1
|
Castorina LV, Grazioli F, Machart P, Mösch A, Errica F. Assessing the generalization capabilities of TCR binding predictors via peptide distance analysis. PLoS One 2025; 20:e0324011. [PMID: 40392871 PMCID: PMC12091837 DOI: 10.1371/journal.pone.0324011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/19/2025] [Indexed: 05/22/2025] Open
Abstract
Understanding the interaction between T Cell Receptors (TCRs) and peptide-bound Major Histocompatibility Complexes (pMHCs) is crucial for comprehending immune responses and developing targeted immunotherapies. While recent machine learning (ML) models show remarkable success in predicting TCR-pMHC binding within training data, these models often fail to generalize to peptides outside their training distributions, raising concerns about their applicability in therapeutic settings. Understanding and improving the generalization of these models is therefore critical to ensure real-world applications. To address this issue, we evaluate the effect of the distance between training and testing peptide distributions on ML model empirical risk assessments, using sequence-based and 3D structure-based distance metrics. In our analysis we use several state-of-the-art models for TCR-peptide binding prediction: Attentive Variational Information Bottleneck (AVIB), NetTCR-2.0 and -2.2, and ERGO II (pre-trained autoencoder) and ERGO II (LSTM). In this work, we introduce a novel approach for assessing the generalization capabilities of TCR binding predictors: the Distance Split (DS) algorithm. The DS algorithm controls the distance between training and testing peptides based on both sequence and structure, allowing for a more nuanced evaluation of model performance. We show that lower 3D shape similarity between training and test peptides is associated with a harder out-of-distribution task definition, which is more interesting when measuring the ability to generalize to unseen peptides. However, we observe the opposite effect when splitting using sequence-based similarity. These findings highlight the importance of using a distance-based splitting approach to benchmark models. This could then be used to estimate a confidence score on predictions on novel and unseen peptides, based on how different they are from the training ones. Additionally, our results may hint that employing 3D shape to complement sequence information could improve the accuracy of TCR-pMHC binding predictors.
Collapse
Affiliation(s)
- Leonardo V. Castorina
- School of Informatics, University of Edinburgh, Edinburgh, United Kingdom
- NEC Laboratories Europe, Heidelberg, Germany
| | | | | | - Anja Mösch
- NEC Laboratories Europe, Heidelberg, Germany
| | | |
Collapse
|
2
|
Morgan J, Lindsay AE. Modulation of antigen discrimination by duration of immune contacts in a kinetic proofreading model of T cell activation with extreme statistics. PLoS Comput Biol 2023; 19:e1011216. [PMID: 37647345 PMCID: PMC10497171 DOI: 10.1371/journal.pcbi.1011216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/12/2023] [Accepted: 08/05/2023] [Indexed: 09/01/2023] Open
Abstract
T cells form transient cell-to-cell contacts with antigen presenting cells (APCs) to facilitate surface interrogation by membrane bound T cell receptors (TCRs). Upon recognition of molecular signatures (antigen) of pathogen, T cells may initiate an adaptive immune response. The duration of the T cell/APC contact is observed to vary widely, yet it is unclear what constructive role, if any, such variations might play in immune signaling. Modeling efforts describing antigen discrimination often focus on steady-state approximations and do not account for the transient nature of cellular contacts. Within the framework of a kinetic proofreading (KP) mechanism, we develop a stochastic First Receptor Activation Model (FRAM) describing the likelihood that a productive immune signal is produced before the expiry of the contact. Through the use of extreme statistics, we characterize the probability that the first TCR triggering is induced by a rare agonist antigen and not by that of an abundant self-antigen. We show that defining positive immune outcomes as resilience to extreme statistics and sensitivity to rare events mitigates classic tradeoffs associated with KP. By choosing a sufficient number of KP steps, our model is able to yield single agonist sensitivity whilst remaining non-reactive to large populations of self antigen, even when self and agonist antigen are similar in dissociation rate to the TCR but differ largely in expression. Additionally, our model achieves high levels of accuracy even when agonist positive APCs encounters are rare. Finally, we discuss potential biological costs associated with high classification accuracy, particularly in challenging T cell environments.
Collapse
Affiliation(s)
- Jonathan Morgan
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, South Bend, Indiana, United States of America
- Biophysics Graduate Program, University of Notre Dame, South Bend, Indiana, United States of America
| | - Alan E. Lindsay
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, South Bend, Indiana, United States of America
| |
Collapse
|
3
|
Lantz O, Teyton L. Identification of T cell antigens in the 21st century, as difficult as ever. Semin Immunol 2022; 60:101659. [PMID: 36183497 PMCID: PMC10332289 DOI: 10.1016/j.smim.2022.101659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Identifying antigens recognized by T cells is still challenging, particularly for innate like T cells that do not recognize peptides but small metabolites or lipids in the context of MHC-like molecules or see non-MHC restricted antigens. The fundamental reason for this situation is the low affinity of T cell receptors for their ligands coupled with a level of degeneracy that makes them bind to similar surfaces on antigen presenting cells. Herein we will describe non-exhaustively some of the methods that were used to identify peptide antigens and briefly mention the high throughput methods more recently proposed for that purpose. We will then present how the molecules recognized by innate like T cells (NKT, MAIT and γδ T cells) were discovered. We will show that serendipity was instrumental in many cases.
Collapse
Affiliation(s)
- Olivier Lantz
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France; Laboratoire d'Immunologie Clinique, Institut Curie, Paris 75005, France; Centre d'investigation Clinique en Biothérapie Gustave-Roussy Institut Curie (CIC-BT1428) Institut Curie, Paris 75005, France
| | - Luc Teyton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
4
|
Abstract
T cells recognize and respond to self antigens in both cancer and autoimmunity. One strategy to influence this response is to incorporate amino acid substitutions into these T cell-specific epitopes. This strategy is being reconsidered now with the goal of increasing time to regression with checkpoint blockade therapies in cancer and antigen-specific immunotherapies in autoimmunity. We discuss how these amino acid substitutions change the interactions with the MHC class I or II molecule and the responding T cell repertoire. Amino acid substitutions in epitopes that are the most effective in therapies bind more strongly to T cell receptor and/or MHC molecules and cross-react with the same repertoire of T cells as the natural antigen.
Collapse
Affiliation(s)
- Jill E Slansky
- Department of Immunology and Microbiology, University of Colorado School of Medicine, 12800 E. 19thAvenue, Aurora, CO 80045, USA.
| | - Maki Nakayama
- Department of Immunology and Microbiology, University of Colorado School of Medicine, 12800 E. 19thAvenue, Aurora, CO 80045, USA; Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, 1775 Aurora Court, Aurora, CO 80045, USA
| |
Collapse
|
5
|
Dahal-Koirala S, Ciacchi L, Petersen J, Risnes LF, Neumann RS, Christophersen A, Lundin KEA, Reid HH, Qiao SW, Rossjohn J, Sollid LM. Discriminative T-cell receptor recognition of highly homologous HLA-DQ2-bound gluten epitopes. J Biol Chem 2018; 294:941-952. [PMID: 30455354 DOI: 10.1074/jbc.ra118.005736] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/05/2018] [Indexed: 12/13/2022] Open
Abstract
Celiac disease (CeD) provides an opportunity to study the specificity underlying human T-cell responses to an array of similar epitopes presented by the same human leukocyte antigen II (HLA-II) molecule. Here, we investigated T-cell responses to the two immunodominant and highly homologous HLA-DQ2.5-restricted gluten epitopes, DQ2.5-glia-α1a (PFPQPELPY) and DQ2.5-glia-ω1 (PFPQPEQPF). Using HLA-DQ2.5-DQ2.5-glia-α1a and HLA-DQ2.5-DQ2.5-glia-ω1 tetramers and single-cell αβ T-cell receptor (TCR) sequencing, we observed that despite similarity in biased variable-gene usage in the TCR repertoire responding to these nearly identical peptide-HLA-II complexes, most of the T cells are specific for either of the two epitopes. To understand the molecular basis of this exquisite fine specificity, we undertook Ala substitution assays revealing that the p7 residue (Leu/Gln) is critical for specific epitope recognition by both DQ2.5-glia-α1a- and DQ2.5-glia-ω1-reactive T-cell clones. We determined high-resolution binary crystal structures of HLA-DQ2.5 bound to DQ2.5-glia-α1a (2.0 Å) and DQ2.5-glia-ω1 (2.6 Å). These structures disclosed that differences around the p7 residue subtly alter the neighboring substructure and electrostatic properties of the HLA-DQ2.5-peptide complex, providing the fine specificity underlying the responses against these two highly homologous gluten epitopes. This study underscores the ability of TCRs to recognize subtle differences in the peptide-HLA-II landscape in a human disease setting.
Collapse
Affiliation(s)
- Shiva Dahal-Koirala
- From the Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway.,the K. G. Jebsen Centre for Coeliac Disease Research, University of Oslo, 0424 Oslo, Norway
| | - Laura Ciacchi
- the Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute and.,the Australian Research Council (ARC) Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Jan Petersen
- the Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute and.,the Australian Research Council (ARC) Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Louise Fremgaard Risnes
- From the Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway.,the K. G. Jebsen Centre for Coeliac Disease Research, University of Oslo, 0424 Oslo, Norway
| | - Ralf Stefan Neumann
- the K. G. Jebsen Centre for Coeliac Disease Research, University of Oslo, 0424 Oslo, Norway
| | - Asbjørn Christophersen
- the K. G. Jebsen Centre for Coeliac Disease Research, University of Oslo, 0424 Oslo, Norway
| | - Knut E A Lundin
- the K. G. Jebsen Centre for Coeliac Disease Research, University of Oslo, 0424 Oslo, Norway.,the Department of Gastroenterology, Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway, and
| | - Hugh H Reid
- the Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute and.,the Australian Research Council (ARC) Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Shuo-Wang Qiao
- From the Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway.,the K. G. Jebsen Centre for Coeliac Disease Research, University of Oslo, 0424 Oslo, Norway
| | - Jamie Rossjohn
- the Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute and .,the Australian Research Council (ARC) Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia.,the Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, Wales, United Kingdom
| | - Ludvig M Sollid
- From the Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway, .,the K. G. Jebsen Centre for Coeliac Disease Research, University of Oslo, 0424 Oslo, Norway
| |
Collapse
|
6
|
Hart DP, Uzun N, Skelton S, Kakoschke A, Househam J, Moss DS, Shepherd AJ. Factor VIII cross-matches to the human proteome reduce the predicted inhibitor risk in missense mutation hemophilia A. Haematologica 2018; 104:599-608. [PMID: 30266735 PMCID: PMC6395325 DOI: 10.3324/haematol.2018.195669] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 09/27/2018] [Indexed: 12/27/2022] Open
Abstract
Single missense mutations in the F8 gene encoding the coagulation protein factor VIII give rise predominantly to non-severe hemophilia A. Despite only a single amino acid sequence difference between the replacement, therapeutic factor VIII and the patient's endogenous factor VIII, therapeutic factor VIII may still be perceived as foreign by the recipient's immune system and trigger an immune response (inhibitor). Inhibitor formation is a life-long risk for patients with non-severe hemophilia A treated with therapeutic factor VIII, but remains difficult to predict. The aim of this study was to understand whether fortuitous, primary sequence cross-matches between therapeutic factor VIII and proteins in the human proteome are the reason why certain F8 mutations are not associated with inhibitor formation. We predicted which therapeutic factor VIII differences are potentially perceived as foreign by helper T cells - a necessary precursor to inhibitor development - and then scanned potentially immunogenic peptides against more than 100,000 proteins in the proteome. As there are hundreds of disease-causing F8 missense mutations and the human leukocyte antigen gene complex governing peptide presentation to helper T cells is highly polymorphic, these calculations pose a huge combinatorial challenge that we addressed computationally. We found that cross-matches between therapeutic factor VIII and the human proteome are commonplace and have a profound impact on the predicted risk of inhibitor development. Our results emphasize the importance of knowing both the F8 missense mutation and the human leukocyte antigen alleles of a patient with missense mutation hemophilia A if his underlying risk of inhibitor development is to be estimated.
Collapse
Affiliation(s)
- Daniel P Hart
- Blizard Institute, Barts and The London School of Medicine and Dentistry, QMUL, London.,The Royal London Hospital Haemophilia Centre, Barts Health NHS Trust, London
| | - Nazmiye Uzun
- Department of Biological Sciences and Institute of Structural and Molecular Biology, Birkbeck, University of London, UK
| | - Stuart Skelton
- Blizard Institute, Barts and The London School of Medicine and Dentistry, QMUL, London.,Department of Biological Sciences and Institute of Structural and Molecular Biology, Birkbeck, University of London, UK
| | - Alison Kakoschke
- Department of Biological Sciences and Institute of Structural and Molecular Biology, Birkbeck, University of London, UK
| | - Jacob Househam
- Department of Biological Sciences and Institute of Structural and Molecular Biology, Birkbeck, University of London, UK
| | - David S Moss
- Department of Biological Sciences and Institute of Structural and Molecular Biology, Birkbeck, University of London, UK
| | - Adrian J Shepherd
- Department of Biological Sciences and Institute of Structural and Molecular Biology, Birkbeck, University of London, UK
| |
Collapse
|
7
|
Campos-García VR, Herrera-Fernández D, Espinosa-de la Garza CE, González G, Vallejo-Castillo L, Avila S, Muñoz-García L, Medina-Rivero E, Pérez NO, Gracia-Mora I, Pérez-Tapia SM, Salazar-Ceballos R, Pavón L, Flores-Ortiz LF. Process signatures in glatiramer acetate synthesis: structural and functional relationships. Sci Rep 2017; 7:12125. [PMID: 28935954 PMCID: PMC5608765 DOI: 10.1038/s41598-017-12416-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/04/2017] [Indexed: 12/14/2022] Open
Abstract
Glatiramer Acetate (GA) is an immunomodulatory medicine approved for the treatment of multiple sclerosis, whose mechanisms of action are yet to be fully elucidated. GA is comprised of a complex mixture of polypeptides with different amino acid sequences and structures. The lack of sensible information about physicochemical characteristics of GA has contributed to its comprehensiveness complexity. Consequently, an unambiguous determination of distinctive attributes that define GA is of highest relevance towards dissecting its identity. Herein we conducted a study of characteristic GA heterogeneities throughout its manufacturing process (process signatures), revealing a strong impact of critical process parameters (CPPs) on the reactivity of amino acid precursors; reaction initiation and polymerization velocities; and peptide solubility, susceptibility to hydrolysis, and size-exclusion properties. Further, distinctive GA heterogeneities were correlated to defined immunological and toxicological profiles, revealing that GA possesses a unique repertoire of active constituents (epitopes) responsible of its immunological responses, whose modification lead to altered profiles. This novel approach established CPPs influence on intact GA peptide mixture, whose physicochemical identity cannot longer rely on reduced properties (based on complete or partial GA degradation), providing advanced knowledge on GA structural and functional relationships to ensure a consistent manufacturing of safe and effective products.
Collapse
Affiliation(s)
- Víctor R Campos-García
- Unidad de Investigación y Desarrollo, Probiomed S.A. de C.V., Cruce de Carreteras Acatzingo-Zumpahuacán s/n, Colonia Los Shiperes, Tenancingo, 52400, Estado de México, Mexico
| | - Daniel Herrera-Fernández
- Unidad de Investigación y Desarrollo, Probiomed S.A. de C.V., Cruce de Carreteras Acatzingo-Zumpahuacán s/n, Colonia Los Shiperes, Tenancingo, 52400, Estado de México, Mexico
| | - Carlos E Espinosa-de la Garza
- Unidad de Investigación y Desarrollo, Probiomed S.A. de C.V., Cruce de Carreteras Acatzingo-Zumpahuacán s/n, Colonia Los Shiperes, Tenancingo, 52400, Estado de México, Mexico
| | - German González
- Unidad de Investigación y Desarrollo, Probiomed S.A. de C.V., Cruce de Carreteras Acatzingo-Zumpahuacán s/n, Colonia Los Shiperes, Tenancingo, 52400, Estado de México, Mexico
| | - Luis Vallejo-Castillo
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala s/n, Colonia Santo Tomás, 11340, Ciudad de México, Mexico
- Departamento de Farmacología, Cinvestav-IPN, Avenida Instituto Politécnico Nacional 2508, Colonia San Pedro Zacatenco, 07360, Ciudad de México, Mexico
| | - Sandra Avila
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala s/n, Colonia Santo Tomás, 11340, Ciudad de México, Mexico
| | - Leslie Muñoz-García
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala s/n, Colonia Santo Tomás, 11340, Ciudad de México, Mexico
| | - Emilio Medina-Rivero
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala s/n, Colonia Santo Tomás, 11340, Ciudad de México, Mexico
| | - Néstor O Pérez
- Unidad de Investigación y Desarrollo, Probiomed S.A. de C.V., Cruce de Carreteras Acatzingo-Zumpahuacán s/n, Colonia Los Shiperes, Tenancingo, 52400, Estado de México, Mexico
| | - Isabel Gracia-Mora
- Departamento de Quı́mica Inorgánica y Nuclear, Facultad de Quı́mica, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, Investigación Científica 70, 04510, Ciudad de México, Mexico
| | - Sonia Mayra Pérez-Tapia
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala s/n, Colonia Santo Tomás, 11340, Ciudad de México, Mexico
- Unidad de Investigación, Desarrollo e Innovación Médica y Biotecnológica (UDIMEB), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala s/n, Colonia Santo Tomás, 11340, Ciudad de México, Mexico
| | - Rodolfo Salazar-Ceballos
- Unidad de Investigación y Desarrollo, Probiomed S.A. de C.V., Cruce de Carreteras Acatzingo-Zumpahuacán s/n, Colonia Los Shiperes, Tenancingo, 52400, Estado de México, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, 14370, Ciudad de México, Mexico
| | - Luis F Flores-Ortiz
- Unidad de Investigación y Desarrollo, Probiomed S.A. de C.V., Cruce de Carreteras Acatzingo-Zumpahuacán s/n, Colonia Los Shiperes, Tenancingo, 52400, Estado de México, Mexico.
| |
Collapse
|
8
|
Contractile actomyosin arcs promote the activation of primary mouse T cells in a ligand-dependent manner. PLoS One 2017; 12:e0183174. [PMID: 28817635 PMCID: PMC5560663 DOI: 10.1371/journal.pone.0183174] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/31/2017] [Indexed: 12/16/2022] Open
Abstract
Mechano-transduction is an emerging but still poorly understood component of T cell activation. Here we investigated the ligand-dependent contribution made by contractile actomyosin arcs populating the peripheral supramolecular activation cluster (pSMAC) region of the immunological synapse (IS) to T cell receptor (TCR) microcluster transport and proximal signaling in primary mouse T cells. Using super resolution microscopy, OT1-CD8+ mouse T cells, and two ovalbumin (OVA) peptides with different affinities for the TCR, we show that the generation of organized actomyosin arcs depends on ligand potency and the ability of myosin 2 to contract actin filaments. While weak ligands induce disorganized actomyosin arcs, strong ligands result in organized actomyosin arcs that correlate well with tension-sensitive CasL phosphorylation and the accumulation of ligands at the IS center. Blocking myosin 2 contractility greatly reduces the difference in the extent of Src and LAT phosphorylation observed between the strong and the weak ligand, arguing that myosin 2-dependent force generation within actin arcs contributes to ligand discrimination. Together, our data are consistent with the idea that actomyosin arcs in the pSMAC region of the IS promote a mechano-chemical feedback mechanism that amplifies the accumulation of critical signaling molecules at the IS.
Collapse
|
9
|
Sauer EL, Cloake NC, Greer JM. Taming the TCR: antigen-specific immunotherapeutic agents for autoimmune diseases. Int Rev Immunol 2015; 34:460-85. [PMID: 25970132 DOI: 10.3109/08830185.2015.1027822] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Current treatments for autoimmune diseases are typically non-specific anti-inflammatory agents that affect not only the autoreactive cells but also the parts of the immune system that are required to maintain health. There is a need for the development of antigen-specific therapeutic agents that can effectively prevent the autoimmune attack while leaving the rest of the immune system functioning as normal. The simplest way to achieve this is using the autoantigen itself as a tolerizing agent; however, there is some risk involved with administering a potentially pathogenic antigen. In this review, we focus instead on the development and use of modified T cell receptor (TCR) ligands, in which the peptide ligand is modified to change the response by the T cell from a disease inducing to a protective response, and still retain the antigen-specificity necessary to target the autoreactive T cells. We review the use of modified TCR ligands as therapeutic agents in animal models of autoimmunity and in human autoimmune disease, and finally consider how they need to be improved in order to use them effectively in patients with autoimmune disease.
Collapse
Affiliation(s)
- Evan L Sauer
- a UQ Centre for Clinical Research , The University of Queensland , Brisbane , Queensland , Australia
| | - Nancy C Cloake
- a UQ Centre for Clinical Research , The University of Queensland , Brisbane , Queensland , Australia
| | - Judith M Greer
- a UQ Centre for Clinical Research , The University of Queensland , Brisbane , Queensland , Australia
| |
Collapse
|
10
|
Nelson RW, Beisang D, Tubo NJ, Dileepan T, Wiesner DL, Nielsen K, Wüthrich M, Klein BS, Kotov DI, Spanier JA, Fife BT, Moon JJ, Jenkins MK. T cell receptor cross-reactivity between similar foreign and self peptides influences naive cell population size and autoimmunity. Immunity 2015; 42:95-107. [PMID: 25601203 DOI: 10.1016/j.immuni.2014.12.022] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 10/25/2014] [Accepted: 12/22/2014] [Indexed: 01/06/2023]
Abstract
T cell receptor (TCR) cross-reactivity between major histocompatibility complex II (MHCII)-binding self and foreign peptides could influence the naive CD4(+) T cell repertoire and autoimmunity. We found that nonamer peptides that bind to the same MHCII molecule only need to share five amino acids to cross-react on the same TCR. This property was biologically relevant because systemic expression of a self peptide reduced the size of a naive cell population specific for a related foreign peptide by deletion of cells with cross-reactive TCRs. Reciprocally, an incompletely deleted naive T cell population specific for a tissue-restricted self peptide could be triggered by related microbial peptides to cause autoimmunity. Thus, TCR cross-reactivity between similar self and foreign peptides can reduce the size of certain foreign peptide-specific T cell populations and might allow T cell populations specific for tissue-restricted self peptides to cause autoimmunity after infection.
Collapse
|
11
|
Birnbaum ME, Garcia KC. Self-determination in the T cell repertoire. Immunity 2015; 42:8-10. [PMID: 25607452 DOI: 10.1016/j.immuni.2014.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The number of T cells specific for various antigens can vary dramatically. In this issue of Immunity, Nelson et al. (2015) report that these differences might be, at least in part, set by the number of cross-reactive self peptides encountered by T cells during development.
Collapse
Affiliation(s)
- Michael E Birnbaum
- Departments of Molecular and Cellular Physiology and Structural Biology, Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - K Christopher Garcia
- Departments of Molecular and Cellular Physiology and Structural Biology, Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; The Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
12
|
Løset GÅ, Berntzen G, Frigstad T, Pollmann S, Gunnarsen KS, Sandlie I. Phage Display Engineered T Cell Receptors as Tools for the Study of Tumor Peptide-MHC Interactions. Front Oncol 2015; 4:378. [PMID: 25629004 PMCID: PMC4290511 DOI: 10.3389/fonc.2014.00378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/17/2014] [Indexed: 11/13/2022] Open
Abstract
Cancer immunotherapy has finally come of age, demonstrated by recent progress in strategies that engage the endogenous adaptive immune response in tumor killing. Occasionally, significant and durable tumor regression has been achieved. A giant leap forward was the demonstration that the pre-existing polyclonal T cell repertoire could be re-directed by use of cloned T cell receptors (TCRs), to obtain a defined tumor-specific pool of T cells. However, the procedure must be performed with caution to avoid deleterious cross-reactivity. Here, the use of engineered soluble TCRs may represent a safer, yet powerful, alternative. There is also a need for deeper understanding of the processes that underlie antigen presentation in disease and homeostasis, how tumor-specific peptides are generated, and how epitope spreading evolves during tumor development. Due to its plasticity, the pivotal interaction where a TCR engages a peptide/MHC (pMHC) also requires closer attention. For this purpose, phage display as a tool to evolve cloned TCRs represents an attractive avenue to generate suitable reagents allowing the study of defined pMHC presentation, TCR engagement, as well as for the discovery of novel therapeutic leads. Here, we highlight important aspects of the current status in this field.
Collapse
Affiliation(s)
- Geir Åge Løset
- Nextera AS , Oslo , Norway ; Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway ; Department of Biosciences, University of Oslo , Oslo , Norway
| | | | | | | | - Kristin S Gunnarsen
- Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Inger Sandlie
- Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway ; Department of Biosciences, University of Oslo , Oslo , Norway
| |
Collapse
|
13
|
Shepherd AJ, Skelton S, Sansom CE, Gomez K, Moss DS, Hart DP. A large-scale computational study of inhibitor risk in non-severe haemophilia A. Br J Haematol 2014; 168:413-20. [PMID: 25244644 DOI: 10.1111/bjh.13131] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/29/2014] [Indexed: 01/13/2023]
Abstract
Over 500 missense F8 mutations have been reported to cause non-severe haemophilia A. Some F8 genotypes appear to confer a higher risk of inhibitor formation than others and individuals with the same F8 genotype may have differing risks of inhibitor formation. We present an in silico strategy demonstrating the heterogeneity of factor VIII (FVIII)-derived antigen presentation whilst identifying patterns of human leucocyte antigen (HLA) peptide binding that might predict future inhibitor risk. A well-validated computational tool, NetMHCII, enabled large-scale comparison of predicted antigen presentation between endogenous, mutated FVIII-derived peptides and wild-type, therapeutic FVIII-derived peptides spanning all F8 missense mutation positions reported to The Haemophilia A Mutation, Structure and Resource Site (HADB). We identify 40 F8 genotypes to be 'low risk' at a 50% inhibitory concentration (IC50 )-binding threshold of 300 nmol/l (P = 0·00005), defined as absence of novel peptide-major histocompatibility complex (MHC) surfaces for all 14 common HLA-DR alleles assessed. Analysing each of the possible 7280 F8 genotype/HLA-DR permutations individually at an IC50 threshold of 300 nmol/l, 65% are predicted to not generate a novel peptide-MHC surface that would be necessary to engage T cell help for subsequent anti-FVIII antibody generation. This study demonstrates the future importance of interpreting F8 genotype in the context of an individual's HLA profile to personalize inhibitor risk prediction.
Collapse
Affiliation(s)
- Adrian J Shepherd
- Department of Biological Sciences and Institute of Structural and Molecular Biology, Birkbeck, University of London, London, UK
| | | | | | | | | | | |
Collapse
|
14
|
|
15
|
Persaud SP, Parker CR, Lo WL, Weber KS, Allen PM. Intrinsic CD4+ T cell sensitivity and response to a pathogen are set and sustained by avidity for thymic and peripheral complexes of self peptide and MHC. Nat Immunol 2014; 15:266-74. [PMID: 24487322 PMCID: PMC3944141 DOI: 10.1038/ni.2822] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/20/2013] [Indexed: 12/13/2022]
Abstract
Interactions of T cell antigen receptors (TCRs) with complexes of self peptide and major histocompatibility complex (MHC) are crucial to T cell development, but their role in peripheral T cell responses remains unclear. Specific and nonspecific stimulation of LLO56 and LLO118 T cells, which transgenically express a TCR specific for the same Listeria monocytogenes epitope, elicited distinct interleukin 2 (IL-2) and phosphorylated kinase Erk responses, the strength of which was set in the thymus and maintained in the periphery in proportion to the avidity of the binding of the TCR to the self peptide-MHC complex. Deprivation of self peptide-MHC substantially compromised the population expansion of LLO56 T cells in response to L. monocytogenes in vivo. Despite their very different self-reactivity, LLO56 T cells and LLO118 T cells bound cognate peptide-MHC with an identical affinity, which challenges associations made between these parameters. Our findings highlight a crucial role for selecting ligands encountered during thymic 'education' in determining the intrinsic functionality of CD4+ T cells.
Collapse
Affiliation(s)
- Stephen P Persaud
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chelsea R Parker
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Wan-Lin Lo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, USA
| | - Paul M Allen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
16
|
Roy BM, Zhukov DV, Maynard JA. Flanking residues are central to DO11.10 T cell hybridoma stimulation by ovalbumin 323-339. PLoS One 2012; 7:e47585. [PMID: 23110081 PMCID: PMC3479146 DOI: 10.1371/journal.pone.0047585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 09/18/2012] [Indexed: 11/30/2022] Open
Abstract
T cell activation requires formation of a tri-molecular interaction between a major histocompatibility complex (MHC), peptide, and T cell receptor. In a common model system, the ovalbumin epitope 323–339 binds the murine class II MHC, I-Ad, in at least three distinct registers. The DO11.10 T cell recognizes the least stable of these, as determined by peptide-MHC dissociation rates. Using exogenous peptides and peptide insertions into a carrier protein in combination with IL-2 secretion assays, we show that the alternate registers do not competitively inhibit display of the active register four. In contrast, this weakly binding register is stabilized by the presence of n-terminal flanking residues active in MHC binding. The DO11.10 hybridoma is sensitive to the presence of specific wild-type residues extending to at least the P-3 peptide position. Transfer of the P-4 to P-2 flanking residues to a hen egg lysozyme epitope also presented by I-Ad increases the activity of that epitope substantially. These results illustrate the inherent complexity in delineating the interaction of multiple registers based on traditional thermodynamic measurements and demonstrate the potential of flanking residue modification for increasing the activity of weakly bound epitopes. The latter technique represents an alternative to substitution of anchor residues within a weakly bound register, which we show can significantly decrease the activity of the epitope to a responding T cell.
Collapse
Affiliation(s)
- Benjamin M. Roy
- Department of Chemical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Chemical Engineering, University of Texas at Austin, Austin Texas, United States of America
| | - Dmitriy V. Zhukov
- Department of Chemical Engineering, University of Texas at Austin, Austin Texas, United States of America
| | - Jennifer A. Maynard
- Department of Chemical Engineering, University of Texas at Austin, Austin Texas, United States of America
- * E-mail:
| |
Collapse
|
17
|
Jordan KR, Buhrman JD, Sprague J, Moore BL, Gao D, Kappler JW, Slansky JE. TCR hypervariable regions expressed by T cells that respond to effective tumor vaccines. Cancer Immunol Immunother 2012; 61:1627-38. [PMID: 22350070 PMCID: PMC3410973 DOI: 10.1007/s00262-012-1217-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 01/30/2012] [Indexed: 12/31/2022]
Abstract
A major goal of immunotherapy for cancer is the activation of T cell responses against tumor-associated antigens (TAAs). One important strategy for improving antitumor immunity is vaccination with peptide variants of TAAs. Understanding the mechanisms underlying the expansion of T cells that respond to the native tumor antigen is an important step in developing effective peptide-variant vaccines. Using an immunogenic mouse colon cancer model, we compare the binding properties and the TCR genes expressed by T cells elicited by peptide variants that elicit variable antitumor immunity directly ex vivo. The steady-state affinity of the natural tumor antigen for the T cells responding to effective peptide vaccines was higher relative to ineffective peptides, consistent with their improved function. Ex vivo analysis showed that T cells responding to the effective peptides expressed a CDR3β motif, which was also shared by T cells responding to the natural antigen and not those responding to the less effective peptide vaccines. Importantly, these data demonstrate that peptide vaccines can expand T cells that naturally respond to tumor antigens, resulting in more effective antitumor immunity. Future immunotherapies may require similar stringent analysis of the responding T cells to select optimal peptides as vaccine candidates.
Collapse
Affiliation(s)
- Kimberly R Jordan
- Integrated Department of Immunology, School of Medicine, University of Colorado Denver, 1400 Jackson Street, Room K511, Denver, CO 80206, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Specificity and detection of insulin-reactive CD4+ T cells in type 1 diabetes in the nonobese diabetic (NOD) mouse. Proc Natl Acad Sci U S A 2011; 108:16729-34. [PMID: 21949373 DOI: 10.1073/pnas.1113954108] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In the nonobese diabetic (NOD) mouse model of type 1 diabetes (T1D), an insulin peptide (B:9-23) is a major target for pathogenic CD4(+) T cells. However, there is no consensus on the relative importance of the various positions or "registers" this peptide can take when bound in the groove of the NOD MHCII molecule, IA(g7). This has hindered structural studies and the tracking of the relevant T cells in vivo with fluorescent peptide-MHCII tetramers. Using mutated B:9-23 peptides and methods for trapping the peptide in particular registers, we show that most, if not all, NOD CD4(+) T cells react to B:9-23 bound in low-affinity register 3. However, these T cells can be divided into two types depending on whether their response is improved or inhibited by substituting a glycine for the B:21 glutamic acid at the p8 position of the peptide. On the basis of these findings, we constructed a set of fluorescent insulin-IA(g7) tetramers that bind to most insulin-specific T-cell clones tested. A mixture of these tetramers detected a high frequency of B:9-23-reactive CD4(+) T cells in the pancreases of prediabetic NOD mice. Our data are consistent with the idea that, within the pancreas, unique processing of insulin generates truncated peptides that lack or contain the B:21 glutamic acid. In the thymus, the absence of this type of processing combined with the low affinity of B:9-23 binding to IA(g7) in register 3 may explain the escape of insulin-specific CD4(+) T cells from the mechanisms that usually eliminate self-reactive T cells.
Collapse
|
19
|
Andersson IE, Batsalova T, Haag S, Dzhambazov B, Holmdahl R, Kihlberg J, Linusson A. (E)-alkene and ethylene isosteres substantially alter the hydrogen-bonding network in class II MHC A(q)/glycopeptide complexes and affect T-cell recognition. J Am Chem Soc 2011; 133:14368-78. [PMID: 21766871 DOI: 10.1021/ja2038722] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The structural basis for antigen presentation by class II major histocompatibility complex (MHC) proteins to CD4(+) T-cells is important for understanding and possibly treating autoimmune diseases. In the work described in this paper, (E)-alkene and ethylene amide-bond isosteres were used to investigate the effect of removing hydrogen-bonding possibilities from the CII259-270 glycopeptide, which is bound by the arthritis-associated murine A(q) class II MHC protein. The isostere-modified glycopeptides showed varying and unexpectedly large losses of A(q) binding that could be linked to the dynamics of the system. Molecular dynamics (MD) simulations revealed that the backbone of CII259-270 and the A(q) protein are able to form up to 11 hydrogen bonds, but fewer than this number are present at any one time. Most of the strong hydrogen-bond interactions were formed by the N-terminal part of the glycopeptide, i.e., in the region where the isosteric replacements were made. The structural dynamics also revealed that hydrogen bonds were strongly coupled to each other; the loss of one hydrogen-bond interaction had a profound effect on the entire hydrogen-bonding network. The A(q) binding data revealed that an ethylene isostere glycopeptide unexpectedly bound more strongly to A(q) than the corresponding (E)-alkene, which is in contrast to the trend observed for the other isosteres. Analysis of the MD trajectories revealed that the complex conformation of this ethylene isostere was structurally different and had an altered molecular interaction pattern compared to the other A(q)/glycopeptide complexes. The introduced amide-bond isosteres also affected the interactions of the glycopeptide/A(q) complexes with T-cell receptors. The dynamic variation of the patterns and strengths of the hydrogen-bond interactions in the class II MHC system is of critical importance for the class II MHC/peptide/TCR signaling system.
Collapse
Affiliation(s)
- Ida E Andersson
- Department of Chemistry, Umeå University, SE-901 87 Umeå, Sweden
| | | | | | | | | | | | | |
Collapse
|
20
|
Agudelo WA, Galindo JF, Patarroyo ME. Electrostatic potential as a tool to understand interactions between malaria vaccine candidate peptides and MHC II molecules. Biochem Biophys Res Commun 2011; 410:410-5. [PMID: 21672519 DOI: 10.1016/j.bbrc.2011.05.145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 05/31/2011] [Indexed: 01/17/2023]
Abstract
One of the most important problems in vaccine development consists in understanding receptor-ligand interactions between Class II Major Histocompatibility Complex molecules (MHC II) and antigenic peptides involved in inducing an appropriate immune response. In this study, we used X-ray crystallography structural data provided by the HLA-DRβ1*0301-CLIP peptide interaction to compare native non-immunogenic and specifically-modified immunogenic peptides derived from the malarial SALSA protein, by analyzing molecular electrostatic potential surfaces on the most important regions of the peptide binding groove (Pockets 1, 4, 6 and 9). Important differences were found on the electrostatic potential induced by these peptides, particularly in MHC II conserved residues: Qα9, Sα53, Nα62, Nα69, Yβ30, Yβ60, Wβ61, Qβ70, Kβ71 and Vβ86, the same ones involved in establishing hydrogen bonds between Class II molecule-peptide and the recognition by T cell receptor, it correlating well with the change in their immunological properties. The results clearly suggest that modifications done on the electrostatic potential of these amino acids could favor the induction of different immune responses and therefore, their identification could allow modifying peptides a priori and in silico, so as to render them into immunogenic and protection-inducers and hence suitable components of a chemically-synthesized, multi-antigenic, minimal subunit based vaccine.
Collapse
Affiliation(s)
- William A Agudelo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | | | | |
Collapse
|
21
|
Patarroyo ME, Bermúdez A, Patarroyo MA. Structural and Immunological Principles Leading to Chemically Synthesized, Multiantigenic, Multistage, Minimal Subunit-Based Vaccine Development. Chem Rev 2011; 111:3459-507. [DOI: 10.1021/cr100223m] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Manuel Elkin Patarroyo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50, No. 26-00, Bogotá, Colombia
- Universidad Nacional de Colombia
| | - Adriana Bermúdez
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50, No. 26-00, Bogotá, Colombia
- Universidad del Rosario
| | - Manuel Alfonso Patarroyo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50, No. 26-00, Bogotá, Colombia
- Universidad del Rosario
| |
Collapse
|
22
|
Andersson IE, Andersson CD, Batsalova T, Dzhambazov B, Holmdahl R, Kihlberg J, Linusson A. Design of glycopeptides used to investigate class II MHC binding and T-cell responses associated with autoimmune arthritis. PLoS One 2011; 6:e17881. [PMID: 21423632 PMCID: PMC3058040 DOI: 10.1371/journal.pone.0017881] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 02/13/2011] [Indexed: 01/12/2023] Open
Abstract
The glycopeptide fragment CII259–273 from type II collagen (CII) binds to the murine Aq and human DR4 class II Major Histocompatibility Complex (MHC II) proteins, which are associated with development of murine collagen-induced arthritis (CIA) and rheumatoid arthritis (RA), respectively. It has been shown that CII259–273 can be used in therapeutic vaccination of CIA. This glycopeptide also elicits responses from T-cells obtained from RA patients, which indicates that it has an important role in RA as well. We now present a methodology for studies of (glyco)peptide-receptor interactions based on a combination of structure-based virtual screening, ligand-based statistical molecular design and biological evaluations. This methodology included the design of a CII259–273 glycopeptide library in which two anchor positions crucial for binding in pockets of Aq and DR4 were varied. Synthesis and biological evaluation of the designed glycopeptides provided novel structure-activity relationship (SAR) understanding of binding to Aq and DR4. Glycopeptides that retained high affinities for these MHC II proteins and induced strong responses in panels of T-cell hybridomas were also identified. An analysis of all the responses revealed groups of glycopeptides with different response patterns that are of high interest for vaccination studies in CIA. Moreover, the SAR understanding obtained in this study provides a platform for the design of second-generation glycopeptides with tuned MHC affinities and T-cell responses.
Collapse
Affiliation(s)
| | | | - Tsvetelina Batsalova
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Balik Dzhambazov
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Rikard Holmdahl
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Jan Kihlberg
- Department of Chemistry, Umeå University, Umeå, Sweden
- AstraZeneca R&D Mölndal, Mölndal, Sweden
| | | |
Collapse
|
23
|
Borbulevych OY, Piepenbrink KH, Baker BM. Conformational melding permits a conserved binding geometry in TCR recognition of foreign and self molecular mimics. THE JOURNAL OF IMMUNOLOGY 2011; 186:2950-8. [PMID: 21282516 DOI: 10.4049/jimmunol.1003150] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Molecular mimicry between foreign and self Ags is a mechanism of TCR cross-reactivity and is thought to contribute to the development of autoimmunity. The αβ TCR A6 recognizes the foreign Ag Tax from the human T cell leukemia virus-1 when presented by the class I MHC HLA-A2. In a possible link with the autoimmune disease human T cell leukemia virus-1-associated myelopathy/tropical spastic paraparesis, A6 also recognizes a self peptide from the neuronal protein HuD in the context of HLA-A2. We found in our study that the complexes of the HuD and Tax epitopes with HLA-A2 are close but imperfect structural mimics and that in contrast with other recent structures of TCRs with self Ags, A6 engages the HuD Ag with the same traditional binding mode used to engage Tax. Although peptide and MHC conformational changes are needed for recognition of HuD but not Tax and the difference of a single hydroxyl triggers an altered TCR loop conformation, TCR affinity toward HuD is still within the range believed to result in negative selection. Probing further, we found that the HuD-HLA-A2 complex is only weakly stable. Overall, these findings help clarify how molecular mimicry can drive self/nonself cross-reactivity and illustrate how low peptide-MHC stability can permit the survival of T cells expressing self-reactive TCRs that nonetheless bind with a traditional binding mode.
Collapse
Affiliation(s)
- Oleg Y Borbulevych
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | |
Collapse
|
24
|
Affiliation(s)
- Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
25
|
Valkenburg SA, Gras S, Guillonneau C, La Gruta NL, Thomas PG, Purcell AW, Rossjohn J, Doherty PC, Turner SJ, Kedzierska K. Protective efficacy of cross-reactive CD8+ T cells recognising mutant viral epitopes depends on peptide-MHC-I structural interactions and T cell activation threshold. PLoS Pathog 2010; 6:e1001039. [PMID: 20711359 PMCID: PMC2920842 DOI: 10.1371/journal.ppat.1001039] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Accepted: 07/13/2010] [Indexed: 11/19/2022] Open
Abstract
Emergence of a new influenza strain leads to a rapid global spread of the virus due to minimal antibody immunity. Pre-existing CD8+ T-cell immunity directed towards conserved internal viral regions can greatly ameliorate the disease. However, mutational escape within the T cell epitopes is a substantial issue for virus control and vaccine design. Although mutations can result in a loss of T cell recognition, some variants generate cross-reactive T cell responses. In this study, we used reverse genetics to modify the influenza NP336–374 peptide at a partially-solvent exposed residue (N->A, NPN3A mutation) to assess the availability, effectiveness and mechanism underlying influenza-specific cross-reactive T cell responses. The engineered virus induced a diminished CD8+ T cell response and selected a narrowed T cell receptor (TCR) repertoire within two Vβ regions (Vβ8.3 and Vβ9). This can be partially explained by the H-2DbNPN3A structure that showed a loss of several contacts between the NPN3A peptide and H-2Db, including a contact with His155, a position known to play an important role in mediating TCR-pMHC-I interactions. Despite these differences, common cross-reactive TCRs were detected in both the naïve and immune NPN3A-specific TCR repertoires. However, while the NPN3A epitope primes memory T-cells that give an equivalent recall response to the mutant or wild-type (wt) virus, both are markedly lower than wt->wt challenge. Such decreased CD8+ responses elicited after heterologous challenge resulted in delayed viral clearance from the infected lung. Furthermore, mice first exposed to the wt virus give a poor, low avidity response following secondary infection with the mutant. Thus, the protective efficacy of cross-reactive CD8+ T cells recognising mutant viral epitopes depend on peptide-MHC-I structural interactions and functional avidity. Our study does not support vaccine strategies that include immunization against commonly selected cross-reactive variants with mutations at partially-solvent exposed residues that have characteristics comparable to NPN3A. Introduction of a new influenza strain into human circulation leads to a rapid global spread of the virus due to minimal antibody immunity. Established T-cell immunity towards conserved viral regions provides some protection against influenza and promotes rapid recovery. However, influenza viruses mutate to escape the protective immunity. We found that established T cell immunity can recognise influenza mutants with variations at positions that are partially involved in T cell recognition. However, an initial priming with the mutated variant decreases recognition of the original parental virus. This finding results from a markedly lower functional quality and limited structural interactions of the mutant. In terms of possible vaccination strategies for rapidly changing viruses or tumours, it appears that priming with cross-reactive mutants that display such characteristics would be of no benefit as the same level of T cell immunity against such mutants can be elicited by exposure to the original virus.
Collapse
Affiliation(s)
- Sophie A. Valkenburg
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Australia
| | - Stephanie Gras
- The Protein Crystallography Unit, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - Carole Guillonneau
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Australia
| | - Nicole L. La Gruta
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Australia
| | - Paul G. Thomas
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Anthony W. Purcell
- Department of Biochemistry and Molecular Biology, the Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Australia
| | - Jamie Rossjohn
- The Protein Crystallography Unit, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - Peter C. Doherty
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Australia
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Stephen J. Turner
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Australia
- * E-mail:
| |
Collapse
|
26
|
Stadinski BD, Zhang L, Crawford F, Marrack P, Eisenbarth GS, Kappler JW. Diabetogenic T cells recognize insulin bound to IAg7 in an unexpected, weakly binding register. Proc Natl Acad Sci U S A 2010; 107:10978-83. [PMID: 20534455 PMCID: PMC2890771 DOI: 10.1073/pnas.1006545107] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
A peptide derived from the insulin B chain contains a major epitope for diabetogenic CD4(+) T cells in the NOD mouse model of type 1 diabetes (T1D). This peptide can fill the binding groove of the NOD MHCII molecule, IA(g7), in a number of ways or "registers." We show here that a diverse set of NOD anti-insulin T cells all recognize this peptide bound in the same register. Surprisingly, this register results in the poorest binding of peptide to IA(g7). The poor binding is due to an incompatibility between the p9 amino acid of the peptide and the unique IA(g7) p9 pocket polymorphisms that are strongly associated with susceptibility to T1D. Our findings suggest that the association of autoimmunity with particular MHCII alleles may be do to poorer, rather than more favorable, binding of the critical self-epitopes, allowing T-cell escape from thymic deletion.
Collapse
Affiliation(s)
- Brian D. Stadinski
- Integrated Department of Immunology, University of Colorado and National Jewish Health, Denver, CO 80206
- Howard Hughes Medical Institute, National Jewish Health, Denver, CO 80206; and
| | - Li Zhang
- Barbara Davis Center for Childhood Diabetes, Aurora, CO 80045
| | - Frances Crawford
- Integrated Department of Immunology, University of Colorado and National Jewish Health, Denver, CO 80206
- Howard Hughes Medical Institute, National Jewish Health, Denver, CO 80206; and
| | - Philippa Marrack
- Integrated Department of Immunology, University of Colorado and National Jewish Health, Denver, CO 80206
- Howard Hughes Medical Institute, National Jewish Health, Denver, CO 80206; and
| | - George S. Eisenbarth
- Integrated Department of Immunology, University of Colorado and National Jewish Health, Denver, CO 80206
- Barbara Davis Center for Childhood Diabetes, Aurora, CO 80045
| | - John W. Kappler
- Integrated Department of Immunology, University of Colorado and National Jewish Health, Denver, CO 80206
- Howard Hughes Medical Institute, National Jewish Health, Denver, CO 80206; and
| |
Collapse
|
27
|
Abdul-Alim CS, Li Y, Yee C. Conditional superagonist CTL ligands for the promotion of tumor-specific CTL responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:6514-21. [PMID: 20483791 PMCID: PMC3313650 DOI: 10.4049/jimmunol.0900448] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although it has been demonstrated that CTLs can be raised against tumor-associated self-antigens, achieving consistent and effective clinical responses has proven challenging. Superagonist altered peptide ligands (APLs) can often elicit potent antitumor CTL responses where the native tumor-associated epitope fails. Current methods have identified a limited number of superagonist APLs, including the prototypic 27L mutant of MART-1. However, more comprehensive screening strategies would be desirable. In this study, we use a novel genetic screen, involving recombinant technology and class I Ag cross-presentation, to search for supraoptimal superagonists of the 27L MART-1 mutant by surveying the effectiveness of virtually every single amino acid substitution mutant of 27L to activate human Ag-specific CTL clones recognizing the wild-type MART-1(26-35) epitope. We identify three novel mutant epitopes with superagonist properties that are functionally superior to 27L; however, the ability of a given analogue to act as superagonist varies among patients and suggests that a given superagonist APL may be ideally suited to different patients. These findings endorse the use of comprehensive methods to establish panels of potential superagonist APLs to individualize tumor peptide vaccines among patients.
Collapse
Affiliation(s)
- C. Siddiq Abdul-Alim
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle WA 98109
| | - Yongqing Li
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle WA 98109
| | - Cassian Yee
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle WA 98109
- Department of Medicine, Division of Oncology, University of Washington Medical Center, Seattle, WA 98195
| |
Collapse
|
28
|
Persaud SP, Donermeyer DL, Weber KS, Kranz DM, Allen PM. High-affinity T cell receptor differentiates cognate peptide-MHC and altered peptide ligands with distinct kinetics and thermodynamics. Mol Immunol 2010; 47:1793-801. [PMID: 20334923 PMCID: PMC2860700 DOI: 10.1016/j.molimm.2010.02.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 02/21/2010] [Indexed: 01/13/2023]
Abstract
Interactions between the T cell receptor and cognate peptide-MHC are crucial initiating events in the adaptive immune response. These binding events are highly specific yet occur with micromolar affinity. Even weaker interactions between TCR and self-pMHC complexes play critical regulatory roles in T cell development, maintenance and coagonist activity. Due to their low-affinity, the kinetics and thermodynamics of such weak interactions are difficult to study. In this work, we used M15, a high-affinity TCR engineered from the 3.L2 TCR system, to study the binding properties, thermodynamics, and specificity of two altered peptide ligands (APLs). Our affinity measurements of the high-affinity TCR support the view that the wild type TCR binds these APLs in the millimolar affinity range, and hence very low affinities can still elicit biological functions. Finally, single methylene differences among the APLs gave rise to strikingly different binding thermodynamics. These minor changes in the pMHC antigen were associated with significant and unpredictable changes in both the entropy and enthalpy of the reaction. As the identical TCR was analyzed with several structurally similar ligands, the distinct thermodynamic binding profiles provide a mechanistic perspective on how exquisite antigen specificity is achieved by the T cell receptor.
Collapse
Affiliation(s)
- Stephen P. Persaud
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - David L. Donermeyer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - K. Scott Weber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - David M. Kranz
- Department of Biochemistry, University of Illinois, Urbana, IL 61801
| | - Paul M. Allen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
29
|
Patarroyo MA, Bermúdez A, López C, Yepes G, Patarroyo ME. 3D analysis of the TCR/pMHCII complex formation in monkeys vaccinated with the first peptide inducing sterilizing immunity against human malaria. PLoS One 2010; 5:e9771. [PMID: 20333301 PMCID: PMC2841639 DOI: 10.1371/journal.pone.0009771] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 02/25/2010] [Indexed: 11/18/2022] Open
Abstract
T-cell receptor gene rearrangements were studied in Aotus monkeys developing high antibody titers and sterilizing immunity against the Plasmodium falciparum malaria parasite upon vaccination with the modified synthetic peptide 24112, which was identified in the Merozoite Surface Protein 2 (MSP-2) and is known to bind to HLA-DRβ1*0403 molecules with high capacity. Spectratyping analysis showed a preferential usage of Vβ12 and Vβ6 TCR gene families in 67% of HLA-DRβ1*0403-like genotyped monkeys. Docking of peptide 24112 into the HLA-DRβ1*0401–HA peptide–HA1.7TCR complex containing the VDJ rearrangements identified in fully protected monkeys showed a different structural signature compared to nonprotected monkeys. These striking results show the exquisite specificity of the TCR/pMHCII complex formation needed for inducing sterilizing immunity and provide important hints for a logical and rational methodology to develop multiepitopic, minimal subunit-based synthetic vaccines against infectious diseases, among them malaria.
Collapse
Affiliation(s)
- Manuel A. Patarroyo
- Fundación Instituto de Inmunología de Colombia, Bogotá, Cundinamarca, Colombia
- Universidad del Rosario, Bogotá, Cundinamarca, Colombia
| | - Adriana Bermúdez
- Fundación Instituto de Inmunología de Colombia, Bogotá, Cundinamarca, Colombia
- Universidad del Rosario, Bogotá, Cundinamarca, Colombia
| | - Carolina López
- Fundación Instituto de Inmunología de Colombia, Bogotá, Cundinamarca, Colombia
- Universidad del Rosario, Bogotá, Cundinamarca, Colombia
| | - Gloria Yepes
- Fundación Instituto de Inmunología de Colombia, Bogotá, Cundinamarca, Colombia
| | - Manuel E. Patarroyo
- Fundación Instituto de Inmunología de Colombia, Bogotá, Cundinamarca, Colombia
- Universidad Nacional de Colombia, Bogotá, Cundinamarca, Colombia
- * E-mail:
| |
Collapse
|
30
|
Baumgartner CK, Ferrante A, Nagaoka M, Gorski J, Malherbe LP. Peptide-MHC class II complex stability governs CD4 T cell clonal selection. THE JOURNAL OF IMMUNOLOGY 2009; 184:573-81. [PMID: 20007533 DOI: 10.4049/jimmunol.0902107] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The clonal composition of the T cell response can affect its ability to mediate infection control or to induce autoimmunity, but the mechanisms regulating the responding TCR repertoire remain poorly defined. In this study, we immunized mice with wild-type or mutated peptides displaying varying binding half-lives with MHC class II molecules to measure the impact of peptide-MHC class II stability on the clonal composition of the CD4 T cell response. We found that, although all peptides elicited similar T cell response size on immunization, the clonotypic diversity of the CD4 T cell response correlated directly with the half-life of the immunizing peptide. Peptides with short half-lives focused CD4 T cell response toward high-affinity clonotypes expressing restricted public TCR, whereas peptides with longer half-lives broadened CD4 T cell response by recruiting lower-affinity clonotypes expressing more diverse TCR. Peptides with longer half-lives did not cause the elimination of high-affinity clonotypes, and at a low dose, they also skewed CD4 T cell response toward higher-affinity clonotypes. Taken collectively, our results suggest the half-life of peptide-MHC class II complexes is the primary parameter that dictates the clonotypic diversity of the responding CD4 T cell compartment.
Collapse
|
31
|
Sadegh-Nasseri S, Dalai SK, Korb Ferris LC, Mirshahidi S. Suboptimal engagement of the T-cell receptor by a variety of peptide-MHC ligands triggers T-cell anergy. Immunology 2009; 129:1-7. [PMID: 20002785 DOI: 10.1111/j.1365-2567.2009.03206.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
T cells recognize antigen via the T-cell receptor (TCR) and produce a spectrum of responses that range from activation to anergy or cell death. The variety of outcomes may be dictated by the strength of the signals transmitted upon cognate recognition of the TCR. The physiological outcome of TCR engagement is determined by several factors, including the avidity of the ligand for TCR, the duration of engagement, and the presence and nature of accessory molecules present on antigen-presenting cells (APCs). In this review, we discuss a model of anergy induced by presentation of low densities of peptide-major histocompatibility complex (MHC) ligand in CD4(+) T cells and compare it to anergy induced by altered peptide ligands in an effort to identify a unifying mechanism. We suggest that altered peptide ligand (APL) and low densities of agonist ligands induce anergy by engaging less than optimal numbers of TCRs. The physiological impacts of anergy in memory CD4(+) T cells are discussed.
Collapse
|
32
|
Manipulating antigenic ligand strength to selectively target myelin-reactive CD4+ T cells in EAE. J Neuroimmune Pharmacol 2009; 5:176-88. [PMID: 19904613 DOI: 10.1007/s11481-009-9181-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 10/15/2009] [Indexed: 10/20/2022]
Abstract
The development of antigen-specific therapies for the selective tolerization of autoreactive T cells remains the Holy Grail for the treatment of T-cell-mediated autoimmune diseases such as multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). This quest remains elusive, however, as the numerous antigen-specific strategies targeting myelin-specific T cells over the years have failed to result in clinical success. In this review, we revisit the antigen-based therapies used in the treatment of myelin-specific CD4+ T cells in the context of the functional avidity and the strength of signal of the encephalitogenic CD4+ T cell repertoire. In light of differences in activation thresholds, we propose that autoreactive T cells are not all equal, and therefore tolerance induction strategies must incorporate ligand strength in order to be successful in treating EAE and ultimately the human disease MS.
Collapse
|
33
|
Lo WL, Felix NJ, Walters JJ, Rohrs H, Gross ML, Allen PM. An endogenous peptide positively selects and augments the activation and survival of peripheral CD4+ T cells. Nat Immunol 2009; 10:1155-61. [PMID: 19801984 PMCID: PMC2764840 DOI: 10.1038/ni.1796] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 08/31/2009] [Indexed: 11/16/2022]
Abstract
Although CD4+ and CD8+ T cells differ in their positively selecting signal strength, endogenous positively selecting ligands have only been identified for MHC class I-restricted T cell receptors (TCRs). Here we screened for ligands that can positively select MHC class II-restricted TCRs, using thymocytes from four I-Ek restricted TCR transgenic mice and a large panel of self peptides. One peptide, gp250, induced positive selection of AND CD4+ T cells, had no homology with the AND TCR agonist ligand, and was recognized with a high degree of specificity. gp250 acted as a co-agonist to initiate activation and enhance survival of peripheral AND CD4+ T cells. Thus, positively selecting ligands play critical roles in thymocyte development and in the activation and maintenance of peripheral T cells.
Collapse
Affiliation(s)
- Wan-Lin Lo
- Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | |
Collapse
|
34
|
Varkony H, Weinstein V, Klinger E, Sterling J, Cooperman H, Komlosh T, Ladkani D, Schwartz R. The glatiramoid class of immunomodulator drugs. Expert Opin Pharmacother 2009; 10:657-68. [DOI: 10.1517/14656560902802877] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
35
|
Beltrami A, Rossmann M, Fiorillo MT, Paladini F, Sorrentino R, Saenger W, Kumar P, Ziegler A, Uchanska-Ziegler B. Citrullination-dependent differential presentation of a self-peptide by HLA-B27 subtypes. J Biol Chem 2008; 283:27189-99. [PMID: 18650441 DOI: 10.1074/jbc.m802818200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Inflammatory processes are accompanied by the posttranslational modification of certain arginine residues within proteins to yield citrulline, although it is largely unknown how this modification influences antigen presentation. We employed crystallographic and functional studies to investigate whether the exchange of arginine to citrulline affects the display of a peptide by two human major histocompatibility antigen class I subtypes, HLA-B(*)2705 and HLA-B(*)2709. Both differ only in residue 116 within the peptide binding groove despite their differential association with ankylosing spondylitis, an inflammatory rheumatic disorder. The crystal structures described here show that a modified self-peptide, pVIPR-U5 (RRKWURWHL; U = citrulline), is presented by the two HLA-B27 molecules in distinct conformations. These binding modes differ not only drastically from each other but also from the conformations exhibited by the non-citrullinated peptide in a given subtype. The differential reactivity of HLA-B27-restricted cytotoxic T cells with modified or unmodified pVIPR supports the structural findings and shows that the presentation of citrullinated peptides has the potential to influence immune responses.
Collapse
Affiliation(s)
- Alessandra Beltrami
- Institut für Immungenetik, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Freie Universität Berlin, Thielallee 73, 14195 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Silano M, Di Benedetto R, Maialetti F, De Vincenzi A, Calcaterra R, Trecca A, De Vincenzi M. A 10-residue peptide from durum wheat promotes a shift from a Th1-type response toward a Th2-type response in celiac disease. Am J Clin Nutr 2008; 87:415-23. [PMID: 18258633 DOI: 10.1093/ajcn/87.2.415] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Celiac disease (CD) is a Th1-driven autoimmune permanent enteropathy that is triggered by dietary gluten. Molecules able to shift the immune response from a Th1- to a Th2-type response have been suggested as therapeutic agents for Th1 autoimmune diseases. OBJECTIVE We sought to investigate the possibility that a decapeptide from durum wheat (p10mer, QQPQDAVQPF), which was previously shown to prevent the activation of celiac peripheral lymphocytes, may promote a shift from a Th1- to a Th2-type immune response in gluten-specific intestinal T cells of CD patients. DESIGN Intestinal T lymphocyte lines derived from 8 children with CD were incubated with gliadin peptides both alone and simultaneously with p10mer. Cell proliferation and the production of interferon-gamma and interleukin-10 by these T cells were measured. RESULTS The incubation of celiac intestinal T cells with deamidated gliadin peptides resulted in a significant (P < 0.008) increase in cell proliferation and interferon-gamma release, whereas the simultaneous exposure to p10mer totally abolished the cell proliferation and cytokine release. Moreover, incubation with p10mer maintained an elevated release of interleukin-10, whereas exposure of the cells to culture medium only did not. The replacement of the residues of aspartic acid in position 5 or those of alanine in position 6 in the sequence of p10mer resulted in peptides with no activity in the activation experiments. CONCLUSION In vitro, p10mer showed the ability to shift the pathogenic immune response of a CD patient from a Th1- to a Th2-type response.
Collapse
Affiliation(s)
- Marco Silano
- Division of Food Science, Human Nutrition and Health, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Rinderknecht CH, Belmares MP, Catanzarite TLW, Bankovich AJ, Holmes TH, Garcia KC, Nanda NK, Busch R, Kovats S, Mellins ED. Posttranslational regulation of I-Ed by affinity for CLIP. THE JOURNAL OF IMMUNOLOGY 2007; 179:5907-15. [PMID: 17947664 DOI: 10.4049/jimmunol.179.9.5907] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Several MHC class II alleles linked with autoimmune diseases form unusually low stability complexes with CLIP, leading us to hypothesize that this is an important feature contributing to autoimmune pathogenesis. To investigate cellular consequences of altering class II/CLIP affinity, we evaluated invariant chain (Ii) mutants with varying CLIP affinity for a mouse class II allele, I-E(d), which has low affinity for wild-type CLIP and is associated with a mouse model of spontaneous, autoimmune joint inflammation. Increasing CLIP affinity for I-E(d) resulted in increased cell surface and total cellular abundance and half-life of I-E(d). This reveals a post-endoplasmic reticulum chaperoning capacity of Ii via its CLIP peptides. Quantitative effects on I-E(d) were less pronounced in DM-expressing cells, suggesting complementary chaperoning effects mediated by Ii and DM, and implying that the impact of allelic variation in CLIP affinity on immune responses will be highest in cells with limited DM activity. Differences in the ability of cell lines expressing wild-type or high-CLIP-affinity mutant Ii to present Ag to T cells suggest a model in which increased CLIP affinity for class II serves to restrict peptide loading to DM-containing compartments, ensuring proper editing of antigenic peptides.
Collapse
Affiliation(s)
- Cornelia H Rinderknecht
- Program in Immunology, Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Borbulevych OY, Insaidoo FK, Baxter TK, Powell DJ, Johnson LA, Restifo NP, Baker BM. Structures of MART-126/27-35 Peptide/HLA-A2 complexes reveal a remarkable disconnect between antigen structural homology and T cell recognition. J Mol Biol 2007; 372:1123-36. [PMID: 17719062 PMCID: PMC2134917 DOI: 10.1016/j.jmb.2007.07.025] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Revised: 07/07/2007] [Accepted: 07/10/2007] [Indexed: 11/24/2022]
Abstract
Small structural changes in peptides presented by major histocompatibility complex (MHC) molecules often result in large changes in immunogenicity, supporting the notion that T cell receptors are exquisitely sensitive to antigen structure. Yet there are striking examples of TCR recognition of structurally dissimilar ligands. The resulting unpredictability of how T cells will respond to different or modified antigens impacts both our understanding of the physical bases for TCR specificity as well as efforts to engineer peptides for immunomodulation. In cancer immunotherapy, epitopes and variants derived from the MART-1/Melan-A protein are widely used as clinical vaccines. Two overlapping epitopes spanning amino acid residues 26 through 35 are of particular interest: numerous clinical studies have been performed using variants of the MART-1 26-35 decamer, although only the 27-35 nonamer has been found on the surface of targeted melanoma cells. Here, we show that the 26-35 and 27-35 peptides adopt strikingly different conformations when bound to HLA-A2. Nevertheless, clonally distinct MART-1(26/27-35)-reactive T cells show broad cross-reactivity towards these ligands. Simultaneously, however, many of the cross-reactive T cells remain unable to recognize anchor-modified variants with very subtle structural differences. These dichotomous observations challenge our thinking about how structural information on unligated peptide/MHC complexes should be best used when addressing questions of TCR specificity. Our findings also indicate that caution is warranted in the design of immunotherapeutics based on the MART-1 26/27-35 epitopes, as neither cross-reactivity nor selectivity is predictable based on the analysis of the structures alone.
Collapse
Affiliation(s)
- Oleg Y. Borbulevych
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame IN 46556, USA
- Walther Cancer Research Center, 251 Nieuwland Science Hall, University of Notre Dame Notre Dame, IN 46530, USA
| | - Francis K. Insaidoo
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame IN 46556, USA
| | - Tiffany K. Baxter
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame IN 46556, USA
| | - Daniel J. Powell
- National Cancer Institute, National Institutes of Health Bethesda, MD 20892, USA
| | - Laura A. Johnson
- National Cancer Institute, National Institutes of Health Bethesda, MD 20892, USA
| | - Nicholas P. Restifo
- National Cancer Institute, National Institutes of Health Bethesda, MD 20892, USA
| | - Brian M. Baker
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame IN 46556, USA
- Walther Cancer Research Center, 251 Nieuwland Science Hall, University of Notre Dame Notre Dame, IN 46530, USA
| |
Collapse
|
41
|
Wucherpfennig KW, Allen PM, Celada F, Cohen IR, De Boer R, Garcia KC, Goldstein B, Greenspan R, Hafler D, Hodgkin P, Huseby ES, Krakauer DC, Nemazee D, Perelson AS, Pinilla C, Strong RK, Sercarz EE. Polyspecificity of T cell and B cell receptor recognition. Semin Immunol 2007; 19:216-24. [PMID: 17398114 PMCID: PMC2034306 DOI: 10.1016/j.smim.2007.02.012] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Accepted: 02/26/2007] [Indexed: 02/06/2023]
Abstract
A recent workshop discussed the recognition of multiple distinct ligands by individual T cell and B cell receptors and the implications of this discovery for lymphocyte biology. The workshop recommends general use of the term polyspecificity because it emphasizes two fundamental aspects, the inherent specificity of receptor recognition and the ability to recognize multiple ligands. Many different examples of polyspecificity and the structural mechanisms were discussed, and the group concluded that polyspecificity is a general, inherent feature of TCR and antibody recognition. This review summarizes the relevance of polyspecificity for lymphocyte development, activation and disease processes.
Collapse
Affiliation(s)
- Kai W Wucherpfennig
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mitaksov V, Truscott SM, Lybarger L, Connolly J, Hansen TH, Fremont DH. Structural engineering of pMHC reagents for T cell vaccines and diagnostics. CHEMISTRY & BIOLOGY 2007; 14:909-22. [PMID: 17719490 PMCID: PMC3601489 DOI: 10.1016/j.chembiol.2007.07.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2006] [Revised: 04/19/2007] [Accepted: 07/02/2007] [Indexed: 11/15/2022]
Abstract
MHC class I peptide complexes (pMHC) are routinely used to enumerate T cell populations and are currently being evaluated as vaccines to tumors and specific pathogens. Herein, we describe the structures of three generations of single-chain pMHC progressively designed for the optimal presentation of covalently associated epitopes. Our ultimate design employs a versatile disulfide trap between an invariant MHC residue and a short C-terminal peptide extension. This general strategy is nondisruptive of native pMHC conformation and T cell receptor engagement. Indeed, cell-surface-expressed MHC complexes with disulfide-trapped epitopes are refractory to peptide exchange, suggesting they will make safe and effective vaccines. Furthermore, we find that disulfide-trap stabilized, recombinant pMHC reagents reliably detect polyclonal CD8 T cell populations as proficiently as conventional reagents and are thus well suited to monitor or modulate immune responses during pathogenesis.
Collapse
Affiliation(s)
- Vesselin Mitaksov
- Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven M. Truscott
- Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lonnie Lybarger
- Cell Biology & Anatomy, University of Arizona Health Sciences Center, Tucson, AZ 85724, U.S.A
| | - Janet Connolly
- Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ted H. Hansen
- Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daved H. Fremont
- Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Biochem. & Mol. Biophysics, Washington University School of Medicine, St. Louis, MO 63110, U.S.A
| |
Collapse
|
43
|
Truscott SM, Lybarger L, Martinko JM, Mitaksov VE, Kranz DM, Connolly JM, Fremont DH, Hansen TH. Disulfide bond engineering to trap peptides in the MHC class I binding groove. THE JOURNAL OF IMMUNOLOGY 2007; 178:6280-9. [PMID: 17475856 DOI: 10.4049/jimmunol.178.10.6280] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunodominant peptides in CD8 T cell responses to pathogens and tumors are not always tight binders to MHC class I molecules. Furthermore, antigenic peptides that bind weakly to the MHC can be problematic when designing vaccines to elicit CD8 T cells in vivo or for the production of MHC multimers for enumerating pathogen-specific T cells in vitro. Thus, to enhance peptide binding to MHC class I, we have engineered a disulfide bond to trap antigenic peptides into the binding groove of murine MHC class I molecules expressed as single-chain trimers or SCTs. These SCTs with disulfide traps, termed dtSCTs, oxidized properly in the endoplasmic reticulum, transited to the cell surface, and were recognized by T cells. Introducing a disulfide trap created remarkably tenacious MHC/peptide complexes because the peptide moiety of the dtSCT was not displaced by high-affinity competitor peptides, even when relatively weak binding peptides were incorporated into the dtSCT. This technology promises to be useful for DNA vaccination to elicit CD8 T cells, in vivo study of CD8 T cell development, and construction of multivalent MHC/peptide reagents for the enumeration and tracking of T cells-particularly when the antigenic peptide has relatively weak affinity for the MHC.
Collapse
Affiliation(s)
- Steven M Truscott
- Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Mazza C, Auphan-Anezin N, Gregoire C, Guimezanes A, Kellenberger C, Roussel A, Kearney A, van der Merwe PA, Schmitt-Verhulst AM, Malissen B. How much can a T-cell antigen receptor adapt to structurally distinct antigenic peptides? EMBO J 2007; 26:1972-83. [PMID: 17363906 PMCID: PMC1847653 DOI: 10.1038/sj.emboj.7601605] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2006] [Accepted: 01/23/2007] [Indexed: 01/15/2023] Open
Abstract
Binding degeneracy is thought to constitute a fundamental property of the T-cell antigen receptor (TCR), yet its structural basis is poorly understood. We determined the crystal structure of a complex involving the BM3.3 TCR and a peptide (pBM8) bound to the H-2K(bm8) major histocompatibility complex (MHC) molecule, and compared it with the structures of the BM3.3 TCR bound to H-2K(b) molecules loaded with two peptides that had a minimal level of primary sequence identity with pBM8. Our findings provide a refined structural view of the basis of BM3.3 TCR cross-reactivity and a structural explanation for the long-standing paradox that a TCR antigen-binding site can be both specific and degenerate. We also measured the thermodynamic features and biological penalties that incurred during cross-recognition. Our data illustrate the difficulty for a given TCR in adapting to distinct peptide-MHC surfaces while still maintaining affinities that result in functional in vivo responses. Therefore, when induction of protective effector T cells is used as the ultimate criteria for adaptive immunity, TCRs are probably much less degenerate than initially assumed.
Collapse
Affiliation(s)
- Catherine Mazza
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerrannée, Marseille Cedex 9, France
- INSERM, U631, Marseille Cedex 9, France
- CNRS, UMR6102, Marseille Cedex 9, France
| | - Nathalie Auphan-Anezin
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerrannée, Marseille Cedex 9, France
- INSERM, U631, Marseille Cedex 9, France
- CNRS, UMR6102, Marseille Cedex 9, France
| | - Claude Gregoire
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerrannée, Marseille Cedex 9, France
- INSERM, U631, Marseille Cedex 9, France
- CNRS, UMR6102, Marseille Cedex 9, France
| | - Annick Guimezanes
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerrannée, Marseille Cedex 9, France
- INSERM, U631, Marseille Cedex 9, France
- CNRS, UMR6102, Marseille Cedex 9, France
| | - Christine Kellenberger
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerrannée, Marseille Cedex 9, France
- INSERM, U631, Marseille Cedex 9, France
- CNRS, UMR6102, Marseille Cedex 9, France
| | - Alain Roussel
- AFMB UMR6098 CNRS, Parc Scientifique de Luminy, Marseille, Cedex 09, France
| | - Alice Kearney
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | | - Anne-Marie Schmitt-Verhulst
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerrannée, Marseille Cedex 9, France
- INSERM, U631, Marseille Cedex 9, France
- CNRS, UMR6102, Marseille Cedex 9, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerrannée, Marseille Cedex 9, France
- INSERM, U631, Marseille Cedex 9, France
- CNRS, UMR6102, Marseille Cedex 9, France
| |
Collapse
|
45
|
Felix NJ, Donermeyer DL, Horvath S, Walters JJ, Gross ML, Suri A, Allen PM. Alloreactive T cells respond specifically to multiple distinct peptide-MHC complexes. Nat Immunol 2007; 8:388-97. [PMID: 17322886 DOI: 10.1038/ni1446] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Accepted: 01/31/2007] [Indexed: 01/07/2023]
Abstract
The molecular basis underlying the specificity of alloreactive T cells for peptide-major histocompatibility complex ligands has been elusive. Here we describe a screen of 60 I-E(k)-alloreactive T cells and 83 naturally processed peptides that identified 9 reactive T cells. Three of the T cells responded to multiple, distinct peptides that shared no sequence homology. These T cells recognized each peptide-major histocompatibility complex ligand specifically and used a distinct constellation of I-E(k) contact residues for each interaction. Our studies show that alloreactive T cells have a 'germline-encoded' capacity to recognize multiple, distinct ligands and thus show 'polyspecificity', not degeneracy. Our findings help to explain the high frequency of alloreactive T cells and provide insight into the nature of T cell specificity.
Collapse
Affiliation(s)
- Nathan J Felix
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63130, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
gammadelta T cells contribute to host immune competence uniquely. This is most likely because they have distinctive antigen-recognition properties. While the basic organization of gammadelta T-cell receptor (TCR) loci is similar to that of alphabeta TCR loci, there is a striking difference in how the diversity of gammadelta TCRs is generated. gammadelta and alphabeta T cells have different antigen-recognition requirements and almost certainly recognize a different set of antigens. While it is unclear what most gammadelta T cells recognize, the non-classical major histocompatibility complex class I molecules T10 and T22 were found to be the natural ligands for a sizable population (0.2-2%) of murine gammadelta T cells. The recognition of T10/T22 may be a way by which gammadelta T cells regulate cells of the immune system, and this system has been used to determine the antigen-recognition determinants of gammadelta T cells. T10/T22-specific gammadelta T cells have TCRs that are diverse in both V gene usage and CDR3 sequences. Their Vgamma usage reflects their tissue origin, and their antigen specificity is conferred by a motif in the TCR delta chain that is encoded by V and D segments and by P-nucleotide addition. Sequence variations around this motif modulate affinities between TCRs and T10/T22. That this CDR3 motif is important in antigen recognition is confirmed by the crystal structure of a gammadelta TCR bound to its ligand. The significance of these observations is discussed in the context of gammadelta T-cell biology.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens/immunology
- Base Sequence
- Complementarity Determining Regions/chemistry
- Complementarity Determining Regions/immunology
- Histocompatibility Antigens/chemistry
- Histocompatibility Antigens/immunology
- Humans
- Lymphocyte Activation/immunology
- Molecular Sequence Data
- Protein Binding/immunology
- Protein Structure, Quaternary
- Receptors, Antigen, T-Cell, gamma-delta/chemistry
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- T-Lymphocyte Subsets/immunology
Collapse
Affiliation(s)
- Yueh-hsiu Chien
- Department of Microbiology and Immunology, Program in Immunology, Stanford University, Stanford, CA, USA.
| | | |
Collapse
|
47
|
Donermeyer DL, Weber KS, Kranz DM, Allen PM. The study of high-affinity TCRs reveals duality in T cell recognition of antigen: specificity and degeneracy. THE JOURNAL OF IMMUNOLOGY 2007; 177:6911-9. [PMID: 17082606 DOI: 10.4049/jimmunol.177.10.6911] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
TCRs exhibit a high degree of Ag specificity, even though their affinity for the peptide/MHC ligand is in the micromolar range. To explore how Ag specificity is achieved, we studied murine T cells expressing high-affinity TCRs engineered by in vitro evolution for binding to hemoglobin peptide/class II complex (Hb/I-Ek). These TCRs were shown previously to maintain Ag specificity, despite having up to 800-fold higher affinity. We compared the response of the high-affinity TCRs and the low-affinity 3.L2 TCR toward a comprehensive set of peptides containing single substitutions at each TCR contact residue. This specificity analysis revealed that the increase in affinity resulted in a dramatic increase in the number of stimulatory peptides. The apparent discrepancy between observed degeneracy in the recognition of single amino acid-substituted Hb peptides and overall Ag specificity of the high-affinity TCRs was examined by generating chimeric peptides between the stimulatory Hb and nonstimulatory moth cytochrome c peptides. These experiments showed that MHC anchor residues significantly affected TCR recognition of peptide. The high-affinity TCRs allowed us to estimate the affinity, in the millimolar range, of immunologically relevant interactions of the TCR with peptide/MHC ligands that were previously unmeasurable because of their weak nature. Thus, through the study of high-affinity TCRs, we demonstrated that a TCR is more tolerant of single TCR contact residue substitutions than other peptide changes, revealing that recognition of Ag by T cells can exhibit both specificity and degeneracy.
Collapse
Affiliation(s)
- David L Donermeyer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
48
|
McMahan RH, McWilliams JA, Jordan KR, Dow SW, Wilson DB, Slansky JE. Relating TCR-peptide-MHC affinity to immunogenicity for the design of tumor vaccines. J Clin Invest 2006; 116:2543-51. [PMID: 16932807 PMCID: PMC1551931 DOI: 10.1172/jci26936] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Accepted: 06/20/2006] [Indexed: 12/22/2022] Open
Abstract
One approach to enhancing the T cell response to tumors is vaccination with mimotopes, mimics of tumor epitopes. While mimotopes can stimulate proliferation of T cells that recognize tumor-associated antigens (TAAs), this expansion does not always correlate with control of tumor growth. We hypothesized that vaccination with mimotopes of optimal affinity in this interaction will improve antitumor immunity. Using a combinatorial peptide library and a cytotoxic T lymphocyte clone that recognizes a TAA, we identified a panel of mimotopes that, when complexed with MHC, bound the TAA-specific TCR with a range of affinities. As expected, in vitro assays showed that the affinity of the TCR-peptide-MHC (TCR-pMHC) interaction correlated with activity of the T cell clone. However, only vaccination with mimotopes in the intermediate-affinity range elicited functional T cells and provided protection against tumor growth in vivo. Vaccination with mimotopes with the highest-affinity TCR-pMHC interactions elicited TAA-specific T cells to the tumor, but did not control tumor growth at any of the peptide concentrations tested. Further analysis of these T cells showed functional defects in response to the TAA. Thus, stimulation of an antitumor response by mimotopes may be optimal with peptides that increase but do not maximize the affinity of the TCR-pMHC interaction.
Collapse
Affiliation(s)
- Rachel H. McMahan
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, Colorado, USA.
Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA.
Torrey Pines Institute for Molecular Studies, San Diego, California, USA
| | - Jennifer A. McWilliams
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, Colorado, USA.
Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA.
Torrey Pines Institute for Molecular Studies, San Diego, California, USA
| | - Kimberly R. Jordan
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, Colorado, USA.
Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA.
Torrey Pines Institute for Molecular Studies, San Diego, California, USA
| | - Steven W. Dow
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, Colorado, USA.
Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA.
Torrey Pines Institute for Molecular Studies, San Diego, California, USA
| | - Darcy B. Wilson
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, Colorado, USA.
Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA.
Torrey Pines Institute for Molecular Studies, San Diego, California, USA
| | - Jill E. Slansky
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, Colorado, USA.
Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA.
Torrey Pines Institute for Molecular Studies, San Diego, California, USA
| |
Collapse
|
49
|
Patarroyo ME, Bermúdez A, Salazar LM, Espejo F. High non-protective, long-lasting antibody levels in malaria are associated with haplotype shifting in MHC–peptide–TCR complex formation: a new mechanism for immune evasion. Biochimie 2006; 88:775-84. [PMID: 16483708 DOI: 10.1016/j.biochi.2006.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2005] [Accepted: 01/06/2006] [Indexed: 11/19/2022]
Abstract
An effective malarial vaccine must contain multiple immunogenic, protection-inducing epitopes able to block and destroy the P. falciparum malaria parasite, the most lethal form of this disease in the world. Our strategy has consisted in using conserved peptides blocking parasite binding to red blood cells; however, these peptides are non-immunogenic and non-protection-inducing. Modifying their critical residues can make them immunogenic. Such peptides induced antibody titers (determined by immunofluorescence antibody test, IFA) and made the latter reactive (determined by Western blot) and protection inducing against experimental challenge with a highly infective Aotus monkey adapted P. falciparum strain. Modified peptides also induce highly non-protective long-lasting antibody levels. Modifications performed might allow them to bind specifically to different HLA-DRbeta purified molecules. These immunological and biological activities are associated with modifications in their three-dimensional structure as determined by (1)H-NMR. It was found that modified, high non-protective long-lasting antibody level peptides bound to HLA-DR molecules from a different haplotype (to which immunogenic, protection-inducers bind) and had 4.6 +/- 1.4 A shorter distances between residues fitting into these molecules' Pocket 1 to Pocket 9, suggesting fitting into an inappropriate HLA-DR molecule. A multi-component, subunit-based, malarial vaccine is therefore feasible if modified peptides are suitably modified for an appropriate fit into the correct HLA-DRbeta1* molecule in order to form a proper MHC-II-peptide-TCR complex.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Protozoan/immunology
- Aotidae
- Blotting, Western
- Computer Simulation
- HLA-DR Antigens/chemistry
- HLA-DR Antigens/immunology
- HLA-DR Antigens/metabolism
- Humans
- Major Histocompatibility Complex/immunology
- Malaria/immunology
- Malaria Vaccines/immunology
- Malaria Vaccines/metabolism
- Models, Molecular
- Molecular Sequence Data
- Peptides/chemical synthesis
- Peptides/chemistry
- Peptides/immunology
- Plasmodium falciparum/immunology
- Protein Binding
- Protein Conformation
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Sequence Homology, Amino Acid
Collapse
Affiliation(s)
- Manuel E Patarroyo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Cra. 50 No. 26-00 Bogotá, Colombia.
| | | | | | | |
Collapse
|
50
|
Patarroyo ME, Cifuentes G, Baquero J. Comparative molecular and three-dimensional analysis of the peptide-MHC II binding region in both human and Aotus MHC-DRB molecules confirms their usefulness in antimalarial vaccine development. Immunogenetics 2006; 58:598-606. [PMID: 16791622 DOI: 10.1007/s00251-006-0128-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Accepted: 05/03/2006] [Indexed: 11/28/2022]
Abstract
A vaccine against malaria is desperately needed, and Aotus monkeys are highly susceptible to experimental infection with malarial parasites. A thorough analysis of this monkey's immune system molecules was thus undertaken in our institute. Cloning and sequencing, followed by three-dimensional analysis, has revealed high homology with some HLA-DRB1 molecules in terms of their peptide binding region pockets. Molecules such as HLA-DRB1*03, 11, 08, and HLA-DRB1*04 are so similar to Aotus MHC-DRB molecules that peptides identified as binding to these molecules and inducing protective immunity in these monkeys could be used in humans without further refinement, while small modifications seem to be needed for those binding to HLA-DRB1*07, HLA-DRB1*15, 16, and HLA-DRB1*10-like molecules, making this New World monkey an excellent model for tailor-made vaccine development, especially against malaria.
Collapse
Affiliation(s)
- M E Patarroyo
- Fundación Instituto de Inmunología de Colombia, Carrera. 50 No. 26-00 Bogotá, Colombia.
| | | | | |
Collapse
|