein-Barr virus-encoded nuclear antigen 1 (EBNA1) mRNA, we demonstrate that destabilization of G-quadruplexes using antisense oligonucleotides increases EBNA1 mRNA translation. In contrast, pretreatment with a G-quadruplex-stabilizing small molecule, pyridostatin, decreases EBNA1 synthesis, highlighting the importance of G-quadruplexes within virally encoded transcripts as unique regulatory signals for translational control and immune evasion. Furthermore, these findings suggest alternative therapeutic strategies focused on targeting RNA structure within viral ORFs.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/genetics , G-Quadruplexes , Protein Biosynthesis , RNA, Messenger/genetics , Base Sequence
4.
Blood ; 120(16): 3237-45, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-22942184

ABSTRACT

T-box transcription factors T-bet (Tbx21) and Eomesodermin (Eomes) are critical players in CD8(+) cytotoxic T lymphocyte effector function and differentiation, but how the expression of these transcription factors is regulated remains poorly defined. Here we show that dominant T cells directed toward human CMV, expressing significantly higher levels of T-bet with graded loss of Eomes expression (T-bet(hi)Eomes(hi/lo)), are more efficient in recognizing endogenously processed peptide-major histocompatibility complexes (pMHC) compared with subdominant virus-specific T cells expressing lower levels of T-bet and high levels of Eomes (T-bet(int)Eomes(hi)). Paradoxically, the T-bet(hi)Eomes(hi/lo) dominant populations that efficiently recognized endogenous antigen demonstrated lower intrinsic avidity for pMHC, whereas T-bet(int)Eomes(hi) subdominant populations were characterized by higher pMHC avidity and less efficient recognition of virus-infected cells. Importantly, differential endogenous viral antigen recognition by CMV-specific CD8(+) T cells also correlated with the differentiation status and expression of perforin, granzyme B and K. Furthermore, we demonstrate that the expression of T-bet correlates with clonal expansion, differentiation status, and expression of perforin, granzyme B and K in antigen-specific T cells. These findings illustrate how endogenous viral antigen presentation during persistent viral infection may influence the transcriptional program of virus-specific T cells and their functional profile in the peripheral blood of humans.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/microbiology , Flow Cytometry , Granzymes/metabolism , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Major Histocompatibility Complex/immunology , Perforin/metabolism , T-Box Domain Proteins/immunology
5.
PLoS Pathog ; 8(12): e1003112, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23300450

ABSTRACT

Unique purine-rich mRNA sequences embedded in the coding sequences of a distinct group of gammaherpesvirus maintenance proteins underlie the ability of the latently infected cell to minimize immune recognition. The Epstein-Barr virus nuclear antigen, EBNA1, a well characterized lymphocryptovirus maintenance protein has been shown to inhibit in cis antigen presentation, due in part to a large internal repeat domain encoding glycine and alanine residues (GAr) encoded by a purine-rich mRNA sequence. Recent studies have suggested that it is the purine-rich mRNA sequence of this repeat region rather than the encoded GAr polypeptide that directly inhibits EBNA1 self-synthesis and contributes to immune evasion. To test this hypothesis, we generated a series of EBNA1 internal repeat frameshift constructs and assessed their effects on cis-translation and endogenous antigen presentation. Diverse peptide sequences resulting from alternative repeat reading frames did not alleviate the translational inhibition characteristic of EBNA1 self-synthesis or the ensuing reduced surface presentation of EBNA1-specific peptide-MHC class I complexes. Human cells expressing the EBNA1 frameshift variants were also poorly recognized by antigen-specific T-cells. Furthermore, a comparative analysis of the mRNA sequences of the corresponding repeat regions of different viral maintenance homologues highlights the high degree of identity between the nucleotide sequences despite very little homology in the encoded amino acid sequences. Based on these combined observations, we propose that the cis-translational inhibitory effect of the EBNA1 internal repeat sequence operates mechanistically at the nucleotide level, potentially through RNA secondary structural elements, and is unlikely to be mediated through the GAr polypeptide. The demonstration that the EBNA1 repeat mRNA sequence and not the encoded protein sequence underlies immune evasion in this class of virus suggests a novel approach to therapeutic development through the use of anti-sense strategies or small molecules targeting EBNA1 mRNA structure.


Subject(s)
Amino Acid Sequence/genetics , Antigen Presentation/immunology , Base Sequence/genetics , Epstein-Barr Virus Nuclear Antigens , RNA, Messenger/genetics , Antigen Presentation/genetics , Cell Line , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/immunology , Epstein-Barr Virus Nuclear Antigens/metabolism , Frameshift Mutation , Genes, MHC Class I , HEK293 Cells , Humans , Immune Evasion , Molecular Sequence Data , Repetitive Sequences, Amino Acid/genetics , Repetitive Sequences, Amino Acid/immunology , Sequence Alignment , T-Lymphocytes, Cytotoxic/immunology
6.
Proc Natl Acad Sci U S A ; 107(7): 3146-51, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133771

ABSTRACT

EBV causes infectious mononucleosis and is associated with certain malignancies. EBV nuclear antigen 1 (EBNA1) mediates EBV genome replication, partition, and transcription, and is essential for persistence of the viral genome in host cells. Here we demonstrate that Hsp90 inhibitors decrease EBNA1 expression and translation, and that this effect requires the Gly-Ala repeat domain of EBNA1. Hsp90 inhibitors induce the death of established, EBV-transformed lymphoblastoid cell lines at doses nontoxic to normal cells, and this effect is substantially reversed when lymphoblastoid cell lines are stably infected with a retrovirus expressing a functional EBNA1 mutant lacking the Gly-Ala repeats. Hsp90 inhibitors prevent EBV transformation of primary B cells, and strongly inhibit the growth of EBV-induced lymphoproliferative disease in SCID mice. These results suggest that Hsp90 inhibitors may be particularly effective for treating EBV-induced diseases requiring the continued presence of the viral genome.


Subject(s)
Benzoquinones/pharmacology , Epstein-Barr Virus Nuclear Antigens/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Herpesvirus 4, Human , Lactams, Macrocyclic/pharmacology , Lymphoproliferative Disorders/drug therapy , Animals , Apoptosis/drug effects , Benzoquinones/therapeutic use , Cell Line, Tumor , DNA Primers/genetics , Dipeptides/genetics , Gene Expression Regulation, Viral/drug effects , Humans , Immunoblotting , Immunoprecipitation , Lactams, Macrocyclic/therapeutic use , Lymphoproliferative Disorders/virology , Mice , Mice, SCID , Reverse Transcriptase Polymerase Chain Reaction , Virus Replication/drug effects
7.
Mol Immunol ; 46(8-9): 1911-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19157553

ABSTRACT

The major ligands presented by MHC class I molecules after natural antigen processing are peptides of eight to ten residues in length, and it is widely accepted that the binding preferences of MHC class I molecules play a dominant role in dictating this classic feature of antigen presentation. In this report, we have reassessed the peptide size specificity of class I human leukocyte antigens (HLAs). By lengthening previously defined T cell epitopes by central amino acid insertion, we demonstrate that the peptide length specificity of some common HLA class I alleles (HLA-B*3501, B*0702 and A*2402) is very broad, and includes peptides of up to 25 residues. These data suggest that the length limitation of naturally processed MHC class I-associated peptides is primarily controlled by peptide availability after antigen processing rather than the binding specificity of MHC class I molecules. Furthermore, the findings provide an explanation for recent reports highlighting that epitopes of >10 amino acids play a minor but significant role in virus-specific immune surveillance by CD8(+) T cells.


Subject(s)
Amino Acid Sequence/physiology , HLA-B Antigens/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Binding Sites/physiology , Cells, Cultured , HLA-B Antigens/genetics , Humans , Molecular Sequence Data , Protein Binding/genetics , Substrate Specificity
8.
Semin Cancer Biol ; 18(6): 397-408, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18977445

ABSTRACT

Upon primary infection, EBV establishes a latent infection in B cells, characterized by maintenance of the viral genome in the absence of viral replication. The Epstein-Barr Nuclear Antigen 1 (EBNA1) plays a crucial role in maintenance of the viral DNA episome and is consistently expressed in all EBV-associated malignancies. Compared to other EBV latent gene products, EBNA1 is poorly recognized by CD8(+) T lymphocytes. Recent studies are discussed that shed new light on the mechanisms that underlie this unusual lack of CD8(+) T cell activation. Whereas the latent phase is characterized by the expression of a limited subset of viral gene products, the full repertoire of over 80 EBV lytic gene products is expressed during the replicative phase. Despite this abundance of potential T cell antigens, which indeed give rise to a strong response of CD4(+) and CD8(+) T lymphocytes, the virus can replicate successfully. Evidence is accumulating that this paradoxical situation is the result of actions of multiple viral gene products, inhibiting discrete stages of the MHC class I and class II antigen presentation pathways. Immediately after initiation of the lytic cycle, BNLF2a prevents peptide-loading of MHC class I molecules through inhibition of the Transporter associated with Antigen Processing, TAP. This will reduce presentation of viral antigens by the large ER-resident pool of MHC class I molecules. Synthesis of new MHC class I molecules is blocked by BGLF5. Viral-IL10 causes a reduction in mRNA levels of TAP1 and bli/LMP2, a subunit of the immunoproteasome. MHC class I molecules present at the cell surface are downregulated by BILF1. Also the antigen presenting capacity of MHC class II molecules is severely compromised by multiple EBV lytic gene products, including gp42/gH/gL, BGLF5, and vIL-10. In this review, we discuss how concerted actions of these EBV lytic proteins result in highly effective interference with CD8(+) and CD4(+) T cell surveillance, thereby providing the virus with a window for undisturbed generation of viral progeny.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Nuclear Antigens/genetics , Herpesvirus 4, Human/genetics , ATP-Binding Cassette Transporters/immunology , ATP-Binding Cassette Transporters/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cytokines/immunology , Cytokines/metabolism , Deoxyribonucleases/immunology , Deoxyribonucleases/metabolism , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/immunology , Epstein-Barr Virus Nuclear Antigens/metabolism , Glycoproteins/immunology , Glycoproteins/metabolism , Herpesvirus 4, Human/immunology , Humans , Immunity , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Viral Proteins/immunology , Viral Proteins/metabolism
9.
PLoS One ; 3(9): e3254, 2008 Sep 22.
Article in English | MEDLINE | ID: mdl-18806872

ABSTRACT

Previous studies have indicated that Epstein-Barr virus (EBV) can modulate the Wnt pathway in virus-infected cells and this effect is mediated by EBV-encoded oncogene latent membrane protein 1 (LMP1). Here we have reassessed the role of LMP1 in regulating the expression of various mediators of the canonical Wnt cascade. Contradicting the previous finding, we found that the levels of E-cadherin, beta-catenin, Glycogen Synthase Kinase 3ss (GSK3beta), axin and alpha-catenin were not affected by the expression of LMP1 sequences from normal B cells or nasopharyngeal carcinoma. Moreover, we also show that LMP1 expression had no detectable effect on the E-cadherin and beta-catenin interaction and did not induce transcriptional activation of beta-catenin. Taken together these studies demonstrate that EBV-mediated activation of Wnt pathway is not dependent on the expression of LMP1.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Herpesvirus 4, Human/metabolism , Viral Matrix Proteins/metabolism , Wnt Proteins/metabolism , Animals , Cadherins/metabolism , Cell Membrane/metabolism , Dogs , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Models, Biological , Transcriptional Activation , alpha Catenin/metabolism , beta Catenin/metabolism
10.
Proc Natl Acad Sci U S A ; 105(27): 9319-24, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18591662

ABSTRACT

Many viruses avoid immune surveillance during latent infection through reduction in the synthesis of virally encoded proteins. Although antigen presentation critically depends on the level of viral protein synthesis, the precise mechanism used to regulate the generation of antigenic peptide precursors remains elusive. Here, we demonstrate that a purine overloaded virally encoded mRNA lacking secondary structure significantly impacts the efficiency of protein translation and prevents endogenous antigen presentation. Reducing this purine bias through the generation of constructs expressing codon-modified sequences, while maintaining the encoded protein sequence, increased the stem-loop structure of the corresponding mRNA and dramatically enhanced self-synthesis of the viral protein. As a consequence, a higher number of HLA-peptide complexes were detected on the surface of cells expressing this viral protein. Furthermore, these cells were more efficiently recognized by virus-specific T cells compared with those expressing the same antigen expressed by a purine-biased mRNA. These findings delineate a mechanism by which viruses regulate self-synthesis of proteins and offer an effective strategy to evade CD8(+) T cell-mediated immune regulation.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Protein Biosynthesis , RNA, Messenger/chemistry , Base Sequence , Cell Line , Codon/genetics , Epitopes/immunology , Epstein-Barr Virus Nuclear Antigens/biosynthesis , Epstein-Barr Virus Nuclear Antigens/chemistry , Epstein-Barr Virus Nuclear Antigens/genetics , Gene Expression Regulation , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Oligonucleotides, Antisense , Protein Structure, Tertiary , Purines , RNA Stability , RNA, Messenger/genetics
11.
J Exp Med ; 204(3): 525-32, 2007 Mar 19.
Article in English | MEDLINE | ID: mdl-17312009

ABSTRACT

A significant proportion of endogenously processed CD8(+) T cell epitopes are derived from newly synthesized proteins and rapidly degrading polypeptides (RDPs). It has been hypothesized that the generation of rapidly degrading polypeptides and CD8(+) T cell epitopes from these RDP precursors may be influenced by the efficiency of protein translation. Here we address this hypothesis by using the Epstein-Barr virus-encoded nuclear antigen 1 protein (EBNA1), with or without its internal glycine-alanine repeat sequence (EBNA1 and EBNA1DeltaGA, respectively), which display distinct differences in translation efficiency. We demonstrate that RDPs constitute a significant proportion of newly synthesized EBNA1 and EBNA1DeltaGA and that the levels of RDPs produced by each of these proteins directly correlate with the translation efficiency of either EBNA1 or EBNA1DeltaGA. As a consequence, a higher number of major histocompatibility complex-peptide complexes can be detected on the surface of cells expressing EBNA1DeltaGA, and these cells are more efficiently recognized by virus-specific cytotoxic T lymphocytes compared to the full-length EBNA1. More importantly, we also demonstrate that the endogenous processing of these CD8(+) T cell epitopes is predominantly determined by the rate at which the RDPs are generated rather than the intracellular turnover of these proteins.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Epstein-Barr Virus Nuclear Antigens/biosynthesis , Epstein-Barr Virus Nuclear Antigens/genetics , Sequence Homology, Amino Acid , Antigen Presentation/genetics , Cell Line , Cell Line, Transformed , Dipeptides/genetics , Epstein-Barr Virus Nuclear Antigens/chemistry , Humans , Repetitive Sequences, Amino Acid/genetics , Sequence Deletion
12.
Eur J Immunol ; 37(2): 328-37, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17236233

ABSTRACT

Lymphocryptoviruses (LCV) that infect humans and Old World primates display a significant degree of genetic identity. These viruses use B lymphocytes as primary host cells to establish a long-term latent infection and express highly homologous latent viral proteins. Of particular interest is the expression of the EBV-encoded nuclear antigen-1 (EBNA1), which plays a crucial role in maintaining the viral genome in B cells. Using human and Old World primate homologues of EBNA1, we show that the internal repeat sequences differentially influence their in vitro translation efficiency. Although the glycine-alanine repeat domain of human LCV (EBV) EBNA1 inhibits its self-synthesis, the repeat domains within the simian LCV homologues of EBNA1 do not inhibit self-synthesis. As a consequence, simian LCV EBNA1-expressing cells are more efficiently recognized by virus-specific CTL when compared to human EBV EBNA1, even though both proteins are highly stable in B cells. Interestingly, we also show that similar to human EBNA1, CD8+ T cell epitopes from simian LCV EBNA1 are predominantly derived from newly synthesized protein rather than the long-lived pool of stable protein. These observations provide additional evidence that supports the theory that immune recognition of EBNA1 can occur without compromising the biological maintenance function of this protein.


Subject(s)
Antigen Presentation/immunology , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/immunology , Lymphocryptovirus/immunology , Protein Biosynthesis , Animals , CD8-Positive T-Lymphocytes/immunology , Electrophoresis, Polyacrylamide Gel , Epitopes, T-Lymphocyte/immunology , Humans , Immunoblotting , Transfection
13.
J Immunol ; 177(7): 4897-906, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16982932

ABSTRACT

Recent studies on Hodgkin's lymphoma (HL) have indicated that patients with active disease display functional impairment of Ag-specific CD8+ T cells due to expansion of regulatory T cells at sites of disease and in the peripheral blood. Adoptive cellular immunotherapy based on EBV-specific CD8+ T cells has been explored with limited success to date. It has been proposed that improved targeting of these CD8+ T cells toward viral Ags that are expressed in HL may enhance future therapeutic vaccine strategies. In this study, we have developed a novel replication-deficient adenoviral Ag presentation system that is designed to encode glycine alanine repeat-deleted EBV nuclear Ag 1 covalently linked to multiple CD8+ T cell epitopes from latent membrane proteins 1 and 2. A single stimulation of CD8+ T cells from healthy virus carriers, and patients with HL with this adenoviral construct in combination with IL-2, was sufficient to reverse the functional T cell impairment and restored both IFN-gamma production and cytolytic function. More importantly, these activated CD8+ T cells responded to tumor cells expressing membrane proteins and recognized novel EBNA1 epitopes. Flow cytometric analysis revealed that a large proportion of T cells expanded from patients with HL were CD62L(high) and CD27(high), and CCR7(low), consistent with early to mid effector T cells. These findings provide an important platform for translation of Ag-specific adoptive immunotherapy for the treatment of EBV-associated malignancies such as HL and nasopharyngeal carcinoma.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Hodgkin Disease/therapy , Immunotherapy, Adoptive , Viral Matrix Proteins/immunology , Adenoviridae/genetics , Amino Acid Sequence , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/immunology , Female , Genetic Vectors , Herpesvirus 4, Human/immunology , Hodgkin Disease/immunology , Hodgkin Disease/virology , Humans , In Vitro Techniques , Lymphocyte Activation/immunology , Male , Polymerase Chain Reaction , Recombinant Proteins/immunology , Tumor Virus Infections/complications , Viral Matrix Proteins/genetics
14.
J Exp Med ; 202(9): 1249-60, 2005 Nov 07.
Article in English | MEDLINE | ID: mdl-16275762

ABSTRACT

Thousands of potentially antigenic peptides are encoded by an infecting pathogen; however, only a small proportion induce measurable CD8(+) T cell responses. To investigate the factors that control peptide immunogenicity, we have examined the cytotoxic T lymphocyte (CTL) response to a previously undefined epitope ((77)APQPAPENAY(86)) from the BZLF1 protein of Epstein-Barr virus (EBV). This peptide binds well to two human histocompatibility leukocyte antigen (HLA) allotypes, HLA-B*3501 and HLA-B*3508, which differ by a single amino acid at position 156 ((156)Leucine vs. (156)Arginine, respectively). Surprisingly, only individuals expressing HLA-B*3508 show evidence of a CTL response to the (77)APQPAPENAY(86) epitope even though EBV-infected cells expressing HLA-B*3501 process and present similar amounts of peptide for CTL recognition, suggesting that factors other than peptide presentation levels are influencing immunogenicity. Functional and structural analysis revealed marked conformational differences in the peptide, when bound to each HLA-B35 allotype, that are dictated by the polymorphic HLA residue 156 and that directly affected T cell receptor recognition. These data indicate that the immunogenicity of an antigenic peptide is influenced not only by how well the peptide binds to major histocompatibility complex (MHC) molecules but also by its bound conformation. It also illustrates a novel mechanism through which MHC polymorphism can further diversify the immune response to infecting pathogens.


Subject(s)
DNA-Binding Proteins/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , HLA-B Antigens/metabolism , Herpesvirus 4, Human/immunology , T-Lymphocytes, Cytotoxic/immunology , Trans-Activators/immunology , Viral Proteins/immunology , Alleles , Clone Cells , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Epitopes, T-Lymphocyte/chemistry , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/metabolism , HLA-B35 Antigen , HLA-B38 Antigen , Herpesvirus 4, Human/metabolism , Humans , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , Protein Conformation , Protein Structure, Tertiary , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology , Trans-Activators/chemistry , Trans-Activators/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism
15.
Eur J Immunol ; 35(3): 996-1007, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15726667

ABSTRACT

Stem cell transplantation (SCT) remains the most effective curative therapy for the majority of hematopoietic malignancies. Unfortunately, SCT is limited by its toxicity and infectious complications that result from profound immunosuppression. In particular, acquisition of exogenous or reactivation of endogenous human cytomegalovirus (HCMV) is common after SCT. More recently, reconstitution of host immunity through augmentation of anti-HCMV T cell responses has been proposed as an exciting candidate therapy to avoid the requirement for antiviral drug use. Here we have developed a novel antigen presentation system based on a replication-deficient adenovirus that encodes multiple HLA class I-restricted epitopes from eight different antigens of HCMV as a polyepitope (referred to as AdCMVpoly). Ex vivo stimulation of peripheral blood mononuclear cells with AdCMVpoly consistently showed rapid stimulation and expansion of multiple epitope-specific T cells that recognized endogenously processed epitopes presented on virus-infected cells. Interestingly, the AdCMVpoly expression system is capable of expanding antigen-specific T cells even in the absence of CD4(+) T cells. These studies show the effectiveness of a polyepitope antigen presentation system for reproducible expansion of antigen-specific T cells from immunocompetent and immunocompromised settings.


Subject(s)
Antigens, Viral/immunology , Cytomegalovirus/immunology , Epitopes, T-Lymphocyte/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Cytotoxic/immunology , Adenoviridae/genetics , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Cell Line , Histocompatibility Antigens Class I/immunology , Humans , Immunologic Techniques , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
16.
Eur J Immunol ; 34(9): 2510-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15307183

ABSTRACT

The BZLF1 antigen of Epstein-Barr virus includes three overlapping sequences of different lengths that conform to the binding motif of human leukocyte antigen (HLA) B*3501. These 9-mer (56LPQGQLTAY64), 11-mer (54EPLPQGQLTAY64), and 13-mer (52LPEPLPQGQLTAY64) peptides all bound well to B*3501; however, the CTL response in individuals expressing this HLA allele was directed strongly and exclusively towards the 11-mer peptide. In contrast, EBV-exposed donors expressing HLA B*3503 showed no significant CTL response to these peptides because the single amino acid difference between B*3501 and B*3503 within the F pocket inhibited HLA binding by these peptides. The extraordinarily long 13-mer peptide was the target for the CTL response in individuals expressing B*3508, which differs from B*3501 at a single position within the D pocket (B*3501, 156Leucine; B*3508, 156Arginine). This minor difference was shown to enhance binding of the 13-mer peptide, presumably through a stabilizing interaction between the negatively charged glutamate at position 3 of the peptide and the positively charged arginine at HLA position 156. The 13-mer epitope defined in this study represents the longest class I-binding viral epitope identified to date as a minimal determinant. Furthermore, the potency of the response indicates that peptides of this length do not present a major structural barrier to CTL recognition.


Subject(s)
Epitopes, T-Lymphocyte , HLA-B35 Antigen/metabolism , Herpesvirus 4, Human/immunology , Peptide Fragments/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes/immunology , Antigen Presentation , Cell Line , DNA-Binding Proteins/immunology , Humans , Immunodominant Epitopes , Polymorphism, Genetic , Trans-Activators/immunology , Viral Proteins/immunology
17.
J Virol ; 78(13): 7131-7, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15194789

ABSTRACT

The geographically constrained distribution of Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC) in southeast Asian populations suggests that both viral and host genetics may influence disease risk. Although susceptibility loci have been mapped within the human genome, the role of viral genetics in the focal distribution of NPC remains an enigma. Here we report a molecular phylogenetic analysis of an NPC-associated viral oncogene, LMP1, in a large panel of EBV isolates from southeast Asia and from Papua New Guinea, Africa, and Australia, regions of the world where NPC is and is not endemic, respectively. This analysis revealed that LMP1 sequences show a distinct geographic structure, indicating that the southeast Asian isolates have evolved as a lineage distinct from those of Papua New Guinea, African, and Australian isolates. Furthermore, a likelihood ratio test revealed that the C termini of the LMP1 sequences of the southeast Asian lineage are under significant positive selection pressure, particularly at some sites within the C-terminal activator regions. We also present evidence that although the N terminus and transmembrane region of LMP1 have undergone recombination, the C-terminal region of the gene has evolved without any history of recombination. Based on these observations, we speculate that selection pressure may be driving the LMP1 sequences in virus isolates from southeast Asia towards a more malignant phenotype, thereby influencing the endemic distribution of NPC in this region.


Subject(s)
Evolution, Molecular , Herpesvirus 4, Human/genetics , Nasopharyngeal Neoplasms/virology , Selection, Genetic , Viral Matrix Proteins/genetics , Amino Acid Sequence , Asia, Southeastern , Cell Line , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/isolation & purification , Humans , Lymphocytes , Molecular Sequence Data , Phylogeny
18.
J Exp Med ; 199(10): 1421-31, 2004 May 17.
Article in English | MEDLINE | ID: mdl-15148340

ABSTRACT

Epstein-Barr virus (EBV)-encoded nuclear antigen (EBNA)1 is thought to escape cytotoxic T lymphocyte (CTL) recognition through either self-inhibition of synthesis or by blockade of proteasomal degradation by the glycine-alanine repeat (GAr) domain. Here we show that EBNA1 has a remarkably varied cell type-dependent stability. However, these different degradation rates do not correspond to the level of major histocompatibility complex class I-restricted presentation of EBNA1 epitopes. In spite of the highly stable expression of EBNA1 in B cells, CTL epitopes derived from this protein are efficiently processed and presented to CD8+ T cells. Furthermore, we show that EBV-infected B cells can readily activate EBNA1-specific memory T cell responses from healthy virus carriers. Functional assays revealed that processing of these EBNA1 epitopes is proteasome and transporter associated with antigen processing dependent. We also show that the endogenous presentation of these epitopes is dependent on the newly synthesized protein rather than the long-lived stable EBNA1. Based on these observations, we propose that defective ribosomal products, not the full-length antigen, are the primary source of endogenously processed CD8+ T cell epitopes from EBNA1.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Nuclear Antigens/immunology , Herpesvirus 4, Human/immunology , B-Lymphocytes/immunology , Cell Line , Cycloheximide/pharmacology , Epitopes/immunology , Epstein-Barr Virus Nuclear Antigens/genetics , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/genetics , Humans , Recombinant Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology , Transfection
19.
Br J Haematol ; 125(3): 267-81, 2004 May.
Article in English | MEDLINE | ID: mdl-15086409

ABSTRACT

Survivors of Hodgkin's lymphoma (HL) frequently have many years to experience the long-term toxicities of combined modality therapies. Also, a significant proportion of HL patients will relapse or have refractory disease, and less than half of these patients will respond to current salvage strategies. 30-50% of HL cases are Epstein-Barr virus associated (EBV-positive HL). The virus is localized to the malignant cells and is clonal. EBV-positive HL is more frequent in childhood, in older adults (>45 years) and in mixed cellularity cases. The survival of EBV-positive HL in the elderly and the immunosuppressed is particularly poor. Despite improvements in our understanding of EBV-positive HL, the true contribution of EBV to the pathogenesis of HL remains unknown. Increased knowledge of the virus' role in the basic biology of HL may generate novel therapeutic strategies for EBV-positive HL and the presence of EBV-latent antigens in the malignant HL cells may represent a target for cellular immunotherapy.


Subject(s)
Epstein-Barr Virus Infections/complications , Hodgkin Disease/virology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/therapy , Hodgkin Disease/immunology , Hodgkin Disease/therapy , Humans , Immune Tolerance , Immunotherapy/methods , Prognosis
20.
Cancer Res ; 64(4): 1483-9, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14973049

ABSTRACT

The EBV-encoded latent membrane proteins (LMP1 and LMP2), which are expressed in various EBV-associated malignancies have been proposed as a potential target for CTL-based therapy. However, the precursor frequency for LMP-specific CTL is generally low, and immunotherapy based on these antigens is often compromised by the poor immunogenicity and potential threat from their oncogenic potential. Here we have developed a replication- incompetent adenoviral vaccine that encodes multiple HLA class I-restricted CTL epitopes from LMP1 and LMP2 as a polyepitope. Immunization with this polyepitope vaccine consistently generated strong LMP-specific CTL responses in HLA A2/K(b) mice, which can be readily detected by both ex vivo and in vivo T-cell assays. Furthermore, a human CTL response to LMP antigens can be rapidly expanded after stimulation with this recombinant polyepitope vector. These expanded T cells displayed strong lysis of autologous target cells sensitized with LMP1 and/or LMP2 CTL epitopes. More importantly, this adenoviral vaccine was also successfully used to reverse the outgrowth of LMP1-expressing tumors in HLA A2/K(b) mice. These studies demonstrate that a replication-incompetent adenovirus polyepitope vaccine is an excellent tool for the induction of a protective CTL response directed toward multiple LMP CTL epitopes restricted through common HLA class I alleles prevalent in different ethnic groups where EBV-associated malignancies are endemic.


Subject(s)
Hodgkin Disease/therapy , Nasopharyngeal Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/therapeutic use , Viral Matrix Proteins/immunology , Viral Vaccines/therapeutic use , Adenoviridae/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Epitopes, T-Lymphocyte , H-2 Antigens/immunology , Humans , Immunization , Mice , Molecular Sequence Data , Viral Matrix Proteins/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...