Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Infect Dis ; 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38060822

ABSTRACT

BACKGROUND: Excessive pulmonary inflammation and damage are characteristic features of severe influenza virus infections. LAT8881 is a synthetic, 16 amino acid cyclic peptide form of a naturally occurring C-terminal fragment of human growth hormone with therapeutic efficacy against influenza. Shorter, linear peptides are typically easier to manufacture and formulate for delivery than larger cyclic peptides. A 6 amino acid linear peptide fragment of LAT8881, LAT9997, was investigated as a potential influenza therapy. METHODS: LAT9997 was evaluated for its potential to limit disease in a preclinical mouse model of severe influenza infection. RESULTS: Intranasal treatment of mice with either LAT8881 or LAT9997 from day 1 following influenza infection significantly improved survival outcomes. Initiating LAT9997 treatment at the onset of severe disease, also significantly improved disease severity. Greater disease resistance in LAT9997-treated mice correlated with reduced lung immunopathology, damage markers, vascular leak, and epithelial cell death. Treatment reduced viral loads, cytokines, and neutrophil infiltration in the airways, yet maintained protective alveolar macrophages in a dose-dependent manner. Sequential trimming of N- and C-terminal amino acids from LAT9997 revealed a structure-activity relationship. CONCLUSIONS: These findings provide preclinical evidence that therapeutic LAT9997 treatment limits viral burden and characteristic features of severe influenza, including hyperinflammation and lung damage.

2.
Cell ; 184(4): 983-999.e24, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33606986

ABSTRACT

Interleukin-12 (IL-12) and IL-23 are heterodimeric cytokines that are produced by antigen-presenting cells to regulate the activation and differentiation of lymphocytes, and they share IL-12Rß1 as a receptor signaling subunit. We present a crystal structure of the quaternary IL-23 (IL-23p19/p40)/IL-23R/IL-12Rß1 complex, together with cryoelectron microscopy (cryo-EM) maps of the complete IL-12 (IL-12p35/p40)/IL-12Rß2/IL-12Rß1 and IL-23 receptor (IL-23R) complexes, which reveal "non-canonical" topologies where IL-12Rß1 directly engages the common p40 subunit. We targeted the shared IL-12Rß1/p40 interface to design a panel of IL-12 partial agonists that preserved interferon gamma (IFNγ) induction by CD8+ T cells but impaired cytokine production from natural killer (NK) cells in vitro. These cell-biased properties were recapitulated in vivo, where IL-12 partial agonists elicited anti-tumor immunity to MC-38 murine adenocarcinoma absent the NK-cell-mediated toxicity seen with wild-type IL-12. Thus, the structural mechanism of receptor sharing used by IL-12 family cytokines provides a protein interface blueprint for tuning this cytokine axis for therapeutics.


Subject(s)
Interleukin-12/chemistry , Interleukin-12/metabolism , Killer Cells, Natural/metabolism , Receptors, Interleukin/chemistry , Receptors, Interleukin/metabolism , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cryoelectron Microscopy , Crystallography, X-Ray , Epitopes/immunology , Female , HEK293 Cells , Humans , Immunity , Interleukin-12/agonists , Interleukin-12 Subunit p40/chemistry , Interleukin-12 Subunit p40/metabolism , Mice, Inbred C57BL , Models, Molecular , Neoplasms/immunology , Neoplasms/pathology , Protein Structure, Quaternary , Receptors, Interleukin/ultrastructure , Receptors, Interleukin-12/metabolism , Signal Transduction , Structure-Activity Relationship
3.
Nat Med ; 24(7): 1005-1014, 2018 07.
Article in English | MEDLINE | ID: mdl-29942088

ABSTRACT

Interleukin-2 (IL-2) has been shown to suppress immune pathologies by preferentially expanding regulatory T cells (Tregs). However, this therapy has been limited by off-target complications due to pathogenic cell expansion. Recent efforts have been focused on developing a more selective IL-2. It is well documented that certain anti-mouse IL-2 antibodies induce conformational changes that result in selective targeting of Tregs. We report the generation of a fully human anti-IL-2 antibody, F5111.2, that stabilizes IL-2 in a conformation that results in the preferential STAT5 phosphorylation of Tregs in vitro and selective expansion of Tregs in vivo. When complexed with human IL-2, F5111.2 induced remission of type 1 diabetes in the NOD mouse model, reduced disease severity in a model of experimental autoimmune encephalomyelitis and protected mice against xenogeneic graft-versus-host disease. These results suggest that IL-2-F5111.2 may provide an immunotherapy to treat autoimmune diseases and graft-versus-host disease.


Subject(s)
Antibodies/chemistry , Antibodies/pharmacology , Interleukin-2/immunology , T-Lymphocytes, Regulatory/drug effects , Animals , Antibodies/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/immunology , Humans , Immunoglobulin Fab Fragments/metabolism , Immunotherapy , Kinetics , Mice, Inbred C57BL , Models, Molecular , Muromegalovirus/drug effects , Phosphorylation/drug effects , Protein Binding/drug effects , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Up-Regulation/drug effects
4.
Structure ; 26(2): 187-198.e4, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29336885

ABSTRACT

Coagulation factor XIa is a candidate target for anticoagulants that better separate antithrombotic efficacy from bleeding risk. We report a co-crystal structure of the FXIa protease domain with DEF, a human monoclonal antibody that blocks FXIa function and prevents thrombosis in animal models without detectable increased bleeding. The light chain of DEF occludes the FXIa S1 subsite and active site, while the heavy chain provides electrostatic interactions with the surface of FXIa. The structure accounts for the specificity of DEF for FXIa over its zymogen and related proteases, its active-site-dependent binding, and its ability to inhibit substrate cleavage. The inactive FXIa protease domain used to obtain the DEF-FXIa crystal structure reversed anticoagulant activity of DEF in plasma and in vivo and the activity of a small-molecule FXIa active-site inhibitor in vitro. DEF and this reversal agent for FXIa active-site inhibitors may help support clinical development of FXIa-targeting anticoagulants.


Subject(s)
Antibodies, Monoclonal/metabolism , Factor XIa/metabolism , Animals , Anticoagulants , Binding Sites, Antibody , Humans , Protein Conformation , Thrombosis/metabolism
5.
Sci Transl Med ; 8(353): 353ra112, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27559095

ABSTRACT

Thrombosis is a major cause of morbidity and mortality. Current antithrombotic drugs are not ideal in that they must balance prevention of thrombosis against bleeding risk. Inhibition of coagulation factor XI (FXI) may offer an improvement over existing antithrombotic strategies by preventing some forms of thrombosis with lower bleeding risk. To permit exploration of this hypothesis in humans, we generated and characterized a series of human immunoglobulin Gs (IgGs) that blocked FXIa active-site function but did not bind FXI zymogen or other coagulation proteases. The most potent of these IgGs, C24 and DEF, inhibited clotting in whole human blood and prevented FeCl3-induced carotid artery occlusion in FXI-deficient mice reconstituted with human FXI and in thread-induced venous thrombosis in rabbits at clinically relevant doses. At doses substantially higher than those required for inhibition of intravascular thrombus formation in these models, DEF did not increase cuticle bleeding in rabbits or cause spontaneous bleeding in macaques over a 2-week study. Anticipating the desirability of a reversal agent, we also generated a human IgG that rapidly reversed DEF activity ex vivo in human plasma and in vivo in rabbits. Thus, an active site-directed FXIa-specific antibody can block thrombosis in animal models and, together with the reversal agent, may facilitate exploration of the roles of FXIa in human disease.


Subject(s)
Factor XI/physiology , Factor XIa/antagonists & inhibitors , Factor XIa/immunology , Hemostasis/physiology , Immunoglobulin G/metabolism , Thrombosis/blood , Animals , Antibodies, Blocking/metabolism , Antibodies, Monoclonal/metabolism , Antibody Specificity , Humans , In Vitro Techniques , Kinetics , Macaca fascicularis , Mice , Rabbits
6.
J Immunol ; 186(10): 5823-32, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21490152

ABSTRACT

T cells specific for the cytochrome c Ag are widely used to investigate many aspects of TCR specificity and interactions with peptide-MHC, but structural information has long been elusive. In this study, we present structures for the well-studied 2B4 TCR, as well as a naturally occurring variant of the 5c.c7 TCR, 226, which is cross-reactive with more than half of possible substitutions at all three TCR-sensitive residues on the peptide Ag. These structures alone and in complex with peptide-MHC ligands allow us to reassess many prior mutagenesis results. In addition, the structure of 226 bound to one peptide variant, p5E, shows major changes in the CDR3 contacts compared with wild-type, yet the TCR V-region contacts with MHC are conserved. These and other data illustrate the ability of TCRs to accommodate large variations in CDR3 structure and peptide contacts within the constraints of highly conserved TCR-MHC interactions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytochromes c/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Antigens, CD/chemistry , Antigens, CD/immunology , Antigens, CD/metabolism , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Cross Reactions , Crystallography, X-Ray , Cytochromes c/metabolism , Humans , Ligands , Mice , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Receptors, Immunologic/chemistry , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Signaling Lymphocytic Activation Molecule Family , Surface Plasmon Resonance
7.
Proc Natl Acad Sci U S A ; 107(12): 5534-9, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20212169

ABSTRACT

Residues within processed protein fragments bound to major histocompatibility complex class I (MHC-I) glycoproteins have been considered to function as a series of "independent pegs" that either anchor the peptide (p) to the MHC-I and/or interact with the spectrum of alphabeta-T-cell receptors (TCRs) specific for the pMHC-I epitope in question. Mining of the extensive pMHC-I structural database established that many self- and viral peptides show extensive and direct interresidue interactions, an unexpected finding that has led us to the idea of "constrained" peptides. Mutational analysis of two constrained peptides (the HLA B44 restricted self-peptide (B44DPalpha-EEFGRAFSF) and an H2-D(b) restricted influenza peptide (D(b)PA, SSLENFRAYV) demonstrated that the conformation of the prominently exposed arginine in both peptides was governed by interactions with MHC-I-orientated flanking residues from the peptide itself. Using reverse genetics in a murine influenza model, we revealed that mutation of an MHC-I-orientated residue (SSLENFRAYV --> SSLENARAYV) within the constrained PA peptide resulted in a diminished cytotoxic T lymphocyte (CTL) response and the recruitment of a limited pMHC-I specific TCR repertoire. Interactions between individual peptide positions can thus impose fine control on the conformation of pMHC-I epitopes, whereas the perturbation of such constraints can lead to a previously unappreciated mechanism of viral escape.


Subject(s)
Histocompatibility Antigens Class I/metabolism , T-Lymphocytes/immunology , Amino Acid Sequence , Amino Acid Substitution , Animals , Antigen Presentation , Epitopes/chemistry , Epitopes/genetics , Epitopes/metabolism , Female , H-2 Antigens/chemistry , H-2 Antigens/genetics , H-2 Antigens/metabolism , HLA-B Antigens/chemistry , HLA-B Antigens/genetics , HLA-B Antigens/metabolism , HLA-B44 Antigen , Histocompatibility Antigen H-2D , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Humans , Ligands , Mice , Mice, Inbred C57BL , Models, Molecular , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Protein Conformation , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology
8.
Nat Immunol ; 10(12): 1245-51, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19838198

ABSTRACT

Interleukin 17 (IL-17)-producing helper T cells (T(H)-17 cells), together with their effector cytokines, including members of the IL-17 family, are emerging as key mediators of chronic inflammatory and autoimmune disorders. Here we present the crystal structure of a complex of IL-17 receptor A (IL-17RA) bound to IL-17F in a 1:2 stoichiometry. The mechanism of complex formation was unique for cytokines and involved the engagement of IL-17 by two fibronectin-type domains of IL-17RA in a groove between the IL-17 homodimer interface. Binding of the first receptor to the IL-17 cytokines modulated the affinity and specificity of the second receptor-binding event, thereby promoting heterodimeric versus homodimeric complex formation. IL-17RA used a common recognition strategy to bind to several members of the IL-17 family, which allows it to potentially act as a shared receptor in multiple different signaling complexes.


Subject(s)
Interleukin-17/chemistry , Interleukin-17/metabolism , Protein Multimerization , Receptors, Interleukin-17/chemistry , Receptors, Interleukin-17/metabolism , Amino Acid Sequence , Cell Line , Conserved Sequence , Crystallography, X-Ray , Humans , Interleukin-17/immunology , Models, Molecular , Molecular Sequence Data , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, Interleukin-17/immunology , Sequence Alignment
9.
Immunity ; 30(6): 777-88, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19464197

ABSTRACT

Ligation of the alphabeta T cell receptor (TCR) by a specific peptide-loaded major histocompatibility complex (pMHC) molecule initiates T cell signaling via the CD3 complex. However, the initial events that link antigen recognition to T cell signal transduction remain unclear. Here we show, via fluorescence-based experiments and structural analyses, that MHC-restricted antigen recognition by the alphabeta TCR results in a specific conformational change confined to the A-B loop within the alpha chain of the constant domain (Calpha). The apparent affinity constant of this A-B loop movement mirrored that of alphabeta TCR-pMHC ligation and was observed in two alphabeta TCRs with distinct pMHC specificities. The Ag-induced A-B loop conformational change could be inhibited by fixing the juxtapositioning of the constant domains and was shown to be reversible upon pMHC disassociation. Notably, the loop movement within the Calpha domain, although specific for an agonist pMHC ligand, was not observed with a pMHC antagonist. Moreover, mutagenesis of residues within the A-B loop impaired T cell signaling in an in vitro system of antigen-specific TCR stimulation. Collectively, our findings provide a basis for the earliest molecular events that underlie Ag-induced T cell triggering.


Subject(s)
Antigens/chemistry , Receptors, Antigen, T-Cell, alpha-beta/chemistry , T-Lymphocytes/immunology , Animals , Antigens/immunology , Humans , Major Histocompatibility Complex/immunology , Mutation/genetics , Peptides/chemistry , Peptides/immunology , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology
10.
Nat Immunol ; 10(2): 143-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19148199

ABSTRACT

The elusive etiology of germline bias of the T cell receptor (TCR) for major histocompatibility complex (MHC) has been clarified by recent 'proof-of-concept' structural results demonstrating the conservation of specific TCR-MHC interfacial contacts in complexes bearing common variable segments and MHC allotypes. We suggest that each TCR variable-region gene product engages each type of MHC through a 'menu' of structurally coded recognition motifs that have arisen through coevolution. The requirement for MHC-restricted T cell recognition during thymic selection and peripheral surveillance has necessitated the existence of such a coded recognition system. Given these findings, a reconsideration of the TCR-peptide-MHC structural database shows that not only have the answers been there all along but also they were predictable by the first principles of physical chemistry.


Subject(s)
Major Histocompatibility Complex/immunology , Protein Structure, Quaternary , Receptors, Antigen, T-Cell/chemistry , Animals , Genes, T-Cell Receptor , Humans , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
11.
J Exp Med ; 206(1): 209-19, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19139173

ABSTRACT

Human leukocyte antigen (HLA) gene polymorphism plays a critical role in protective immunity, disease susceptibility, autoimmunity, and drug hypersensitivity, yet the basis of how HLA polymorphism influences T cell receptor (TCR) recognition is unclear. We examined how a natural micropolymorphism in HLA-B44, an important and large HLA allelic family, affected antigen recognition. T cell-mediated immunity to an Epstein-Barr virus determinant (EENLLDFVRF) is enhanced when HLA-B*4405 was the presenting allotype compared with HLA-B*4402 or HLA-B*4403, each of which differ by just one amino acid. The micropolymorphism in these HLA-B44 allotypes altered the mode of binding and dynamics of the bound viral epitope. The structure of the TCR-HLA-B*4405(EENLLDFVRF) complex revealed that peptide flexibility was a critical parameter in enabling preferential engagement with HLA-B*4405 in comparison to HLA-B*4402/03. Accordingly, major histocompatibility complex (MHC) polymorphism can alter the dynamics of the peptide-MHC landscape, resulting in fine-tuning of T cell responses between closely related allotypes.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HLA-B Antigens/genetics , Polymorphism, Single Nucleotide , Receptors, Antigen, T-Cell/immunology , Amino Acid Substitution , Binding Sites , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Epitopes, T-Lymphocyte/genetics , Epstein-Barr Virus Nuclear Antigens/immunology , HLA Antigens/genetics , HLA Antigens/metabolism , HLA-B Antigens/chemistry , HLA-B Antigens/metabolism , HLA-B44 Antigen , Humans , Hydrogen Bonding , Kinetics , Models, Molecular , Protein Binding , Protein Conformation , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Recombinant Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology
12.
Curr Opin Immunol ; 20(5): 575-80, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18678247

ABSTRACT

T-cells play a critical role in protective immunity, with their broad receptor repertoire capable of engaging diverse foreign pMHC landscapes. While the versatility and specificity of this MHC-restricted response is the hallmark of adaptive immunity, unwanted TCR interactions can profoundly effect the health of the host leading for instance to allograft rejection or autoimmunity. In allogeneic transplantation, such adverse reactions can occur by an indirect pathway when the TCR interacts with self-MHC molecules presenting allogeneic MHC derived peptides. Direct T-cell alloreactivity involves recognition of the allogeneic molecule itself either through molecular mimicry or by novel pMHC binding modes. By contrast, auto-reactive TCRs are considered to interact in a manner distinct from cognate pMHC interactions. Here we review recent advances in the field, focusing on structural data pertaining to alloreactivity and auto-reactivity and discuss implications for T-cell mediated transplant rejection and autoimmune disorders.


Subject(s)
Autoimmunity/immunology , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Humans , Major Histocompatibility Complex/genetics , Protein Binding/immunology , Protein Conformation , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/metabolism
13.
J Mol Biol ; 377(5): 1297-303, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18339401

ABSTRACT

Human leukocyte antigen (HLA)-E is a non-classical major histocompatibility complex class I molecule that binds peptides derived from the leader sequences of other HLA class I molecules. Natural killer cell recognition of these HLA-E molecules, via the CD94-NKG2 natural killer family, represents a central innate mechanism for monitoring major histocompatibility complex expression levels within a cell. The leader sequence-derived peptides bound to HLA-E exhibit very limited polymorphism, yet subtle differences affect the recognition of HLA-E by the CD94-NKG2 receptors. To better understand the basis for this peptide-specific recognition, we determined the structure of HLA-E in complex with two leader peptides, namely, HLA-Cw*07 (VMAPRALLL), which is poorly recognised by CD94-NKG2 receptors, and HLA-G*01 (VMAPRTLFL), a high-affinity ligand of CD94-NKG2 receptors. A comparison of these structures, both of which were determined to 2.5-A resolution, revealed that allotypic variations in the bound leader sequences do not result in conformational changes in the HLA-E heavy chain, although subtle changes in the conformation of the peptide within the binding groove of HLA-E were evident. Accordingly, our data indicate that the CD94-NKG2 receptors interact with HLA-E in a manner that maximises the ability of the receptors to discriminate between subtle changes in both the sequence and conformation of peptides bound to HLA-E.


Subject(s)
HLA Antigens/immunology , Histocompatibility Antigens Class I/immunology , NK Cell Lectin-Like Receptor Subfamily D/immunology , Receptors, Immunologic/immunology , HLA Antigens/chemistry , Histocompatibility Antigens Class I/chemistry , Humans , Immunity, Innate/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Protein Conformation , Receptors, Immunologic/metabolism , HLA-E Antigens
14.
Mol Immunol ; 45(3): 583-98, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17869342

ABSTRACT

A great paradox in cellular immunology is how T cell allorecognition exists at high frequencies (up to 10%) despite the stringent requirements of discriminating 'self' from 'non-self' imposed by MHC restriction. Thus, in tissue transplantation, a substantial proportion of the recipient's T cells will have the ability to recognize the graft and instigate an immune response against the transplanted tissue, ultimately resulting in graft rejection--a manifestation of T cell alloreactivity. Transplantation of human organs and lymphoid cells as treatment for otherwise life-threatening diseases has become a more routine medical procedure making this problem of great importance. Immunologists have gained important insights into the mechanisms of T cell alloreactivity from cytotoxic T cell assays, affinity-avidity studies, and crystal structures of peptide-MHC (pMHC) molecules and T cell receptors (TCRs) both alone and in complex. Despite the clinical significance of alloreactivity, the crystal structure of an alloreactive human TCR in complex with both cognate pMHC and an allogeneic pMHC complex has yet to be determined. This review highlights some of the important findings from studies characterizing the way in which alloreactive T cell receptors and pMHC molecules interact in an attempt to resolve this great irony of the cellular immune response.


Subject(s)
Antigen Presentation/immunology , Histocompatibility Antigens/immunology , Isoantigens/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Transplantation Immunology , Animals , Graft Rejection/immunology , Humans , Receptors, Antigen, T-Cell/immunology , Tissue Transplantation
15.
Nat Immunol ; 8(9): 975-83, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17694060

ABSTRACT

All complexes of T cell receptors (TCRs) bound to peptide-major histocompatibility complex (pMHC) molecules assume a stereotyped binding 'polarity', despite wide variations in TCR-pMHC docking angles. However, existing TCR-pMHC crystal structures have failed to show broadly conserved pairwise interaction motifs. Here we determined the crystal structures of two TCRs encoded by the variable beta-chain 8.2 (V(beta)8.2), each bound to the MHC class II molecule I-A(u), and did energetic mapping of V(alpha) and V(beta) contacts with I-A(u). Together with two previously solved structures of V(beta)8.2-containing TCR-MHC complexes, we found four TCR-I-A complexes with structurally superimposable interactions between the V(beta) loops and the I-A alpha-helix. This examination of a narrow 'slice' of the TCR-MHC repertoire demonstrates what is probably one of many germline-derived TCR-MHC interaction 'codons'.


Subject(s)
Histocompatibility Antigens Class II/chemistry , Major Histocompatibility Complex , Protein Structure, Quaternary , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Amino Acid Sequence , Animals , Crystallization , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice , Molecular Sequence Data , Protein Binding , Receptors, Antigen, T-Cell, alpha-beta/genetics , Surface Plasmon Resonance
16.
Proc Natl Acad Sci U S A ; 103(17): 6641-6, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16617112

ABSTRACT

The underlying basis of major histocompatibility complex (MHC) restriction is unclear. Nevertheless, current data suggest that a common thermodynamic signature dictates alphabeta T cell receptor (TcR) ligation. To evaluate whether this thermodynamic signature defines MHC restriction, we have examined the thermodynamic basis of a highly characterized immunodominant TcR interacting with its cognate peptide-MHC-I ligand. Surprisingly, we observed this interaction to be governed by favorable enthalpic and entropic forces, which is in contrast to the prevailing generality, namely, enthalpically driven interactions combined with markedly unfavorable entropic forces. We conclude that extrinsic molecular factors, such as coreceptor ligation, conformational adjustments involved in TcR signaling, or constraints dictated by higher-order arrangement of ligated TcRs, might play a greater role in guiding MHC restriction than appreciated previously.


Subject(s)
HLA-B Antigens/chemistry , HLA-B Antigens/metabolism , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Amino Acid Sequence , Binding Sites , Entropy , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , HLA-B8 Antigen , Humans , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Ligands , Models, Molecular , Multiprotein Complexes , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction , Surface Plasmon Resonance , T-Lymphocytes, Cytotoxic/immunology , Thermodynamics , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/metabolism
17.
Nat Immunol ; 7(3): 256-64, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16474394

ABSTRACT

In contrast to antigen-specific immunity orchestrated by major histocompatibility complex (MHC) class Ia molecules, the ancestrally related nonclassical MHC class Ib molecules generally mediate innate immune responses. Here we have demonstrated the structural basis by which the MHC class Ib molecule HLA-E mediates an adaptive MHC-restricted cytotoxic T lymphocyte response to human cytomegalovirus. Highly constrained by host genetics, the response showed notable fine specificity for position 8 of the viral peptide, which is the sole discriminator of self versus nonself. Despite the evolutionary divergence of MHC class Ia and class Ib molecules, the structure of the T cell receptor-MHC class Ib complex was very similar to that of conventional T cell receptor-MHC class Ia complexes. These results emphasize the evolutionary 'ambiguity' of HLA-E, which not only interacts with innate immune receptors but also has the functional capacity to mediate virus-specific cytotoxic T lymphocyte responses during adaptive immunity.


Subject(s)
Histocompatibility Antigens Class I/chemistry , Receptors, Antigen, T-Cell/chemistry , T-Lymphocytes/immunology , Animals , Cytomegalovirus/immunology , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Humans , Lymphocyte Activation/immunology , Protein Structure, Quaternary , Receptors, Antigen, T-Cell/immunology
18.
IUBMB Life ; 57(8): 575-82, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16118115

ABSTRACT

Alphabeta T-cell receptor (TcR) recognition of antigenic peptides bound to the major histocompatibility complex (pMHC), is integral to the cellular immune system. Crystallographic studies over the last decade have provided significant insight into this unique trimolecular recognition event. The TcR-pMHC structural information has been paralleled by biophysical studies that have further explored the emerging binding models in an attempt to answer fundamental immunological questions regarding MHC restriction, T-cell immunodominance and TcR cross-reactivity. However, despite the important data that has been generated regarding TcR-pMHC interactions, the scope of this information is still incomplete due to the limited range of TcRs that have been studied. These limitations are primarily due to difficulties in obtaining high yields of recombinant alphabeta TcR for crystallographic and biophysical analysis; here we will discuss some of the protein engineering strategies that have been employed to expand the pool of recombinant TcRs suitable for crystallographic studies and the subsequent studies that have utilized these proteins.


Subject(s)
Biophysics/methods , Immunity, Cellular/genetics , Models, Molecular , Protein Engineering/methods , Receptors, Antigen, T-Cell, alpha-beta/genetics , Recombinant Proteins/genetics , Humans , Major Histocompatibility Complex/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Recombinant Proteins/metabolism
19.
J Immunol ; 174(9): 5593-601, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843558

ABSTRACT

Alloreactive T lymphocytes are central mediators of graft-versus-host disease and allograft rejection. A public CTL clonotype with specificity for the alloantigens HLA-B*4402 and B*4405 is often expanded to large numbers in healthy HLA-B*0801(+) individuals, driven by cross-reactive stimulation with the common, persistent herpesvirus EBV. Since such alloreactive memory CTL expansions have the potential to influence transplantation outcome, altered peptide ligands (APLs) of the target HLA-B*0801-binding EBV peptide, FLRGRAYGL, were screened as specific antagonists for this immunodominant clonotype. One APL, FLRGRFYGL, exerted powerful antagonism of a prototypic T cell clone expressing this immunodominant TCR when costimulated with target cells presenting HLA-B*0801(FLRGRAYGL). Significantly, this APL also reduced the lysis of allogeneic target cells expressing HLA-B*4402 by up to 99%. The affinities of the agonist and antagonist complexes for the public TCR, measured using solution and solid-phase assays, were 8 and 138 muM, respectively. Surprisingly, the half-life of the agonist and antagonist complexes was similar, yet the association rate for the antagonist complex was significantly slower. These observations were further supported by structural studies that suggested a large conformational hurdle was required to ligate the immunodominant TCR to the HLA-B*0801 antagonist complex. By defining an antagonist APL against an immunodominant alloreactive TCR, these findings raise the prospect of exploiting such peptides to inhibit clinical alloreactivity, particularly against clonal T cell expansions that react with alloantigens.


Subject(s)
Antigens, Viral/immunology , Cytotoxicity, Immunologic/immunology , Graft Rejection/immunology , Herpesvirus 4, Human/immunology , Isoantigens/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/immunology , Antigens, Viral/metabolism , Cell Line, Transformed , Clone Cells , Cross-Priming/immunology , Cytotoxicity Tests, Immunologic/methods , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Graft Rejection/virology , HLA-B Antigens/immunology , HLA-B Antigens/metabolism , HLA-B44 Antigen , HLA-B8 Antigen/chemistry , HLA-B8 Antigen/immunology , HLA-B8 Antigen/metabolism , Half-Life , Humans , Immunodominant Epitopes/immunology , Immunodominant Epitopes/metabolism , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Protein Binding/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/virology
20.
Nat Immunol ; 6(2): 171-80, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15640805

ABSTRACT

The energetic bases of T cell recognition are unclear. Here, we studied the 'energetic landscape' of peptide-major histocompatibility complex (pMHC) recognition by an immunodominant alphabeta T cell receptor (TCR). We quantified and evaluated the effect of natural and systematic substitutions in the complementarity-determining region (CDR) loops on ligand binding in the context of the structural detail of each component of the immunodominant TCR-pMHC complex. The CDR1 and CDR2 loops contributed minimal energy through direct recognition of the antigen and instead had a chief function in stabilizing the ligated CDR3 loops. The underlying energetic basis for recognition lay in the CDR3 loops. Therefore the energetic burden of the CDR loops in the TCR-pMHC interaction is variable among TCRs, reflecting the inherent adaptability of the TCR in ligating different ligands.


Subject(s)
Complementarity Determining Regions/immunology , Complementarity Determining Regions/metabolism , Histocompatibility Antigens/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Base Sequence , Cells, Cultured , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Histocompatibility Antigens/chemistry , Humans , Kinetics , Ligands , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Peptides/chemistry , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Surface Plasmon Resonance , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...