Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 17(1): 87-94, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26523866

RESUMEN

The T cell antigen receptor (TCR)-peptide-major histocompatibility complex (MHC) interface is composed of conserved and diverse regions, yet the relative contribution of each in shaping recognition by T cells remains unclear. Here we isolated cross-reactive peptides with limited homology, which allowed us to compare the structural properties of nine peptides for a single TCR-MHC pair. The TCR's cross-reactivity was rooted in highly similar recognition of an apical 'hot-spot' position in the peptide with tolerance of sequence variation at ancillary positions. Furthermore, we found a striking structural convergence onto a germline-mediated interaction between the TCR CDR1α region and the MHC α2 helix in twelve TCR-peptide-MHC complexes. Our studies suggest that TCR-MHC germline-mediated constraints, together with a focus on a small peptide hot spot, might place limits on peptide antigen cross-reactivity.


Asunto(s)
Antígenos/inmunología , Reacciones Cruzadas/inmunología , Activación de Linfocitos/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Secuencia de Aminoácidos , Animales , Antígenos/química , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/inmunología , Unión Proteica/inmunología , Conformación Proteica , Receptores de Antígenos de Linfocitos T alfa-beta/química
2.
Proc Natl Acad Sci U S A ; 120(51): e2312057120, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38085776

RESUMEN

Neoepitopes arising from amino acid substitutions due to single nucleotide polymorphisms are targets of T cell immune responses to cancer and are of significant interest in the development of cancer vaccines. However, understanding the characteristics of rare protective neoepitopes that truly control tumor growth has been a challenge, due to their scarcity as well as the challenge of verifying true, neoepitope-dependent tumor control in humans. Taking advantage of recent work in mouse models that circumvented these challenges, here, we compared the structural and physical properties of neoepitopes that range from fully protective to immunologically inactive. As neoepitopes are derived from self-peptides that can induce immune tolerance, we studied not only how the various neoepitopes differ from each other but also from their wild-type counterparts. We identified multiple features associated with protection, including features that describe how neoepitopes differ from self as well as features associated with recognition by diverse T cell receptor repertoires. We demonstrate both the promise and limitations of neoepitope structural analysis and predictive modeling and illustrate important aspects that can be incorporated into neoepitope prediction pipelines.


Asunto(s)
Neoplasias , Humanos , Animales , Ratones , Epítopos , Neoplasias/genética , Linfocitos T , Péptidos/metabolismo , Antígenos de Neoplasias
3.
J Immunol ; 211(3): 325-332, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37459192

RESUMEN

Recognition of peptide/MHC complexes by αß TCRs has traditionally been viewed through the lens of conventional receptor-ligand theory. Recent work, however, has shown that TCR recognition and T cell signaling can be profoundly influenced and tuned by mechanical forces. One outcome of applied force is the catch bond, where TCR dissociation rates decrease (half-lives increase) when limited force is applied. Although catch bond behavior is believed to be widespread in biology, its counterintuitive nature coupled with the difficulties of describing mechanisms at the structural level have resulted in considerable mystique. In this review, we demonstrate that viewing catch bonds through the lens of energy landscapes, barriers, and the ensuing reaction rates can help demystify catch bonding and provide a foundation on which atomic-level TCR catch bond mechanisms can be built.


Asunto(s)
Receptores de Antígenos de Linfocitos T , Linfocitos T , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Membrana Celular/metabolismo , Transducción de Señal , Unión Proteica
4.
J Immunol ; 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37966257

RESUMEN

Identification of neoepitopes that can control tumor growth in vivo remains a challenge even 10 y after the first genomics-defined cancer neoepitopes were identified. In this study, we identify a neoepitope, resulting from a mutation in the junction plakoglobin (Jup) gene (chromosome 11), from the mouse colon cancer line MC38-FABF (C57BL/6). This neoepitope, Jup mutant (JupMUT), was detected during mass spectrometry of MHC class I-eluted peptides from the tumor. JupMUT has a predicted binding affinity of 564 nM for the Kb molecule and a higher predicted affinity of 82 nM for Db. However, whereas structural modeling of JupMUT and its unmutated counterpart Jup wild-type indicates that there are little conformational differences between the two epitopes bound to Db, large structural divergences are predicted between the two epitopes bound to Kb. Together with in vitro binding data with RMA-S cells, these data suggest that Kb rather than Db is the relevant MHC class I molecule of JupMUT. Immunization of naive C57BL/6 mice with JupMUT elicits CD8-dependent tumor control of a MC38-FABF challenge. Despite the CD8 dependence of JupMUT-mediated tumor control in vivo, CD8+ T cells from JupMUT-immunized mice do not produce higher levels of IFN-γ than do naive mice. The structural and immunological characteristics of JupMUT are substantially different from those of many other neoepitopes that have been shown to mediate tumor control.

5.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33468649

RESUMEN

Presentation of peptides by class I MHC proteins underlies T cell immune responses to pathogens and cancer. The association between peptide binding affinity and immunogenicity has led to the engineering of modified peptides with improved MHC binding, with the hope that these peptides would be useful for eliciting cross-reactive immune responses directed toward their weak binding, unmodified counterparts. Increasing evidence, however, indicates that T cell receptors (TCRs) can perceive such anchor-modified peptides differently than wild-type (WT) peptides, although the scope of discrimination is unclear. We show here that even modifications at primary anchors that have no discernible structural impact can lead to substantially stronger or weaker T cell recognition depending on the TCR. Surprisingly, the effect of peptide anchor modification can be sensed by a TCR at regions distant from the site of modification, indicating a through-protein mechanism in which the anchor residue serves as an allosteric modulator for TCR binding. Our findings emphasize caution in the use and interpretation of results from anchor-modified peptides and have implications for how anchor modifications are accounted for in other circumstances, such as predicting the immunogenicity of tumor neoantigens. Our data also highlight an important need to better understand the highly tunable dynamic nature of class I MHC proteins and the impact this has on various forms of immune recognition.


Asunto(s)
Antígeno HLA-A2/química , Péptidos/química , Receptores de Antígenos de Linfocitos T alfa-beta/química , Células Th2/inmunología , Regulación Alostérica , Sitios de Unión , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Humanos , Células Jurkat , Cinética , Modelos Moleculares , Péptidos/genética , Péptidos/inmunología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Ingeniería de Proteínas , Dominios y Motivos de Interacción de Proteínas , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Células Th2/citología , Termodinámica
6.
J Biol Chem ; 296: 100686, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33891944

RESUMEN

Using a variety of activating and inhibitory receptors, natural killer (NK) cells protect against disease by eliminating cells that have downregulated class I major histocompatibility complex (MHC) proteins, such as in response to cell transformation or viral infection. The inhibitory murine NK receptor Ly49C specifically recognizes the class I MHC protein H-2Kb. Unusual among NK receptors, Ly49C exhibits a peptide-dependent sensitivity to H-2Kb recognition, which has not been explained despite detailed structural studies. To gain further insight into Ly49C peptide sensitivity, we examined Ly49C recognition biochemically and through the lens of dynamic allostery. We found that the peptide sensitivity of Ly49C arises through small differences in H-2Kb-binding affinity. Although molecular dynamics simulations supported a role for peptide-dependent protein dynamics in producing these differences in binding affinity, calorimetric measurements indicated an enthalpically as opposed to entropically driven process. A quantitative linkage analysis showed that this emerges from peptide-dependent dynamic tuning of electrostatic interactions across the Ly49C-H-2Kb interface. We propose a model whereby different peptides alter the flexibility of H-2Kb, which in turn changes the strength of electrostatic interactions across the protein-protein interface. Our results provide a quantitative assessment of how peptides alter Ly49C-binding affinity, suggest the underlying mechanism, and demonstrate peptide-driven allostery at work in class I MHC proteins. Lastly, our model provides a solution for how dynamic allostery could impact binding of some, but not all, class I MHC partners depending on the structural and chemical composition of the interfaces.


Asunto(s)
Células Asesinas Naturales/metabolismo , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Regulación Alostérica , Animales , Cinética , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Subfamilia A de Receptores Similares a Lectina de Células NK/química , Unión Proteica , Dominios Proteicos , Especificidad por Sustrato
7.
Bioinformatics ; 36(22-23): 5377-5385, 2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33355667

RESUMEN

MOTIVATION: The binding of T-cell receptors (TCRs) to their target peptide MHC (pMHC) ligands initializes the cell-mediated immune response. In autoimmune diseases such as multiple sclerosis, the TCR erroneously recognizes self-peptides as foreign and activates an immune response against healthy cells. Such responses can be triggered by cross-recognition of the autoreactive TCR with foreign peptides. Hence, it would be desirable to identify such foreign-antigen triggers to provide a mechanistic understanding of autoimmune diseases. However, the large sequence space of foreign antigens presents an obstacle in the identification of cross-reactive peptides. RESULTS: Here, we present an in silico modeling and scoring method which exploits the structural properties of TCR-pMHC complexes to predict the binding of cross-reactive peptides. We analyzed three mouse TCRs and one human TCR isolated from a patient with multiple sclerosis. Cross-reactive peptides for these TCRs were previously identified via yeast display coupled with deep sequencing, providing a robust dataset for evaluating our method. Modeling query peptides in their associated TCR-pMHC crystal structures, our method accurately selected the top binding peptides from sets containing more than a hundred thousand unique peptides. AVAILABILITY AND IMPLEMENTATION: Analyses were performed using custom Python and R scripts available at https://github.com/weng-lab/antigen-predict. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

8.
Nat Chem Biol ; 16(11): 1269-1276, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32807968

RESUMEN

T-cell recognition of peptides incorporating nonsynonymous mutations, or neoepitopes, is a cornerstone of tumor immunity and forms the basis of new immunotherapy approaches including personalized cancer vaccines. Yet as they are derived from self-peptides, the means through which immunogenic neoepitopes overcome immune self-tolerance are often unclear. Here we show that a point mutation in a non-major histocompatibility complex anchor position induces structural and dynamic changes in an immunologically active ovarian cancer neoepitope. The changes pre-organize the peptide into a conformation optimal for recognition by a neoepitope-specific T-cell receptor, allowing the receptor to bind the neoepitope with high affinity and deliver potent T-cell signals. Our results emphasize the importance of structural and physical changes relative to self in neoepitope immunogenicity. Considered broadly, these findings can help explain some of the difficulties in identifying immunogenic neoepitopes from sequence alone and provide guidance for developing novel, neoepitope-based personalized therapies.


Asunto(s)
Aciltransferasas/metabolismo , Epítopos de Linfocito T/metabolismo , Tolerancia Inmunológica/efectos de los fármacos , Inmunoterapia/métodos , Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Aciltransferasas/genética , Dominio Catalítico , Femenino , Genoma Humano , Humanos , Cinética , Simulación de Dinámica Molecular , Mutación , Neoplasias Ováricas/metabolismo , Unión Proteica , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad , Linfocitos T/metabolismo , Termodinámica
9.
Semin Cell Dev Biol ; 84: 30-41, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30449534

RESUMEN

The role of the αß T cell receptor (TCR) in identifying immunological targets and signaling appropriate responses provides for exciting translational opportunities. Yet TCRs mediate one of the most complex protein-protein interactions in biology, with intricate signaling and selection mechanisms adding additional layers of sophistication. In this review, we discuss how these complexities influence the development and optimization of TCR-based therapeutics, focusing on the intersection between structure, affinity, and specificity. We highlight similarities between TCRs and germline antibodies in molecular recognition, but emphasize that engineering TCRs by mimicking antibody maturation may not translate into improved biological outcomes. A key point is the need to distinguish TCR biochemical recognition from T cell functional recognition and the complications this distinction has for efforts in TCR engineering. We suggest learning from natural immunity and taking advantage of structural features and state-of-the-art protein design principles as a means to optimize TCRs for therapeutic use.


Asunto(s)
Anticuerpos/uso terapéutico , Inmunoterapia , Unión Proteica , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Animales , Anticuerpos/genética , Mutación de Línea Germinal/genética , Humanos , Unión Proteica/genética , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología
10.
Biochemistry ; 59(43): 4163-4175, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33074657

RESUMEN

T cell receptors (TCRs) orchestrate cellular immunity by recognizing peptides presented by a range of major histocompatibility complex (MHC) proteins. Naturally occurring TCRs bind the composite peptide/MHC surface, recognizing peptides that are structurally and chemically compatible with the TCR binding site. Here we describe a molecularly evolved TCR variant that binds the human class I MHC protein HLA-A2 independent of the bound peptide, achieved by a drastic perturbation of the TCR binding geometry that places the molecule far from the peptide binding groove. This unique geometry is unsupportive of normal T cell signaling. A substantial divergence between affinity measurements in solution and in two dimensions between proximal cell membranes leads us to attribute the lack of signaling to steric hindrance that limits binding in the confines of a cell-cell interface. Our results provide an example of how receptor binding geometry can impact T cell function and provide further support for the view that germline-encoded residues in TCR binding loops evolved to drive productive TCR recognition and signaling.


Asunto(s)
Receptores de Antígenos de Linfocitos T/metabolismo , Sitios de Unión , Antígenos HLA-A/metabolismo , Humanos , Complejo Mayor de Histocompatibilidad/genética , Complejo Mayor de Histocompatibilidad/fisiología , Unión Proteica , Conformación Proteica
11.
Proteins ; 88(3): 503-513, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31589793

RESUMEN

Recognition of antigenic peptides bound to major histocompatibility complex (MHC) proteins by αß T cell receptors (TCRs) is a hallmark of T cell mediated immunity. Recent data suggest that variations in TCR binding geometry may influence T cell signaling, which could help explain outliers in relationships between physical parameters such as TCR-pMHC binding affinity and T cell function. Traditionally, TCR binding geometry has been described with simple descriptors such as the crossing angle, which quantifies what has become known as the TCR's diagonal binding mode. However, these descriptors often fail to reveal distinctions in binding geometry that are apparent through visual inspection. To provide a better framework for relating TCR structure to T cell function, we developed a comprehensive system for quantifying the geometries of how TCRs bind peptide/MHC complexes. We show that our system can discern differences not clearly revealed by more common methods. As an example of its potential to impact biology, we used it to reveal differences in how TCRs bind class I and class II peptide/MHC complexes, which we show allow the TCR to maximize access to and "read out" the peptide antigen. We anticipate our system will be of use in not only exploring these and other details of TCR-peptide/MHC binding interactions, but also addressing questions about how TCR binding geometry relates to T cell function, as well as modeling structural properties of class I and class II TCR-peptide/MHC complexes from sequence information. The system is available at https://tcr3d.ibbr.umd.edu/tcr_com or for download as a script.


Asunto(s)
Antígenos de Histocompatibilidad Clase II/química , Antígenos de Histocompatibilidad Clase I/química , Receptores de Antígenos de Linfocitos T alfa-beta/química , Sitios de Unión , Cristalografía por Rayos X , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Modelos Moleculares , Análisis de Componente Principal , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/química , Linfocitos T/inmunología , Linfocitos T/metabolismo , Termodinámica
12.
Trends Immunol ; 38(1): 2-4, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27939452

RESUMEN

Major histocompatibility complex (MHC) restriction is a fundamental tenet of T cell biology, but the underlying mechanisms have remained controversial. The extent to which T cell receptors (TCRs) are biased towards MHC proteins in particular has been widely discussed. In a recent paper, Sharon et al. report direct evidence for coevolution between TCR and MHC genes, helping to explain how MHC compatibility and bias can be encoded within TCRs.


Asunto(s)
Presentación de Antígeno , Evolución Molecular , Antígenos de Histocompatibilidad/metabolismo , Complejo Mayor de Histocompatibilidad/genética , Sitios de Carácter Cuantitativo/genética , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Animales , Ligamiento Genético , Antígenos de Histocompatibilidad/genética , Humanos , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/metabolismo , Relación Estructura-Actividad
13.
Nat Chem Biol ; 14(10): 934-942, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30224695

RESUMEN

T cell receptor cross-reactivity allows a fixed T cell repertoire to respond to a much larger universe of potential antigens. Recent work has emphasized the importance of peptide structural and chemical homology, as opposed to sequence similarity, in T cell receptor cross-reactivity. Surprisingly, though, T cell receptors can also cross-react between ligands with little physiochemical commonalities. Studying the clinically relevant receptor DMF5, we demonstrate that cross-recognition of such divergent antigens can occur through mechanisms that involve heretofore unanticipated rearrangements in the peptide and presenting MHC protein, including binding-induced peptide register shifts and extensions from MHC peptide binding grooves. Moreover, cross-reactivity can proceed even when such dramatic rearrangements do not translate into structural or chemical molecular mimicry. Beyond demonstrating new principles of T cell receptor cross-reactivity, our results have implications for efforts to predict and control T cell specificity and cross-reactivity and highlight challenges associated with predicting T cell reactivities.


Asunto(s)
Oligopéptidos/química , Receptores de Antígenos de Linfocitos T/química , Antígenos/química , Autoinmunidad , Reacciones Cruzadas , Cristalografía por Rayos X , Epítopos/química , Humanos , Cinética , Ligandos , Imitación Molecular , Unión Proteica , Dominios Proteicos , Retroviridae , Resonancia por Plasmón de Superficie , Linfocitos T/química
14.
Mol Ther ; 27(2): 300-313, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30617019

RESUMEN

T cell receptors (TCRs) have emerged as a new class of immunological therapeutics. However, though antigen specificity is a hallmark of adaptive immunity, TCRs themselves do not possess the high specificity of monoclonal antibodies. Although a necessary function of T cell biology, the resulting cross-reactivity presents a significant challenge for TCR-based therapeutic development, as it creates the potential for off-target recognition and immune toxicity. Efforts to enhance TCR specificity by mimicking the antibody maturation process and enhancing affinity can inadvertently exacerbate TCR cross-reactivity. Here we demonstrate this concern by showing that even peptide-targeted mutations in the TCR can introduce new reactivities against peptides that bear similarity to the original target. To counteract this, we explored a novel structure-guided approach for enhancing TCR specificity independent of affinity. Tested with the MART-1-specific TCR DMF5, our approach had a small but discernible impact on cross-reactivity toward MART-1 homologs yet was able to eliminate DMF5 cross-recognition of more divergent, unrelated epitopes. Our study provides a proof of principle for the use of advanced structure-guided design techniques for improving TCR specificity, and it suggests new ways forward for enhancing TCRs for therapeutic use.


Asunto(s)
Receptores de Antígenos de Linfocitos T/metabolismo , Inmunidad Adaptativa/fisiología , Anticuerpos Monoclonales/inmunología , Humanos , Antígeno MART-1/inmunología , Estructura Secundaria de Proteína , Resonancia por Plasmón de Superficie , Especificidad del Receptor de Antígeno de Linfocitos T
15.
Proc Natl Acad Sci U S A ; 114(24): E4792-E4801, 2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28572406

RESUMEN

T-cell receptor (TCR) allorecognition is often presumed to be relatively nonspecific, attributable to either a TCR focus on exposed major histocompatibility complex (MHC) polymorphisms or the degenerate recognition of allopeptides. However, paradoxically, alloreactivity can proceed with high peptide and MHC specificity. Although the underlying mechanisms remain unclear, the existence of highly specific alloreactive TCRs has led to their use as immunotherapeutics that can circumvent central tolerance and limit graft-versus-host disease. Here, we show how an alloreactive TCR achieves peptide and MHC specificity. The HCV1406 TCR was cloned from T cells that expanded when a hepatitis C virus (HCV)-infected HLA-A2- individual received an HLA-A2+ liver allograft. HCV1406 was subsequently shown to recognize the HCV nonstructural protein 3 (NS3):1406-1415 epitope with high specificity when presented by HLA-A2. We show that NS3/HLA-A2 recognition by the HCV1406 TCR is critically dependent on features unique to both the allo-MHC and the NS3 epitope. We also find cooperativity between structural mimicry and a crucial peptide "hot spot" and demonstrate its role, along with the MHC, in directing the specificity of allorecognition. Our results help explain the paradox of specificity in alloreactive TCRs and have implications for their use in immunotherapy and related efforts to manipulate TCR recognition, as well as alloreactivity in general.


Asunto(s)
Receptores de Antígenos de Linfocitos T/metabolismo , Secuencia de Aminoácidos , Línea Celular , Reacciones Cruzadas , Cristalografía por Rayos X , Epítopos/metabolismo , Células HEK293 , Antígeno HLA-A2/química , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Hepacivirus/química , Hepacivirus/genética , Hepacivirus/inmunología , Humanos , Inmunoterapia , Isoantígenos/metabolismo , Células Jurkat , Complejo Mayor de Histocompatibilidad , Modelos Moleculares , Imitación Molecular/genética , Imitación Molecular/inmunología , Péptidos/inmunología , Dominios Proteicos , Linfocitos T/inmunología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología
16.
Cancer Immunol Immunother ; 68(11): 1881-1889, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31595324

RESUMEN

Adoptive cell transfer (ACT) using T cell receptor (TCR) gene-modified T cells is an exciting and rapidly evolving field. Numerous preclinical and clinical studies have demonstrated various levels of feasibility, safety, and efficacy using TCR-engineered T cells to treat cancer and viral infections. Although evidence suggests their use can be effective, to what extent and how to improve these therapeutics are still matters of investigation. As TCR affinity has been generally accepted as the central role in defining T cell specificity and sensitivity, selection for and generation of high affinity TCRs has remained a fundamental approach to design more potent T cells. However, traditional methods for affinity-enhancement by random mutagenesis can induce undesirable cross-reactivity causing on- and off-target adverse events, generate exhausted effectors by overstimulation, and ignore other kinetic and cellular parameters that have been shown to impact antigen specificity. In this Focussed Research Review, we comment on the preclinical and clinical potential of TCR gene-modified T cells, summarize our contributions challenging the role TCR affinity plays in antigen recognition, and explore how structure-guided design can be used to manipulate antigen specificity and TCR cross-reactivity to improve the safety and efficacy of TCR gene-modified T cells used in ACT.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Genes Codificadores de los Receptores de Linfocitos T/inmunología , Inmunoterapia , Neoplasias/terapia , Linfocitos T/inmunología , Linfocitos T/trasplante , Animales , Especificidad de Anticuerpos , Reacciones Cruzadas , Genes Codificadores de los Receptores de Linfocitos T/genética , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T/metabolismo
17.
18.
J Immunol ; 199(7): 2203-2213, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28923982

RESUMEN

T cell specificity emerges from a myriad of processes, ranging from the biological pathways that control T cell signaling to the structural and physical mechanisms that influence how TCRs bind peptides and MHC proteins. Of these processes, the binding specificity of the TCR is a key component. However, TCR specificity is enigmatic: TCRs are at once specific but also cross-reactive. Although long appreciated, this duality continues to puzzle immunologists and has implications for the development of TCR-based therapeutics. In this review, we discuss TCR specificity, emphasizing results that have emerged from structural and physical studies of TCR binding. We show how the TCR specificity/cross-reactivity duality can be rationalized from structural and biophysical principles. There is excellent agreement between predictions from these principles and classic predictions about the scope of TCR cross-reactivity. We demonstrate how these same principles can also explain amino acid preferences in immunogenic epitopes and highlight opportunities for structural considerations in predictive immunology.


Asunto(s)
Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T , Membrana Celular/metabolismo , Reacciones Cruzadas , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Humanos , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
19.
Mol Ther ; 26(4): 996-1007, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29503203

RESUMEN

The use of T cell receptor (TCR) gene-modified T cells in adoptive cell transfer has had promising clinical success, but often, simple preclinical evaluation does not necessarily accurately predict treatment efficacy or safety. Preclinical studies generally evaluate one or a limited number of type 1 cytokines to assess antigen recognition. However, recent studies have implicated other "typed" T cells in effective anti-tumor/viral immunity, and limited functional evaluations may underestimate cross-reactivity. In this study, we use an altered peptide ligand (APL) model and multi-dimensional flow cytometry to evaluate polyfunctionality of TCR gene-modified T cells. Evaluating six cytokines and the lytic marker CD107a on a per cell basis revealed remarkably diverse polyfunctional phenotypes within a single T cell culture and among peripheral blood lymphocyte (PBL) donors. This polyfunctional assessment identified unexpected phenotypes, including cells producing both type 1 and type 2 cytokines, and highlighted interferon γneg (IFNγneg) antigen-reactive populations overlooked in our previous studies. Additionally, APLs skewed functional phenotypes to be less polyfunctional, which was not necessarily related to changes in TCR-peptide-major histocompatibility complex (pMHC) affinity. A better understanding of gene-modified T cell functional diversity may help identify optimal therapeutic phenotypes, predict clinical responses, anticipate off-target recognition, and improve the design and delivery of TCR gene-modified T cells.


Asunto(s)
Péptidos/metabolismo , Fenotipo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Biomarcadores , Células Cultivadas , Citocinas/metabolismo , Citotoxicidad Inmunológica , Antígenos de Histocompatibilidad/química , Antígenos de Histocompatibilidad/inmunología , Antígenos de Histocompatibilidad/metabolismo , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva/métodos , Ligandos , Activación de Linfocitos , Modelos Moleculares , Péptidos/química , Unión Proteica , Conformación Proteica , Receptores de Antígenos de Linfocitos T/química , Relación Estructura-Actividad , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
20.
Proc Natl Acad Sci U S A ; 113(9): E1276-85, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26884163

RESUMEN

How T-cell receptors (TCRs) can be intrinsically biased toward MHC proteins while simultaneously display the structural adaptability required to engage diverse ligands remains a controversial puzzle. We addressed this by examining αß TCR sequences and structures for evidence of physicochemical compatibility with MHC proteins. We found that human TCRs are enriched in the capacity to engage a polymorphic, positively charged "hot-spot" region that is almost exclusive to the α1-helix of the common human class I MHC protein, HLA-A*0201 (HLA-A2). TCR binding necessitates hot-spot burial, yielding high energetic penalties that must be offset via complementary electrostatic interactions. Enrichment of negative charges in TCR binding loops, particularly the germ-line loops encoded by the TCR Vα and Vß genes, provides this capacity and is correlated with restricted positioning of TCRs over HLA-A2. Notably, this enrichment is absent from antibody genes. The data suggest a built-in TCR compatibility with HLA-A2 that biases receptors toward, but does not compel, particular binding modes. Our findings provide an instructional example for how structurally pliant MHC biases can be encoded within TCRs.


Asunto(s)
Antígeno HLA-A2/química , Receptores de Antígenos de Linfocitos T alfa-beta/química , Humanos , Conformación Proteica , Electricidad Estática
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA