Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.226
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 37: 547-570, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30699000

ABSTRACT

Adaptive immune recognition is mediated by antigen receptors on B and T cells generated by somatic recombination during lineage development. The high level of diversity resulting from this process posed technical limitations that previously limited the comprehensive analysis of adaptive immune recognition. Advances over the last ten years have produced data and approaches allowing insights into how T cells develop, evolutionary signatures of recombination and selection, and the features of T cell receptors that mediate epitope-specific binding and T cell activation. The size and complexity of these data have necessitated the generation of novel computational and analytical approaches, which are transforming how T cell immunology is conducted. Here we review the development and application of novel biological, theoretical, and computational methods for understanding T cell recognition and discuss the potential for improved models of receptor:antigen interactions.


Subject(s)
Computational Biology/methods , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Antigens/immunology , Antigens/metabolism , Cell Differentiation , Clonal Selection, Antigen-Mediated , Epitopes, T-Lymphocyte/metabolism , High-Throughput Nucleotide Sequencing , Humans , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism
2.
Annu Rev Immunol ; 34: 65-92, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26666651

ABSTRACT

T cell responses display two key characteristics. First, a small population of epitope-specific naive T cells expands by several orders of magnitude. Second, the T cells within this proliferating population take on diverse functional and phenotypic properties that determine their ability to exert effector functions and contribute to T cell memory. Recent technological advances in lineage tracing allow us for the first time to study these processes in vivo at single-cell resolution. Here, we summarize resulting data demonstrating that although epitope-specific T cell responses are reproducibly similar at the population level, expansion potential and diversification patterns of the offspring derived from individual T cells are highly variable during both primary and recall immune responses. In spite of this stochastic response variation, individual memory T cells can serve as adult stem cells that provide robust regeneration of an epitope-specific tissue through population averaging. We discuss the relevance of these findings for T cell memory formation and clinical immunotherapy.


Subject(s)
Adult Stem Cells/immunology , Cell Differentiation , Immunotherapy/methods , Single-Cell Analysis/methods , T-Lymphocytes/immunology , Animals , Biodiversity , Cell Lineage , Cell Proliferation , Cultural Diversity , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Humans , Immunologic Memory , Lymphocyte Activation
3.
Cell ; 174(1): 117-130.e14, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29909981

ABSTRACT

Heterogeneity is a hallmark feature of the adaptive immune system in vertebrates. Following infection, naive T cells differentiate into various subsets of effector and memory T cells, which help to eliminate pathogens and maintain long-term immunity. The current model suggests there is a single lineage of naive T cells that give rise to different populations of effector and memory T cells depending on the type and amounts of stimulation they encounter during infection. Here, we have discovered that multiple sub-populations of cells exist in the naive CD8+ T cell pool that are distinguished by their developmental origin, unique transcriptional profiles, distinct chromatin landscapes, and different kinetics and phenotypes after microbial challenge. These data demonstrate that the naive CD8+ T cell pool is not as homogeneous as previously thought and offers a new framework for explaining the remarkable heterogeneity in the effector and memory T cell subsets that arise after infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Genes, Developmental , Listeria monocytogenes/pathogenicity , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Line, Tumor , Chromatin/metabolism , Cytokines/pharmacology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Immunologic Memory , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Listeria monocytogenes/metabolism , Mice , Mice, Inbred C57BL , Principal Component Analysis , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/transplantation , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
4.
Nat Immunol ; 20(5): 652-662, 2019 05.
Article in English | MEDLINE | ID: mdl-30858620

ABSTRACT

αß T cell antigen receptors (TCRs) bind complexes of peptide and major histocompatibility complex (pMHC) with low affinity, which poses a considerable challenge for the direct identification of αß T cell cognate peptides. Here we describe a platform for the discovery of MHC class II epitopes based on the screening of engineered reporter cells expressing novel pMHC-TCR (MCR) hybrid molecules carrying cDNA-derived peptides. This technology identifies natural epitopes of CD4+ T cells in an unbiased and efficient manner and allows detailed analysis of TCR cross-reactivity that provides recognition patterns beyond discrete peptides. We determine the cognate peptides of virus- and tumor-specific T cells in mouse disease models and present a proof of concept for human T cells. Furthermore, we use MCR to identify immunogenic tumor neo-antigens and show that vaccination with a peptide naturally recognized by tumor-infiltrating lymphocytes efficiently protects mice from tumor challenge. Thus, the MCR technology holds promise for basic research and clinical applications, allowing the personalized identification of T cell-specific neo-antigens in patients.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , T-Cell Antigen Receptor Specificity/immunology , Amino Acid Sequence , Animals , Base Sequence , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Humans , Major Histocompatibility Complex/genetics , Mice, Inbred C57BL , Peptides/genetics , Peptides/immunology , Peptides/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism
5.
Immunity ; 54(3): 586-602.e8, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691136

ABSTRACT

To identify disease-relevant T cell receptors (TCRs) with shared antigen specificity, we analyzed 778,938 TCRß chain sequences from 178 non-small cell lung cancer patients using the GLIPH2 (grouping of lymphocyte interactions with paratope hotspots 2) algorithm. We identified over 66,000 shared specificity groups, of which 435 were clonally expanded and enriched in tumors compared to adjacent lung. The antigenic epitopes of one such tumor-enriched specificity group were identified using a yeast peptide-HLA A∗02:01 display library. These included a peptide from the epithelial protein TMEM161A, which is overexpressed in tumors and cross-reactive epitopes from Epstein-Barr virus and E. coli. Our findings suggest that this cross-reactivity may underlie the presence of virus-specific T cells in tumor infiltrates and that pathogen cross-reactivity may be a feature of multiple cancers. The approach and analytical pipelines generated in this work, as well as the specificity groups defined here, present a resource for understanding the T cell response in cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Epitope Mapping/methods , Epitopes, T-Lymphocyte/genetics , Lung Neoplasms/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology , Algorithms , Antigen Presentation , Antigens, Neoplasm/metabolism , Cells, Cultured , Cross Reactions , Epitopes, T-Lymphocyte/metabolism , HLA-A2 Antigen/metabolism , Humans , Protein Binding , T-Cell Antigen Receptor Specificity
6.
Immunity ; 53(6): 1245-1257.e5, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326767

ABSTRACT

Understanding the hallmarks of the immune response to SARS-CoV-2 is critical for fighting the COVID-19 pandemic. We assessed antibody and T cell reactivity in convalescent COVID-19 patients and healthy donors sampled both prior to and during the pandemic. Healthy donors examined during the pandemic exhibited increased numbers of SARS-CoV-2-specific T cells, but no humoral response. Their probable exposure to the virus resulted in either asymptomatic infection without antibody secretion or activation of preexisting immunity. In convalescent patients, we observed a public and diverse T cell response to SARS-CoV-2 epitopes, revealing T cell receptor (TCR) motifs with germline-encoded features. Bulk CD4+ and CD8+ T cell responses to the spike protein were mediated by groups of homologous TCRs, some of them shared across multiple donors. Overall, our results demonstrate that the T cell response to SARS-CoV-2, including the identified set of TCRs, can serve as a useful biomarker for surveying antiviral immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , Epitopes, T-Lymphocyte/metabolism , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/metabolism , Adolescent , Adult , Antibodies, Viral/metabolism , Asymptomatic Infections , Cells, Cultured , Convalescence , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunity , Immunologic Memory , Lymphocyte Activation , Male , Middle Aged , Pandemics , Receptors, Antigen, T-Cell/metabolism , Spike Glycoprotein, Coronavirus/immunology , Young Adult
7.
Semin Immunol ; 66: 101708, 2023 03.
Article in English | MEDLINE | ID: mdl-36621290

ABSTRACT

The identification of T-cell epitopes is key for a complete molecular understanding of immune recognition mechanisms in infectious diseases, autoimmunity and cancer. T-cell epitopes further provide targets for personalized vaccines and T-cell therapy, with several therapeutic applications in cancer immunotherapy and elsewhere. T-cell epitopes consist of short peptides displayed on Major Histocompatibility Complex (MHC) molecules. The recent advances in mass spectrometry (MS) based technologies to profile the ensemble of peptides displayed on MHC molecules - the so-called immunopeptidome - had a major impact on our understanding of antigen presentation and MHC ligands. On the one hand, these techniques enabled researchers to directly identify hundreds of thousands of peptides presented on MHC molecules, including some that elicited T-cell recognition. On the other hand, the data collected in these experiments revealed fundamental properties of antigen presentation pathways and significantly improved our ability to predict naturally presented MHC ligands and T-cell epitopes across the wide spectrum of MHC alleles found in human and other organisms. Here we review recent computational developments to analyze experimentally determined immunopeptidomes and harness these data to improve our understanding of antigen presentation and MHC binding specificities, as well as our ability to predict MHC ligands. We further discuss the strengths and limitations of the latest approaches to move beyond predictions of antigen presentation and tackle the challenges of predicting TCR recognition and immunogenicity.


Subject(s)
Epitopes, T-Lymphocyte , Neoplasms , Humans , Epitopes, T-Lymphocyte/metabolism , Ligands , Antigen Presentation , Peptides
8.
Immunity ; 47(1): 107-117.e8, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28709804

ABSTRACT

Regulatory T (Treg) cells expressing the transcription factor Foxp3 are critical for the prevention of autoimmunity and the suppression of anti-tumor immunity. The major self-antigens recognized by Treg cells remain undefined, representing a substantial barrier to the understanding of immune regulation. Here, we have identified natural Treg cell ligands in mice. We found that two recurrent Treg cell clones, one prevalent in prostate tumors and the other associated with prostatic autoimmune lesions, recognized distinct non-overlapping MHC-class-II-restricted peptides derived from the same prostate-specific protein. Notably, this protein is frequently targeted by autoantibodies in experimental models of prostatic autoimmunity. On the basis of these findings, we propose a model in which Treg cell responses at peripheral sites converge on those self-proteins that are most susceptible to autoimmune attack, and we suggest that this link could be exploited as a generalizable strategy for identifying the Treg cell antigens relevant to human autoimmunity.


Subject(s)
Autoantigens/metabolism , Epitopes, T-Lymphocyte/metabolism , Prostatic Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/physiology , Animals , Autoantibodies/metabolism , Autoantigens/genetics , Autoantigens/immunology , Cell Differentiation , Clone Cells , Epitope Mapping , Forkhead Transcription Factors/metabolism , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation , Male , Mice
9.
Bioinformatics ; 40(1)2024 01 02.
Article in English | MEDLINE | ID: mdl-38152987

ABSTRACT

MOTIVATION: The binding of a peptide antigen to a Class I major histocompatibility complex (MHC) protein is part of a key process that lets the immune system recognize an infected cell or a cancer cell. This mechanism enabled the development of peptide-based vaccines that can activate the patient's immune response to treat cancers. Hence, the ability of accurately predict peptide-MHC binding is an essential component for prioritizing the best peptides for each patient. However, peptide-MHC binding experimental data for many MHC alleles are still lacking, which limited the accuracy of existing prediction models. RESULTS: In this study, we presented an improved version of MHCSeqNet that utilized sub-word-level peptide features, a 3D structure embedding for MHC alleles, and an expanded training dataset to achieve better generalizability on MHC alleles with small amounts of data. Visualization of MHC allele embeddings confirms that the model was able to group alleles with similar binding specificity, including those with no peptide ligand in the training dataset. Furthermore, an external evaluation suggests that MHCSeqNet2 can improve the prioritization of T cell epitopes for MHC alleles with small amount of training data. AVAILABILITY AND IMPLEMENTATION: The source code and installation instruction for MHCSeqNet2 are available at https://github.com/cmb-chula/MHCSeqNet2.


Subject(s)
Histocompatibility Antigens Class I , Peptides , Humans , Alleles , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/chemistry , Peptides/chemistry , Protein Binding , Epitopes, T-Lymphocyte/metabolism
10.
Bioinformatics ; 39(12)2023 12 01.
Article in English | MEDLINE | ID: mdl-38070156

ABSTRACT

MOTIVATION: T cells play an essential role in adaptive immune system to fight pathogens and cancer but may also give rise to autoimmune diseases. The recognition of a peptide-MHC (pMHC) complex by a T cell receptor (TCR) is required to elicit an immune response. Many machine learning models have been developed to predict the binding, but generalizing predictions to pMHCs outside the training data remains challenging. RESULTS: We have developed a new machine learning model that utilizes information about the TCR from both α and ß chains, epitope sequence, and MHC. Our method uses ProtBERT embeddings for the amino acid sequences of both chains and the epitope, as well as convolution and multi-head attention architectures. We show the importance of each input feature as well as the benefit of including epitopes with only a few TCRs to the training data. We evaluate our model on existing databases and show that it compares favorably against other state-of-the-art models. AVAILABILITY AND IMPLEMENTATION: https://github.com/DaniTheOrange/EPIC-TRACE.


Subject(s)
Receptors, Antigen, T-Cell , T-Lymphocytes , Epitopes , Receptors, Antigen, T-Cell/chemistry , Amino Acid Sequence , T-Lymphocytes/metabolism , Protein Binding , Epitopes, T-Lymphocyte/metabolism
11.
Bioinformatics ; 39(11)2023 11 01.
Article in English | MEDLINE | ID: mdl-37847663

ABSTRACT

SUMMARY: T-cell receptors (TCRs) on T cells recognize and bind to epitopes presented by the major histocompatibility complex in case of an infection or cancer. However, the high diversity of TCRs, as well as their unique and complex binding mechanisms underlying epitope recognition, make it difficult to predict the binding between TCRs and epitopes. Here, we present the utility of transformers, a deep learning strategy that incorporates an attention mechanism that learns the informative features, and show that these models pre-trained on a large set of protein sequences outperform current strategies. We compared three pre-trained auto-encoder transformer models (ProtBERT, ProtAlbert, and ProtElectra) and one pre-trained auto-regressive transformer model (ProtXLNet) to predict the binding specificity of TCRs to 25 epitopes from the VDJdb database (human and murine). Two additional modifications were performed to incorporate gene usage of the TCRs in the four transformer models. Of all 12 transformer implementations (four models with three different modifications), a modified version of the ProtXLNet model could predict TCR-epitope pairs with the highest accuracy (weighted F1 score 0.55 simultaneously considering all 25 epitopes). The modification included additional features representing the gene names for the TCRs. We also showed that the basic implementation of transformers outperformed the previously available methods, i.e. TCRGP, TCRdist, and DeepTCR, developed for the same biological problem, especially for the hard-to-classify labels. We show that the proficiency of transformers in attention learning can be made operational in a complex biological setting like TCR binding prediction. Further ingenuity in utilizing the full potential of transformers, either through attention head visualization or introducing additional features, can extend T-cell research avenues. AVAILABILITY AND IMPLEMENTATION: Data and code are available on https://github.com/InduKhatri/tcrformer.


Subject(s)
Epitopes, T-Lymphocyte , Receptors, Antigen, T-Cell , Humans , Animals , Mice , Epitopes, T-Lymphocyte/metabolism , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/metabolism , Amino Acid Sequence , Major Histocompatibility Complex
12.
PLoS Pathog ; 18(6): e1010403, 2022 06.
Article in English | MEDLINE | ID: mdl-35737741

ABSTRACT

There is now convincing evidence that the successful development of an effective CMV vaccine will require improved formulation and adjuvant selection that is capable of inducing both humoral and cellular immune responses. Here, we have designed a novel bivalent subunit vaccine formulation based on CMV-encoded oligomeric glycoprotein B (gB) and polyepitope protein in combination with human compatible TLR9 agonist CpG1018. The polyepitope protein includes multiple minimal HLA class I-restricted CD8+ T cell epitopes from different antigens of CMV. This subunit vaccine generated durable anti-viral antibodies, CMV-specific CD4+ and CD8+ T cell responses in multiple HLA expressing mice. Antibody responses included broad TH1 isotypes (IgG2a, IgG2b and IgG3) and potently neutralized CMV infection in fibroblasts and epithelial cells. Furthermore, polyfunctional antigen-specific T cell immunity and antiviral antibody responses showed long-term memory maintenance. These observations argue that this novel vaccine strategy, if applied to humans, could facilitate the generation of robust humoral and cellular immune responses which may be more effective in preventing CMV-associated complications in various clinical settings.


Subject(s)
Cytomegalovirus Infections , Immunity, Humoral , Adjuvants, Immunologic , Animals , Antibodies, Viral , CD8-Positive T-Lymphocytes , Epitopes, T-Lymphocyte/metabolism , Humans , Immunity, Cellular , Mice , Oligodeoxyribonucleotides , Toll-Like Receptor 9/metabolism , Vaccines, Combined , Vaccines, Subunit
13.
Circ Res ; 131(3): 258-276, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35766025

ABSTRACT

BACKGROUND: CD (cluster of differentiation) 4+ T-cell responses to APOB (apolipoprotein B) are well characterized in atherosclerotic mice and detectable in humans. CD4+ T cells recognize antigenic peptides displayed on highly polymorphic HLA (human leukocyte antigen)-II. Immunogenicity of individual APOB peptides is largely unknown in humans. Only 1 HLA-II-restricted epitope was validated using the DRB1*07:01-APOB3036-3050 tetramer. We hypothesized that human APOB may contain discrete immunodominant CD4+ T-cell epitopes that trigger atherosclerosis-related autoimmune responses in donors with diverse HLA alleles. METHODS: We selected 20 APOB-derived peptides (APOB20) from an in silico screen and experimentally validated binding to the most commonly occurring human HLA-II alleles. We optimized a restimulation-based workflow to evaluate antigenicity of multiple candidate peptides in HLA-typed donors. This included activation-induced marker assay, intracellular cytokine staining, IFNγ (interferon gamma) enzyme-linked immunospot and cytometric bead array. High-throughput sequencing revealed TCR (T-cell receptor) clonalities of APOB-reactive CD4+ T cells. RESULTS: Using stringent positive, negative, and crossover stimulation controls, we confirmed specificity of expansion-based protocols to detect CD4+ T cytokine responses to the APOB20 pool. Ex vivo assessment of AIM+CD4+ T cells revealed a statistically significant autoimmune response to APOB20 but not to a ubiquitously expressed negative control protein, actin. Resolution of CD4+ T responses to the level of individual peptides using IFNγ enzyme-linked immunospot led to the discovery of 6 immunodominant epitopes (APOB6) that triggered robust CD4+ T activation in most donors. APOB6-specific responding CD4+ T cells were enriched in unique expanded TCR clonotypes and preferentially expressed memory markers. Cytometric bead array analysis detected APOB6-induced secretion of both proinflammatory and regulatory cytokines. In clinical samples from patients with angiographically verified coronary artery disease, APOB6 stimulation induced higher activation and memory phenotypes and augmented secretion of proinflammatory cytokines TNF (tumor necrosis factor) and IFNγ, compared with patients with low coronary artery disease. CONCLUSIONS: Using 3 cohorts, each with ≈20 donors, we discovered and validated 6 immunodominant, HLA-II-restricted APOB epitopes. The immune response to these APOB epitopes correlated with coronary artery disease severity.


Subject(s)
Coronary Artery Disease , Animals , Apolipoproteins B/metabolism , CD4-Positive T-Lymphocytes , Coronary Artery Disease/metabolism , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Humans , Interferon-gamma/metabolism , Major Histocompatibility Complex , Mice , Peptides/genetics
14.
J Immunol ; 208(1): 3-15, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34880106

ABSTRACT

CD8+ T cells play an important role in the control of untreated HIV infection. Several studies have suggested a decisive role of TCRs involved in anti-HIV immunity. HLA-B*27 and B*57 are often associated with a delayed HIV disease progression, but the exact correlates that provide superior immunity against HIV are not known. To investigate if the T cell repertoire underlies the protective effect in disease outcome in HLA-B*27 and B*57+ individuals, we analyzed Ag-specific TCR profiles from progressors (n = 13) and slow progressors (n = 11) expressing either B*27 or B*57. Our data showed no differences in TCR diversity between progressors and slow progressors. Both alleles recruit biased T cell repertoires (i.e., TCR populations skewed toward specific TRBV families or CDR3 regions). This bias was unrelated to disease progression and was remarkably profound for HLA-B*57, in which TRBV family usage and CDR3 sequences were shared to some extent even between epitopes. Conclusively, these data suggest that the T cell repertoires recruited by protective HLA alleles are highly similar between progressors and slow progressors in terms of TCR diversity, TCR usage, and cross-reactivity.


Subject(s)
Complementarity Determining Regions/genetics , HIV Infections/immunology , HIV-1/physiology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/physiology , Alleles , Antigen Presentation , Antigens, Viral/metabolism , Cells, Cultured , Cross Reactions , Disease Progression , Enzyme-Linked Immunospot Assay , Epitopes, T-Lymphocyte/metabolism , Genetic Variation , HLA-B Antigens/genetics , HLA-B Antigens/metabolism , HLA-B27 Antigen/genetics , HLA-B27 Antigen/metabolism , Humans , Lymphocyte Activation
15.
Semin Immunol ; 50: 101418, 2020 08.
Article in English | MEDLINE | ID: mdl-33131981

ABSTRACT

Epitopes, in the context of T cell recognition, are short peptides typically derived by antigen processing, and presented on the cell surface bound to MHC molecules (HLA molecules in humans) for TCR scrutiny. The identification of epitopes is a context-dependent process, with consideration given to, for example, the source pathogen and protein, the host organism, and state of the immune reaction (e.g., following natural infection, vaccination, etc.). In the following review, we consider the various approaches used to define T cell epitopes, including both bioinformatic and experimental approaches, and discuss the concepts of immunodominance and immunoprevalence. We also discuss HLA polymorphism and epitope restriction, and the resulting impact on the identification of, and potential population coverage afforded by, epitopes or epitope-based vaccines. Finally, some examples of the practical application of T cell epitope identification are provided, showing how epitopes have been valuable for deriving novel immunological insights in the context of the immune response to various pathogens and allergens.


Subject(s)
Epitope Mapping/methods , Epitopes, T-Lymphocyte/genetics , Immunodominant Epitopes/metabolism , T-Lymphocytes/immunology , Vaccines/immunology , Animals , Computational Biology , Epitopes, T-Lymphocyte/metabolism , HLA Antigens/genetics , HLA Antigens/metabolism , Humans , Immunoassay , Immunodominant Epitopes/genetics , Polymorphism, Genetic , Protein Binding
16.
Biochemistry ; 62(17): 2517-2529, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37554055

ABSTRACT

Antigen conformation shapes CD4+ T-cell specificity through mechanisms of antigen processing, and the consequences for immunity may rival those from conformational effects on antibody specificity. CD4+ T cells initiate and control immunity to pathogens and cancer and are at least partly responsible for immunopathology associated with infection, autoimmunity, and allergy. The primary trigger for CD4+ T-cell maturation is the presentation of an epitope peptide in the MHC class II antigen-presenting protein (MHCII), most commonly on an activated dendritic cell, and then the T-cell responses are recalled by subsequent presentations of the epitope peptide by the same or other antigen-presenting cells. Peptide presentation depends on the proteolytic fragmentation of the antigen in an endosomal/lysosomal compartment and concomitant loading of the fragments into the MHCII, a multistep mechanism called antigen processing and presentation. Although the role of peptide affinity for MHCII has been well studied, the role of proteolytic fragmentation has received less attention. In this Perspective, we will briefly summarize evidence that antigen resistance to unfolding and proteolytic fragmentation shapes the specificity of the CD4+ T-cell response to selected viral envelope proteins, identify several remarkable examples in which the immunodominant CD4+ epitopes most likely depend on the interaction of processing machinery with antigen conformation, and outline how knowledge of antigen conformation can inform future efforts to design vaccines.


Subject(s)
CD4-Positive T-Lymphocytes , Epitopes, T-Lymphocyte , CD4-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/metabolism , Viral Fusion Proteins/metabolism , Histocompatibility Antigens Class II/metabolism , Antigen Presentation , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/metabolism
17.
J Virol ; 96(18): e0116621, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36069549

ABSTRACT

Studies on Ebola virus disease (EVD) survivors and clinical studies on Ebola virus (EBOV) vaccine candidates have pinpointed the importance of a strong antibody response in protection and survival from EBOV infection. However, little is known about the T cell responses to EBOV or EBOV vaccines. We used HLA-A*02:01 (HLA-A2) transgenic mice to study HLA-A2-specific T cell responses elicited following vaccination with EBOV glycoprotein (EBOV-GP) presented with three different systems: (i) recombinant protein (rEBOV-GP), (ii) vesicular stomatitis replication-competent recombinant virus (VSV-EBOV-GP), and (iii) modified vaccinia Ankara virus recombinant (MVA-EBOV-GP). T cells from immunized animals were analyzed using peptide pools representing the entire GP region and individual peptides. Regardless of the vaccine formulation, we identified a minimal 9mer epitope containing an HLA-A2 motif (FLDPATTS), which was confirmed through HLA-A2 binding affinity and immunization studies. Using binding prediction software, we identified substitutions surrounding position 9 (S9V, P10V, and Q11V) that predicted enhanced binding to the HLA-A2 molecule. This enhanced binding was confirmed through in vitro binding studies and enhanced potency was shown with in vivo immunization studies using the enhanced sequences and the wild-type sequence. Of note, in silico studies predicted the enhanced 9mer epitope carrying the S9V substitution as the best overall HLA-A2 epitope for the full-length EBOV-GP. These results suggest that EBOV-GP-S9V and EBOV-GP-P10V represent more potent in vivo immunogens. Identification and enhancement of EBOV-specific human HLA epitopes could lead to the development of tools and reagents to induce more robust T cell responses in human subjects. IMPORTANCE Vaccine efficacy and immunity to viral infection are often measured by neutralizing antibody titers. T cells are specialized subsets of immune cells with antiviral activity, but this response is variable and difficult to track. We showed that the HLA-A2-specific T cell response to the Ebola virus glycoprotein can be enhanced significantly by a single residue substitution designed to improve an epitope binding affinity to one of the most frequent MHC alleles in the human population. This strategy could be applied to improve T cell responses to Ebola vaccines designed to elicit antibodies and adapted to target MHC alleles of populations in regions where endemic infections, like Ebola virus disease, are still causing outbreaks with concerning pandemic potential.


Subject(s)
Amino Acids , Ebolavirus , Epitopes, T-Lymphocyte , Glycoproteins , Hemorrhagic Fever, Ebola , Amino Acids/metabolism , Animals , Antibodies, Neutralizing , Antibodies, Viral , Ebola Vaccines/genetics , Ebolavirus/genetics , Epitopes, T-Lymphocyte/metabolism , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Mice , Recombinant Proteins , Vaccinia virus , Vesiculovirus
18.
Nat Immunol ; 12(1): 45-53, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21151101

ABSTRACT

Cytotoxic T lymphocytes (CTLs) recognize peptides presented by HLA class I molecules on the cell surface. The C terminus of these CTL epitopes is considered to be produced by the proteasome. Here we demonstrate that the cytosolic endopeptidases nardilysin and thimet oligopeptidase (TOP) complemented proteasome activity. Nardilysin and TOP were required, either together or alone, for the generation of a tumor-specific CTL epitope from PRAME, an immunodominant CTL epitope from Epstein-Barr virus protein EBNA3C, and a clinically important epitope from the melanoma protein MART-1. TOP functioned as C-terminal trimming peptidase in antigen processing, and nardilysin contributed to both the C-terminal and N-terminal generation of CTL epitopes. By broadening the antigenic peptide repertoire, nardilysin and TOP strengthen the immune defense against intracellular pathogens and cancer.


Subject(s)
Antigens, Neoplasm/metabolism , Epitopes, T-Lymphocyte/metabolism , Metalloendopeptidases/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Antigen Presentation/genetics , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , HLA-A3 Antigen/metabolism , Humans , K562 Cells , Metalloendopeptidases/genetics , Metalloendopeptidases/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Peptide Fragments/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , RNA, Small Interfering/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Transgenes/genetics
19.
J Immunol ; 207(8): 2051-2059, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34526376

ABSTRACT

Ag-specific immunotherapy to restore immune tolerance to self-antigens, without global immune suppression, is a long-standing goal in the treatment of autoimmune disorders such as type 1 diabetes (T1D). However, vaccination with autoantigens such as insulin or glutamic acid decarboxylase have largely failed in human T1D trials. Induction and maintenance of peripheral tolerance by vaccination requires efficient autoantigen presentation by APCs. In this study, we show that a lipophilic modification at the N-terminal end of CD4+ epitopes (lipo-peptides) dramatically improves peptide Ag presentation. We designed amphiphilic lipo-peptides to efficiently target APCs in the lymph nodes by binding and trafficking with endogenous albumin. Additionally, we show that lipophilic modification anchors the peptide into the membranes of APCs, enabling a bivalent cell-surface Ag presentation. The s.c. injected lipo-peptide accumulates in the APCs in the lymph node, enhances the potency and duration of peptide Ag presentation by APCs, and induces Ag-specific immune tolerance that controls both T cell- and B cell-mediated immunity. Immunization with an amphiphilic insulin B chain 9-23 peptide, an immunodominant CD4+ T cell epitope in NOD mice, significantly suppresses the activation of T cells, increases inhibitory cytokine production, induces regulatory T cells, and delays the onset and lowers the incidence of T1D. Importantly, treatment with a lipophilic ß-cell peptide mixture delays progression to end-stage diabetes in acutely diabetic NOD mice, whereas the same doses of standard soluble peptides were not effective. Amphiphilic modification effectively enhances Ag presentation for peptide-based immune regulation of autoimmune diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/metabolism , Insulin/metabolism , Peptide Fragments/metabolism , Surface-Active Agents/metabolism , Albumins , Animals , Antigen Presentation , Female , Humans , Immune Tolerance , Immunization , Immunomodulation , Insulin/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Peptide Fragments/immunology
20.
J Immunol ; 206(8): 1709-1718, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33762324

ABSTRACT

Studies of immune responses elicited by bovine viral diarrhea virus (BVDV) vaccines have primarily focused on the characterization of neutralizing B cell and CD4+ T cell epitopes. Despite the availability of commercial vaccines for decades, BVDV prevalence in cattle has remained largely unaffected. There is limited knowledge regarding the role of BVDV-specific CD8+ T cells in immune protection, and indirect evidence suggests that they play a crucial role during BVDV infection. In this study, the presence of BVDV-specific CD8+ T cells that are highly cross-reactive in cattle was demonstrated. Most importantly, novel potent IFN-γ-inducing CD8+ T cell epitopes were identified from different regions of BVDV polyprotein. Eight CD8+ T cell epitopes were identified from the following structural BVDV Ags: Erns, E1, and E2 glycoproteins. In addition, from nonstructural BVDV Ags Npro, NS2-3, NS4A-B, and NS5A-B, 20 CD8+ T cell epitopes were identified. The majority of these IFN-γ-inducing CD8+ T cell epitopes were found to be highly conserved among more than 200 strains from BVDV-1 and -2 genotypes. These conserved epitopes were also validated as cross-reactive because they induced high recall IFN-γ+CD8+ T cell responses ex vivo in purified bovine CD8+ T cells isolated from BVDV-1- and -2-immunized cattle. Altogether, 28 bovine MHC class I-binding epitopes were identified from key BVDV Ags that can elicit broadly reactive CD8+ T cells against diverse BVDV strains. The data presented in this study will lay the groundwork for the development of a contemporary CD8+ T cell-based BVDV vaccine capable of addressing BVDV heterogeneity more effectively than current vaccines.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/immunology , CD8-Positive T-Lymphocytes/immunology , Diarrhea Viruses, Bovine Viral/physiology , Epitopes, T-Lymphocyte/metabolism , Viral Envelope Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Viral Vaccines/immunology , Animals , Cattle , Cells, Cultured , Conserved Sequence/genetics , Cross Reactions , Epitopes, T-Lymphocyte/genetics , Histocompatibility Antigens Class I/metabolism , Interferon-gamma/metabolism , Protein Binding , Viral Envelope Proteins/genetics , Viral Nonstructural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL