Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Soc Psychol ; : 1-18, 2022 Oct 13.
Article in English | MEDLINE | ID: mdl-36226679

ABSTRACT

Three experiments tested how communicating attributes of initially liked or disliked groups might create more extreme attitudes. We gave non-neutral participants information about previously unknown groups and asked them to write social media posts describing the group to others. Participants who wrote social media posts to friends (Experiment 1, n = 332) or undecided strangers (Experiments 2 and 3, ns = 113 and 816) exaggerated and elaborated on initial information, subsequently reporting more extreme attitudes. These effects, mediated by extremity of associations to the target group, were interpreted as consistent with theory and research on going beyond the information given. (100 words).

2.
Mol Ther Oncolytics ; 18: 443-456, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32913893

ABSTRACT

Immuno-oncology approaches that utilize T cell receptors (TCRs) are becoming highly attractive because of their potential to target virtually all cellular proteins, including cancer-specific epitopes, via the recognition of peptide-human leukocyte antigen (pHLA) complexes presented at the cell surface. However, because natural TCRs generally recognize cancer-derived pHLAs with very weak affinities, efforts have been made to enhance their binding strength, in some cases by several million-fold. In this study, we investigated the mechanisms underpinning human TCR affinity enhancement by comparing the crystal structures of engineered enhanced affinity TCRs with those of their wild-type progenitors. Additionally, we performed molecular dynamics simulations to better understand the energetic mechanisms driving the affinity enhancements. These data demonstrate that supra-physiological binding affinities can be achieved without altering native TCR-pHLA binding modes via relatively subtle modifications to the interface contacts, often driven through the addition of buried hydrophobic residues. Individual energetic components of the TCR-pHLA interaction governing affinity enhancements were distinct and highly variable for each TCR, often resulting from additive, or knock-on, effects beyond the mutated residues. This comprehensive analysis of affinity-enhanced TCRs has important implications for the future rational design of engineered TCRs as efficacious and safe drugs for cancer treatment.

3.
Orthop J Sports Med ; 8(6): 2325967120927371, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32613023

ABSTRACT

BACKGROUND: Chronic ankle instability (CAI) is linked to mechanical and functional insufficiencies. Joint mobilization is purported to be effective at treating these deficits. PURPOSE: To examine the effect of different treatment durations of a grade IV anterior-to-posterior ankle joint mobilization on weightbearing dorsiflexion range of motion (WB-DFROM), posterior talar glide (PG), and dynamic postural control in individuals with CAI. STUDY DESIGN: Controlled laboratory study. METHODS: A total of 48 female athletes (mean age, 22.8 ± 4.8 years) with unilateral CAI participated in this study. Participants were randomly assigned to 1 of 3 treatment conditions: 30 seconds, 60 seconds, and 120 seconds. Treatment was provided to the injured limb on 3 separate occasions 48 hours apart and consisted of a Maitland grade IV anterior-to-posterior talar joint mobilization based on the participant's initial group assignment. WB-DFROM; PG; and the anterior (ANT), posteromedial (PM), and posterolateral (PL) reach directions of the Star Excursion Balance Test were measured bilaterally before and after each treatment. The uninjured limb acted as a control. Data were analyzed using 2-way mixed-model analyses of variance, and effect sizes were calculated through use of Hedges g. RESULTS: Significant differences were detected after all treatment sessions for all outcome measures (P ≤ .001) and between treatment groups after sessions 1, 2, and 3 for all outcome measures (P ≤ .001). Effect sizes were very large or huge for all treatment groups for WB-DFROM, PG, and ANT reach direction. Substantial variation was found in effect sizes for PM and PL measures. CONCLUSION: Accessory mobilization is an effective treatment to induce acute changes in ankle motion and dynamic postural control in patients with CAI, with longer treatment durations conferring greater improvements. CLINICAL RELEVANCE: This study adds clarity to the use of joint mobilization treatments and will add to the current clinical practice strategy for patients with CAI.

4.
J Clin Invest ; 130(5): 2673-2688, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32310221

ABSTRACT

Tumor-associated peptide-human leukocyte antigen complexes (pHLAs) represent the largest pool of cell surface-expressed cancer-specific epitopes, making them attractive targets for cancer therapies. Soluble bispecific molecules that incorporate an anti-CD3 effector function are being developed to redirect T cells against these targets using 2 different approaches. The first achieves pHLA recognition via affinity-enhanced versions of natural TCRs (e.g., immune-mobilizing monoclonal T cell receptors against cancer [ImmTAC] molecules), whereas the second harnesses an antibody-based format (TCR-mimic antibodies). For both classes of reagent, target specificity is vital, considering the vast universe of potential pHLA molecules that can be presented on healthy cells. Here, we made use of structural, biochemical, and computational approaches to investigate the molecular rules underpinning the reactivity patterns of pHLA-targeting bispecifics. We demonstrate that affinity-enhanced TCRs engage pHLA using a comparatively broad and balanced energetic footprint, with interactions distributed over several HLA and peptide side chains. As ImmTAC molecules, these TCRs also retained a greater degree of pHLA selectivity, with less off-target activity in cellular assays. Conversely, TCR-mimic antibodies tended to exhibit binding modes focused more toward hot spots on the HLA surface and exhibited a greater degree of crossreactivity. Our findings extend our understanding of the basic principles that underpin pHLA selectivity and exemplify a number of molecular approaches that can be used to probe the specificity of pHLA-targeting molecules, aiding the development of future reagents.


Subject(s)
HLA Antigens/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Amino Acid Sequence , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibody Specificity , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line , Cell Line, Tumor , Crystallography, X-Ray , HLA Antigens/chemistry , HLA Antigens/genetics , Humans , Indicators and Reagents , Models, Molecular , Molecular Dynamics Simulation , Molecular Mimicry/genetics , Molecular Mimicry/immunology , Peptides/chemistry , Peptides/genetics , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology
5.
Eur J Immunol ; 49(7): 1052-1066, 2019 07.
Article in English | MEDLINE | ID: mdl-31091334

ABSTRACT

The HLA-A*02:01-restricted decapeptide EAAGIGILTV, derived from melanoma antigen recognized by T-cells-1 (MART-1) protein, represents one of the best-studied tumor associated T-cell epitopes, but clinical results targeting this peptide have been disappointing. This limitation may reflect the dominance of the nonapeptide, AAGIGILTV, at the melanoma cell surface. The decapeptide and nonapeptide are presented in distinct conformations by HLA-A*02:01 and TCRs from clinically relevant T-cell clones recognize the nonapeptide poorly. Here, we studied the MEL5 TCR that potently recognizes the nonapeptide. The structure of the MEL5-HLA-A*02:01-AAGIGILTV complex revealed an induced fit mechanism of antigen recognition involving altered peptide-MHC anchoring. This "flexing" at the TCR-peptide-MHC interface to accommodate the peptide antigen explains previously observed incongruences in this well-studied system and has important implications for future therapeutic approaches. Finally, this study expands upon the mechanisms by which molecular plasticity can influence antigen recognition by T cells.


Subject(s)
Immunodominant Epitopes/metabolism , Immunotherapy, Adoptive/methods , MART-1 Antigen/metabolism , Melanoma/immunology , Peptides/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Amino Acids , Antigen Presentation , Binding Sites , Cells, Cultured , Clone Cells , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/metabolism , Humans , Lymphocyte Activation , MART-1 Antigen/chemistry , Melanoma/therapy , Peptides/chemistry , Protein Binding , Protein Conformation , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/transplantation
6.
J Sports Med Phys Fitness ; 59(12): 1963-1967, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31933342

ABSTRACT

BACKGROUND: A golf bag filled with a set of clubs provides a substantial load. When carried over distance this can increase the demands placed upon the golfer, leading to discomfort, fatigue and injuries. This study aimed to compare the metabolic demands of 2 methods of golf bag carriage. METHODS: A total of 16 healthy male recreational golfers participated in the study. Participants were given an initial familiarization session in which their self-selected walking speed was determined. This was utilized as the treadmill speed for all subsequent trials. The testing protocol consisted of 3 randomized trials of treadmill walking for 5 minutes in each of three conditions: unloaded, single-strap bag and double-strap bag. Equipment consisted of a double-strap golf bag with a standard set of clubs weighing 12.5kg. For all trials oxygen consumption (L·min-1), VO2 (mL·kg·min-1) respiratory minute volume (VE) (L·min-1), and heart rate (HR) were measured. RESULTS: Results showed that the double-strap bag required significantly less oxygen consumption (1.19±0.19 vs. 1.31±0.16 L·min-1, P<0.01) relative oxygen consumption (14.49±2.06 vs. 15.93±2.25 mL·kg·min-1, P<0.01), reduced respiratory minute volume (29.95±4.19 vs. 32.47±4.26 L·min-1, P<0.05), and lower heart rates (100.14±11.05 vs. 106.96±9.33 BPM, P<0.001) than the single-strap bag. Both methods of carriage showed significantly greater metabolic demands than the unloaded condition (P<0.05). CONCLUSIONS: The decreased metabolic cost of carrying a double-strap golf bag may facilitate a reduction in fatigue and reduced mechanical stress. Golf bag transportation must therefore be recognized as a factor in reducing the risk of injury and improving playing performance.


Subject(s)
Equipment and Supplies , Golf/physiology , Adult , Athletes/statistics & numerical data , Energy Metabolism , Exercise Test , Heart Rate , Humans , Male , Middle Aged , Oxygen Consumption , Respiratory Function Tests , Walking
7.
Front Immunol ; 9: 674, 2018.
Article in English | MEDLINE | ID: mdl-29696015

ABSTRACT

T-cell immunity is controlled by T cell receptor (TCR) binding to peptide major histocompatibility complexes (pMHCs). The nature of the interaction between these two proteins has been the subject of many investigations because of its central role in immunity against pathogens, cancer, in autoimmunity, and during organ transplant rejection. Crystal structures comparing unbound and pMHC-bound TCRs have revealed flexibility at the interaction interface, particularly from the perspective of the TCR. However, crystal structures represent only a snapshot of protein conformation that could be influenced through biologically irrelevant crystal lattice contacts and other factors. Here, we solved the structures of three unbound TCRs from multiple crystals. Superposition of identical TCR structures from different crystals revealed some conformation differences of up to 5 Å in individual complementarity determining region (CDR) loops that are similar to those that have previously been attributed to antigen engagement. We then used a combination of rigidity analysis and simulations of protein motion to reveal the theoretical potential of TCR CDR loop flexibility in unbound state. These simulations of protein motion support the notion that crystal structures may only offer an artifactual indication of TCR flexibility, influenced by crystallization conditions and crystal packing that is inconsistent with the theoretical potential of intrinsic TCR motions.


Subject(s)
Complementarity Determining Regions , Receptors, Antigen, T-Cell/chemistry , Computer Simulation , Crystallization , Crystallography, X-Ray , Protein Conformation
8.
Front Immunol ; 8: 1503, 2017.
Article in English | MEDLINE | ID: mdl-29209312

ABSTRACT

Serial accumulation of mutations to fixation in the SLYNTVATL (SL9) immunodominant, HIV p17 Gag-derived, HLA A2-restricted cytotoxic T lymphocyte epitope produce the SLFNTIAVL triple mutant "ultimate" escape variant. These mutations in solvent-exposed residues are believed to interfere with TCR recognition, although confirmation has awaited structural verification. Here, we solved a TCR co-complex structure with SL9 and the triple escape mutant to determine the mechanism of immune escape in this eminent system. We show that, in contrast to prevailing hypotheses, the main TCR contact residue is 4N and the dominant mechanism of escape is not via lack of TCR engagement. Instead, mutation of solvent-exposed residues in the peptide destabilise the peptide-HLA and reduce peptide density at the cell surface. These results highlight the extraordinary lengths that HIV employs to evade detection by high-affinity TCRs with a broad peptide-binding footprint and necessitate re-evaluation of this exemplar model of HIV TCR escape.

9.
Immunol Cell Biol ; 95(1): 68-76, 2017 01.
Article in English | MEDLINE | ID: mdl-27670790

ABSTRACT

The CD8 co-receptor engages peptide-major histocompatibility complex class I (pMHCI) molecules at a largely invariant site distinct from the T-cell receptor (TCR)-binding platform and enhances the sensitivity of antigen-driven activation to promote effective CD8+ T-cell immunity. A small increase in the strength of the pMHCI/CD8 interaction (~1.5-fold) can disproportionately amplify this effect, boosting antigen sensitivity by up to two orders of magnitude. However, recognition specificity is lost altogether with more substantial increases in pMHCI/CD8 affinity (~10-fold). In this study, we used a panel of MHCI mutants with altered CD8-binding properties to show that TCR-mediated antigen specificity is delimited by a pMHCI/CD8 affinity threshold. Our findings suggest that CD8 can be engineered within certain biophysical parameters to enhance the therapeutic efficacy of adoptive T-cell transfer irrespective of antigen specificity.


Subject(s)
CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Cell Membrane/metabolism , Humans , Lymphocyte Activation/immunology , Mutation/genetics , Peptides/metabolism
10.
Proc Natl Acad Sci U S A ; 113(50): 14378-14383, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911793

ABSTRACT

Human γδ T cells display potent responses to pathogens and malignancies. Of particular interest are those expressing a γδ T-cell receptor (TCR) incorporating TCRδ-chain variable-region-2 [Vδ2(+)], which are activated by pathogen-derived phosphoantigens (pAgs), or host-derived pAgs that accumulate in transformed cells or in cells exposed to aminobisphosphonates. Once activated, Vδ2(+) T cells exhibit multiple effector functions that have made them attractive candidates for immunotherapy. Despite this, clinical trials have reported mixed patient responses, highlighting a need for better understanding of Vδ2(+) T-cell biology. Here, we reveal previously unappreciated functional heterogeneity between the Vδ2(+) T-cell compartments of 63 healthy individuals. In this cohort, we identify distinct "Vδ2 profiles" that are stable over time; that do not correlate with age, gender, or history of phosphoantigen activation; and that develop after leaving the thymus. Multiple analyses suggest these Vδ2 profiles consist of variable proportions of two dominant but contrasting Vδ2(+) T-cell subsets that have divergent transcriptional programs and that display mechanistically distinct cytotoxic potentials. Importantly, an individual's Vδ2 profile predicts defined effector capacities, demonstrated by contrasting mechanisms and efficiencies of killing of a range of tumor cell lines. In short, these data support patient stratification to identify individuals with Vδ2 profiles that have effector mechanisms compatible with tumor killing and suggest that tailored Vδ2-profile-specific activation protocols may maximize the chances of future treatment success.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Adolescent , Adult , Aged , CX3C Chemokine Receptor 1/metabolism , Child , Child, Preschool , Cytotoxicity, Immunologic , Female , Gene Expression Profiling , Genes, T-Cell Receptor delta , Healthy Volunteers , Humans , Immunophenotyping , Lymphocyte Activation/immunology , Male , Middle Aged , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, CCR6/metabolism , Young Adult
11.
J Immunol ; 195(12): 5827-36, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26553072

ABSTRACT

Fluorochrome-conjugated peptide-MHC (pMHC) class I multimers are staple components of the immunologist's toolbox, enabling reliable quantification and analysis of Ag-specific CD8(+) T cells irrespective of functional outputs. In contrast, widespread use of the equivalent pMHC class II (pMHC-II) reagents has been hindered by intrinsically weaker TCR affinities for pMHC-II, a lack of cooperative binding between the TCR and CD4 coreceptor, and a low frequency of Ag-specific CD4(+) T cell populations in the peripheral blood. In this study, we show that peptide flanking regions, extending beyond the central nonamer core of MHC-II-bound peptides, can enhance TCR-pMHC-II binding and T cell activation without loss of specificity. Consistent with these findings, pMHC-II multimers incorporating peptide flanking residue modifications proved superior for the ex vivo detection, characterization, and manipulation of Ag-specific CD4(+) T cells, highlighting an unappreciated feature of TCR-pMHC-II interactions.


Subject(s)
Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Separation/methods , HLA Antigens/immunology , Peptide Fragments/metabolism , Cell Line , Histocompatibility Antigens Class II/metabolism , Humans , Lymphocyte Activation , Protein Binding , Receptors, Antigen, T-Cell/immunology , T-Cell Antigen Receptor Specificity
12.
Immunology ; 146(1): 11-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26076649

ABSTRACT

Analysis of antigen-specific T-cell populations by flow cytometry with peptide-MHC (pMHC) multimers is now commonplace. These reagents allow the tracking and phenotyping of T cells during infection, autoimmunity and cancer, and can be particularly revealing when used for monitoring therapeutic interventions. In 2009, we reviewed a number of 'tricks' that could be used to improve this powerful technology. More recent advances have demonstrated the potential benefits of using higher order multimers and of 'boosting' staining by inclusion of an antibody against the pMHC multimer. These developments now allow staining of T cells where the interaction between the pMHC and the T-cell receptor is over 20-fold weaker (K(D) > 1 mm) than could previously be achieved. Such improvements are particularly relevant when using pMHC multimers to stain anti-cancer or autoimmune T-cell populations, which tend to bear lower affinity T-cell receptors. Here, we update our previous work to include discussion of newer tricks that can produce substantially brighter staining even when using log-fold lower concentrations of pMHC multimer. We further provide a practical guide to using pMHC multimers that includes a description of several common pitfalls and how to circumvent them.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Staining and Labeling/methods , Antibodies/immunology , CD8-Positive T-Lymphocytes/cytology , Flow Cytometry/methods , Fluorescent Dyes , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Peptides/immunology , Protein Multimerization
13.
Eur J Immunol ; 45(2): 584-91, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25471691

ABSTRACT

MHC anchor residue-modified "heteroclitic" peptides have been used in many cancer vaccine trials and often induce greater immune responses than the wild-type peptide. The best-studied system to date is the decamer MART-1/Melan-A26-35 peptide, EAAGIGILTV, where the natural alanine at position 2 has been modified to leucine to improve human leukocyte antigen (HLA)-A*0201 anchoring. The resulting ELAGIGILTV peptide has been used in many studies. We recently showed that T cells primed with the ELAGIGILTV peptide can fail to recognize the natural tumor-expressed peptide efficiently, thereby providing a potential molecular reason for why clinical trials of this peptide have been unsuccessful. Here, we solved the structure of a TCR in complex with HLA-A*0201-EAAGIGILTV peptide and compared it with its heteroclitic counterpart , HLA-A*0201-ELAGIGILTV. The data demonstrate that a suboptimal anchor residue at position 2 enables the TCR to "pull" the peptide away from the MHC binding groove, facilitating extra contacts with both the peptide and MHC surface. These data explain how a TCR can distinguish between two epitopes that differ by only a single MHC anchor residue and demonstrate how weak MHC anchoring can enable an induced-fit interaction with the TCR. Our findings constitute a novel demonstration of the extreme sensitivity of the TCR to minor alterations in peptide conformation.


Subject(s)
Alanine/chemistry , Epitopes, T-Lymphocyte/metabolism , HLA-A2 Antigen/chemistry , Leucine/chemistry , MART-1 Antigen/chemistry , Peptides/chemistry , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Alanine/genetics , Amino Acid Sequence , Amino Acid Substitution , Crystallography, X-Ray , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Leucine/genetics , MART-1 Antigen/genetics , MART-1 Antigen/immunology , Models, Molecular , Molecular Sequence Data , Peptides/genetics , Peptides/immunology , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
14.
J Immunol ; 194(1): 463-74, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25452566

ABSTRACT

Fluorochrome-conjugated peptide-MHC (pMHC) multimers are commonly used in combination with flow cytometry for direct ex vivo visualization and characterization of Ag-specific T cells, but these reagents can fail to stain cells when TCR affinity and/or TCR cell-surface density are low. pMHC multimer staining of tumor-specific, autoimmune, or MHC class II-restricted T cells can be particularly challenging, as these T cells tend to express relatively low-affinity TCRs. In this study, we attempted to improve staining using anti-fluorochrome unconjugated primary Abs followed by secondary staining with anti-Ab fluorochrome-conjugated Abs to amplify fluorescence intensity. Unexpectedly, we found that the simple addition of an anti-fluorochrome unconjugated Ab during staining resulted in considerably improved fluorescence intensity with both pMHC tetramers and dextramers and with PE-, allophycocyanin-, or FITC-based reagents. Importantly, when combined with protein kinase inhibitor treatment, Ab stabilization allowed pMHC tetramer staining of T cells even when the cognate TCR-pMHC affinity was extremely low (KD >1 mM) and produced the best results that we have observed to date. We find that this inexpensive addition to pMHC multimer staining protocols also allows improved recovery of cells that have recently been exposed to Ag, improvements in the recovery of self-specific T cells from PBMCs or whole-blood samples, and the use of less reagent during staining. In summary, Ab stabilization of pMHC multimers during T cell staining extends the range of TCR affinities that can be detected, yields considerably enhanced staining intensities, and is compatible with using reduced amounts of these expensive reagents.


Subject(s)
Flow Cytometry/methods , Fluorescent Antibody Technique/methods , Receptors, Antigen, T-Cell/immunology , Staining and Labeling/methods , T-Lymphocytes/immunology , Antibodies/chemistry , Antibodies/immunology , Cells, Cultured , Fluorescent Dyes/chemistry , Humans , Phycocyanin/chemistry , Protein Binding/immunology , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes/cytology
15.
J Biol Chem ; 289(2): 628-38, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24196962

ABSTRACT

αß T-cell receptors (TCRs) engage antigens using complementarity-determining region (CDR) loops that are either germ line-encoded (CDR1 and CDR2) or somatically rearranged (CDR3). TCR ligands compose a presentation platform (major histocompatibility complex (MHC)) and a variable antigenic component consisting of a short "foreign" peptide. The sequence of events when the TCR engages its peptide-MHC (pMHC) ligand remains unclear. Some studies suggest that the germ line elements of the TCR engage the MHC prior to peptide scanning, but this order of binding is difficult to reconcile with some TCR-pMHC structures. Here, we used TCRs that exhibited enhanced pMHC binding as a result of mutations in either CDR2 and/or CDR3 loops, that bound to the MHC or peptide, respectively, to dissect the roles of these loops in stabilizing TCR-pMHC interactions. Our data show that TCR-peptide interactions play a strongly dominant energetic role providing a binding mode that is both temporally and energetically complementary with a system requiring positive selection by self-pMHC in the thymus and rapid recognition of non-self-pMHC in the periphery.


Subject(s)
Complementarity Determining Regions/metabolism , HLA Antigens/metabolism , Peptides/metabolism , Receptors, Antigen, T-Cell/metabolism , Amino Acid Sequence , Binding, Competitive , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Crystallography, X-Ray , HLA Antigens/chemistry , HLA Antigens/genetics , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Kinetics , Ligands , Models, Molecular , Molecular Sequence Data , Mutation , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , T-Cell Antigen Receptor Specificity
16.
Front Immunol ; 4: 172, 2013.
Article in English | MEDLINE | ID: mdl-23847615

ABSTRACT

Recombinant αß T cell receptors, expressed on T cell membranes, recognize short peptides presented at the cell surface in complex with MHC molecules. There are two main subsets of αß T cells: CD8(+) T cells that recognize mainly cytosol-derived peptides in the context of MHC class I (pMHC-I), and CD4(+) T cells that recognize peptides usually derived from exogenous proteins presented by MHC class II (pMHC-II). Unlike the more uniform peptide lengths (usually 8-13mers) bound in the MHC-I closed groove, MHC-II presented peptides are of a highly variable length. The bound peptides consist of a core bound 9mer (reflecting the binding motif for the particular MHC-II type) but with variable peptide flanking residues (PFRs) that can extend from both the N- and C-terminus of the MHC-II binding groove. Although pMHC-I and pMHC-II play a virtually identical role during T cell responses (T cell antigen presentation) and are very similar in overall conformation, there exist a number of subtle but important differences that may govern the functional dichotomy observed between CD8(+) and CD4(+) T cells. Here, we provide an overview of the impact of structural differences between pMHC-I and pMHC-II and the molecular interactions with the T cell receptor including the functional importance of MHC-II PFRs. We consider how factors such as anatomical location, inflammatory milieu, and particular types of antigen presenting cell might, in theory, contribute to the quantitative (i.e., pMHC ligand frequency) as well as qualitative (i.e., variable PFR) nature of peptide epitopes, and hence offer a means of control and influence of a CD4(+) T cell response. Lastly, we review our recent findings showing how modifications to MHC-II PFRs can modify CD4(+) T cell antigen recognition. These findings may have novel applications for the development of CD4(+) T cell peptide vaccines and diagnostics.

17.
J Biol Chem ; 288(26): 18766-75, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23698002

ABSTRACT

The T-cell receptor (TCR) recognizes peptides bound to major histocompatibility molecules (MHC) and allows T-cells to interrogate the cellular proteome for internal anomalies from the cell surface. The TCR contacts both MHC and peptide in an interaction characterized by weak affinity (KD = 100 nM to 270 µM). We used phage-display to produce a melanoma-specific TCR (α24ß17) with a 30,000-fold enhanced binding affinity (KD = 0.6 nM) to aid our exploration of the molecular mechanisms utilized to maintain peptide specificity. Remarkably, although the enhanced affinity was mediated primarily through new TCR-MHC contacts, α24ß17 remained acutely sensitive to modifications at every position along the peptide backbone, mimicking the specificity of the wild type TCR. Thermodynamic analyses revealed an important role for solvation in directing peptide specificity. These findings advance our understanding of the molecular mechanisms that can govern the exquisite peptide specificity characteristic of TCR recognition.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity , Alanine , Biotinylation , Crystallography, X-Ray , Humans , Hydrogen Bonding , Major Histocompatibility Complex , Molecular Conformation , Mutation , Peptide Library , Peptides/metabolism , Protein Binding , Solvents , Surface Plasmon Resonance , Thermodynamics , Water
18.
Sci Rep ; 2: 629, 2012.
Article in English | MEDLINE | ID: mdl-22953050

ABSTRACT

Successful immunity requires that a limited pool of αß T-cell receptors (TCRs) provide cover for a vast number of potential foreign peptide antigens presented by 'self' major histocompatibility complex (pMHC) molecules. Structures of unligated and ligated MHC class-I-restricted TCRs with different ligands, supplemented with biophysical analyses, have revealed a number of important mechanisms that govern TCR mediated antigen recognition. HA1.7 TCR binding to the influenza hemagglutinin antigen (HA(306-318)) presented by HLA-DR1 or HLA-DR4 represents an ideal system for interrogating pMHC-II antigen recognition. Accordingly, we solved the structure of the unligated HA1.7 TCR and compared it to both complex structures. Despite a relatively rigid binding mode, HA1.7 T-cells could tolerate mutations in key contact residues within the peptide epitope. Thermodynamic analysis revealed that limited plasticity and extreme favorable entropy underpinned the ability of the HA1.7 T-cell clone to cross-react with HA(306-318) presented by multiple MHC-II alleles.


Subject(s)
Cross Reactions , HLA-DR1 Antigen/chemistry , HLA-DR4 Antigen/chemistry , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Amino Acid Sequence , Cells, Cultured , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/immunology , HLA-DR1 Antigen/immunology , HLA-DR4 Antigen/immunology , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Lymphocyte Activation , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Thermodynamics
19.
J Immunol Methods ; 382(1-2): 203-10, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22705983

ABSTRACT

The interaction between the clonotypic αß T cell receptor (TCR), expressed on the T cell surface, and peptide-major histocompatibility complex (pMHC) molecules, expressed on the target cell surface, governs T cell mediated autoimmunity and immunity against pathogens and cancer. Structural investigations of this interaction have been limited because of the challenges inherent in the production of good quality TCR/pMHC protein crystals. Here, we report the development of an 'intelligently designed' crystallization screen that reproducibly generates high quality TCR/pMHC complex crystals suitable for X-ray crystallographic studies, thereby reducing protein consumption. Over the last 2 years, we have implemented this screen to produce 32 T cell related protein structures at high resolution, substantially contributing to the current immune protein database. Protein crystallography, used to study this interaction, has already extended our understanding of the molecular rules that govern T cell immunity. Subsequently, these data may help to guide the intelligent design of T cell based therapies that target human diseases, underlining the importance of developing optimized approaches for crystallizing novel TCR/pMHC complexes.


Subject(s)
Crystallization/methods , Major Histocompatibility Complex , Peptides/chemistry , Receptors, Antigen, T-Cell/chemistry , Crystallography, X-Ray , Humans , Major Histocompatibility Complex/immunology , Peptides/immunology , Protein Conformation , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/chemistry , T-Lymphocytes/immunology
20.
Nat Commun ; 3: 665, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22314361

ABSTRACT

Human CD4(+) αß T cells are activated via T-cell receptor recognition of peptide epitopes presented by major histocompatibility complex (MHC) class II (MHC-II). The open ends of the MHC-II binding groove allow peptide epitopes to extend beyond a central nonamer core region at both the amino- and carboxy-terminus. We have previously found that these non-bound C-terminal residues can alter T cell activation in an MHC allele-transcending fashion, although the mechanism for this effect remained unclear. Here we show that modification of the C-terminal peptide-flanking region of an influenza hemagglutinin (HA(305-320)) epitope can alter T-cell receptor binding affinity, T-cell activation and repertoire selection of influenza-specific CD4(+) T cells expanded from peripheral blood. These data provide the first demonstration that changes in the C-terminus of the peptide-flanking region can substantially alter T-cell receptor binding affinity, and indicate a mechanism through which peptide flanking residues could influence repertoire selection.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Epitopes/chemistry , Influenza, Human/immunology , Orthomyxoviridae/metabolism , Biotinylation , CD4-Positive T-Lymphocytes/metabolism , Flow Cytometry/methods , Histocompatibility Antigens Class II/metabolism , Humans , Influenza Vaccines/immunology , Influenza, Human/virology , Models, Genetic , Models, Molecular , Molecular Conformation , Peptides/chemistry , Protein Structure, Tertiary , Surface Plasmon Resonance , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...