Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 14(7): 699-705, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23666294

ABSTRACT

Activating and inhibitory receptors on natural killer (NK) cells have a crucial role in innate immunity, although the basis of the engagement of activating NK cell receptors is unclear. The activating receptor Ly49H confers resistance to infection with murine cytomegalovirus by binding to the 'immunoevasin' m157. We found that m157 bound to the helical stalk of Ly49H, whereby two m157 monomers engaged the Ly49H dimer. The helical stalks of Ly49H lay centrally across the m157 platform, whereas its lectin domain was not required for recognition. Instead, m157 targeted an 'aromatic peg motif' present in stalks of both activating and inhibitory receptors of the Ly49 family, and substitution of this motif abrogated binding. Furthermore, ligation of m157 to Ly49H or Ly49C resulted in intracellular signaling. Accordingly, m157 has evolved to 'tackle the legs' of a family of NK cell receptors.


Subject(s)
Herpesviridae Infections/immunology , Histocompatibility Antigens Class I/immunology , Immunity, Innate/immunology , Killer Cells, Natural/immunology , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/immunology , Amino Acid Motifs/immunology , Amino Acid Sequence , Animals , Crystallography, X-Ray , Female , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Signal Transduction/immunology , Specific Pathogen-Free Organisms , Surface Plasmon Resonance
2.
Nat Immunol ; 13(12): 1171-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23142773

ABSTRACT

The development and function of natural killer (NK) cells is regulated by the interaction of inhibitory receptors of the Ly49 family with distinct peptide-laden major histocompatibility complex (MHC) class I molecules, although whether the Ly49 family is able bind to other MHC class I-like molecules is unclear. Here we found that the prototypic inhibitory receptor Ly49A bound the highly conserved nonclassical MHC class I molecule H2-M3 with an affinity similar to its affinity for H-2D(d). The specific recognition of H2-M3 by Ly49A regulated the 'licensing' of NK cells and mediated 'missing-self' recognition of H2-M3-deficient bone marrow. Host peptide-H2-M3 was required for optimal NK cell activity against experimental metastases and carcinogenesis. Thus, nonclassical MHC class I molecules can act as cognate ligands for Ly49 molecules. Our results provide insight into the various mechanisms that lead to NK cell tolerance.


Subject(s)
Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Animals , Histocompatibility Antigens Class I/genetics , Immune Tolerance , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Am J Transplant ; 2023 Nov 18.
Article in English | MEDLINE | ID: mdl-37981213

ABSTRACT

Outcomes after lung transplantation (LTx) remain poor, despite advances in sequencing technology and development of algorithms defining immunologic compatibility. Presently, there is no consensus regarding the best approach to define human leukocyte antigen (HLA) compatibility in LTx. In this study, we compared 5 different HLA compatibility tools in a high-resolution HLA-typed, clinically characterized cohort, to determine which approach predicts outcomes after LTx. In this retrospective single-center study, 277 donor-recipient transplant pairs were HLA-typed using next generation sequencing. HLA compatibility was defined using HLAMatchmaker, HLA epitope mismatch algorithm (HLA-EMMA), predicted indirectly recognizable HLA epitopes (PIRCHE), electrostatic mismatch score (EMS), and amino acid mismatches (AAMMs). Associations with HLA mismatching and survival, chronic lung allograft dysfunction (CLAD), and anti-HLA donor-specific antibody (DSA) were calculated using adjusted Cox proportional modeling. Lower HLA class II mismatching was associated with improved survival as defined by HLAMatchmaker (P < .01), HLA-EMMA (P < .05), PIRCHE (P < .05), EMS (P < .001), and AAMM (P < .01). All approaches demonstrated that HLA-DRB1345 matching was associated with freedom from restrictive allograft syndrome and HLA-DQ matching with reduced DSA development. Reducing the level of HLA mismatching, in T cell or B cell epitopes, electrostatic differences, or amino acid, can improve outcomes after LTx and potentially guide immunosuppression strategies.

4.
Nat Immunol ; 12(7): 616-23, 2011 Jun 12.
Article in English | MEDLINE | ID: mdl-21666690

ABSTRACT

Type I natural killer T cells (NKT cells) are characterized by an invariant variable region 14-joining region 18 (V(α)14-J(α)18) T cell antigen receptor (TCR) α-chain and recognition of the glycolipid α-galactosylceramide (α-GalCer) restricted to the antigen-presenting molecule CD1d. Here we describe a population of α-GalCer-reactive NKT cells that expressed a canonical V(α)10-J(α)50 TCR α-chain, which showed a preference for α-glucosylceramide (α-GlcCer) and bacterial α-glucuronic acid-containing glycolipid antigens. Structurally, despite very limited TCRα sequence identity, the V(α)10 TCR-CD1d-α-GlcCer complex had a docking mode similar to that of type I TCR-CD1d-α-GalCer complexes, although differences at the antigen-binding interface accounted for the altered antigen specificity. Our findings provide new insight into the structural basis and evolution of glycolipid antigen recognition and have notable implications for the scope and immunological role of glycolipid-specific T cell responses.


Subject(s)
Galactosylceramides/immunology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Adjuvants, Immunologic/pharmacology , Amino Acid Sequence , Animals , Antigens, Bacterial/immunology , Antigens, CD1d/immunology , Cell Line , Galactosylceramides/pharmacology , Glucuronates/immunology , Humans , Mice , Mice, Mutant Strains , Molecular Sequence Data , Receptors, Antigen, T-Cell, alpha-beta/genetics
5.
Transpl Infect Dis ; 23(3): e13550, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33351991

ABSTRACT

BACKGROUND: Optimizing immunosuppression in lung transplant recipients (LTR) is crucially important in minimizing the risk of infection and rejection. Quantiferon®-Monitor (QFM) is a candidate immune function biomarker which has not yet been rigorously evaluated in the lung transplant setting. The aim of this prospective cohort study was to explore relationships between QFM results, immunosuppression, and infection/rejection in LTR. METHODS: QFM, which measures interferon-γ after stimulation with innate and adaptive immune antigens, was tested before and at 2, 6, 12, 24 and 52 weeks post-transplant. Immunosuppression relationships were assessed with linear mixed effects models. Clinical outcomes were analyzed based on the preceding QFM result. RESULTS: Eighty LTR were included. Median pre-transplant QFM levels were 171 IU/mL (IQR 45-461), decreasing to 3 IU/mL (IQR 1-8) at 2 weeks post-transplant then progressively recovering toward baseline with time from transplant. Prednisolone was strongly inversely associated with QFM level (0.1 mg/kg dose increase correlating with 88 IU/mL QFM decrease, 95% CI 61-114, P < .001). Patients with QFM values <10 and <60 IU/mL were more likely to develop a serious opportunistic infection between 3 and 6 months (HR 6.38, 95% CI 1.37-29.66, P = .02) and 6-12 months (HR 3.25, 95% CI 1.11-9.49, P = .03) post-transplant, respectively. CONCLUSIONS: QFM values declined significantly post-transplant, with patients recovering at different rates. Prednisolone dose significantly impacted QFM results. Low levels were associated with infection beyond 3 months post-transplant, suggesting that QFM may be able to identify overly immunosuppressed patients who could be targeted for dose reduction. Larger prospective studies are needed to further evaluate this promising assay.


Subject(s)
Immunosuppression Therapy , Transplant Recipients , Biomarkers , Graft Rejection , Humans , Lung , Lung Transplantation , Prospective Studies
6.
Proc Natl Acad Sci U S A ; 115(46): 11808-11813, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30381460

ABSTRACT

Natural killer (NK) cells play a critical role in controlling malignancies. Susceptibility or resistance to lung cancer, for example, specifically depends on NK cell function. Nevertheless, intrinsic factors that control NK cell-mediated clearance of lung cancer are unknown. Here we report that NK cells exposed to exogenous major histocompatibility class I (MHCI) provide a significant immunologic barrier to the growth and progression of malignancy. Clearance of lung cancer is facilitated by up-regulation of NKG2D, NKp46, and other activating receptors upon exposure to environmental MHCI. Surface expression of the inhibitory receptor Ly49C/I, on the other hand, is down-regulated upon exposure to tumor-bearing tissue. We thus demonstrate that NK cells exhibit dynamic plasticity in surface expression of both activating and inhibitory receptors based on the environmental context. Our data suggest that altering the activation state of NK cells may contribute to immunologic control of lung and possibly other cancers.


Subject(s)
Antigens, Ly/immunology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , NK Cell Lectin-Like Receptor Subfamily A/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Natural Cytotoxicity Triggering Receptor 1/immunology , Receptors, Immunologic/immunology , Receptors, Natural Killer Cell/metabolism , Animals , Cytotoxicity, Immunologic , Down-Regulation , Histocompatibility Antigens Class I/metabolism , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Up-Regulation
7.
Am J Respir Cell Mol Biol ; 62(4): 430-439, 2020 04.
Article in English | MEDLINE | ID: mdl-31697586

ABSTRACT

We investigated the contribution of human leukocyte antigen A2 (HLA-A2) and HLA-E-restricted CD8+ T cells in patients with Mycobacterium tuberculosis and human immunodeficiency virus 1 (HIV-1) coinfection. HIV-1 downregulates HLA-A, -B, and -C molecules in infected cells, thus influencing recognition by HLA class I-restricted CD8+ T cells but not by HLA-E-restricted CD8+ T cells, owing to the inability of the virus to downmodulate their expression. Therefore, antigen-specific HLA-E-restricted CD8+ T cells could play a protective role in Mycobacterium tuberculosis and HIV-1 coinfection. HLA-E- and HLA-A2-restricted Mycobacterium tuberculosis-specific CD8+ T cells were tested in vitro for cytotoxic and microbicidal activities, and their frequencies and phenotypes were evaluated ex vivo in patients with active tuberculosis and concomitant HIV-1 infection. HIV-1 and Mycobacterium tuberculosis coinfection caused downmodulation of HLA-A2 expression in human monocyte-derived macrophages associated with resistance to lysis by HLA-A2-restricted CD8+ T cells and failure to restrict the growth of intracellular Mycobacterium tuberculosis. Conversely, HLA-E surface expression and HLA-E-restricted cytolytic and microbicidal CD8 responses were not affected. HLA-E-restricted and Mycobacterium tuberculosis-specific CD8+ T cells were expanded in the circulation of patients with Mycobacterium tuberculosis/HIV-1 coinfection, as measured by tetramer staining, but displayed a terminally differentiated and exhausted phenotype that was rescued in vitro by anti-PD-1 (programmed cell death protein 1) monoclonal antibody. Together, these results indicate that HLA-E-restricted and Mycobacterium tuberculosis-specific CD8+ T cells in patients with Mycobacterium tuberculosis/HIV-1 coinfection have an exhausted phenotype and fail to expand in vitro in response to antigen stimulation, which can be restored by blocking the PD-1 pathway using the specific monoclonal antibody nivolumab.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Coinfection/immunology , HIV Infections/immunology , HIV-1/immunology , HLA-A2 Antigen/immunology , Histocompatibility Antigens Class I/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Adult , Antigens, Bacterial/immunology , Down-Regulation/immunology , Female , Humans , Lymphocyte Activation/immunology , Lymphocyte Count/methods , Male , Middle Aged , HLA-E Antigens
8.
Immunity ; 34(3): 327-39, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21376639

ABSTRACT

Natural killer T (NKT) cells respond to a variety of CD1d-restricted antigens (Ags), although the basis for Ag discrimination by the NKT cell receptor (TCR) is unclear. Here we have described NKT TCR fine specificity against several closely related Ags, termed altered glycolipid ligands (AGLs), which differentially stimulate NKT cells. The structures of five ternary complexes all revealed similar docking. Acyl chain modifications did not affect the interaction, but reduced NKT cell proliferation, indicating an affect on Ag processing or presentation. Conversely, truncation of the phytosphingosine chain caused an induced fit mode of TCR binding that affected TCR affinity. Modifications in the glycosyl head group had a direct impact on the TCR interaction and associated cellular response, with ligand potency reflecting the t(1/2) life of the interaction. Accordingly, we have provided a molecular basis for understanding how modifications in AGLs can result in striking alterations in the cellular response of NKT cells.


Subject(s)
Antigens, CD1d/immunology , Epitopes , Natural Killer T-Cells/immunology , Animals , Carbohydrate Sequence , Cell Line , Cell Proliferation , Glycolipids/immunology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Natural Killer T-Cells/cytology , Receptors, Natural Killer Cell/immunology
9.
Am J Transplant ; 19(5): 1577-1581, 2019 05.
Article in English | MEDLINE | ID: mdl-30653828

ABSTRACT

Antibody-mediated rejection, whereby transplant recipient B cells and/or plasma cells produce alloreactive anti-human leukocyte antigen (HLA) antibodies, negatively influences transplant outcomes and is a major contributor to graft loss. An early humoral immune response is suggested by the production of anti-HLA donor-specific antibodies (DSA) that can be measured using solid phase assays. We report the early posttransplant coexistence of a shared anti-HLA antibody profile in 5 solid organ transplant recipients who received organs from the same donor. Retrospective analysis of the donor's serum confirmed the presence of the same anti-HLA profile, suggesting the transfer of donor-derived anti-HLA antibodies, or the cells that produce them, to multiple solid organ transplant recipients. The time frame and extent of transfer suggest a novel variant of the passenger lymphocyte syndrome. These findings have important implications for the consideration of all posttransplant antibody measurements, particularly the interpretation of non-DSAs in the sera of transplant recipients.


Subject(s)
HLA Antigens/immunology , Immunity, Humoral/immunology , Isoantibodies/immunology , Lung Transplantation/methods , Lymphocytes/immunology , Postoperative Complications/immunology , Tissue Donors/supply & distribution , Adult , Female , Humans , Male , Middle Aged , Organ Transplantation , Prognosis , Retrospective Studies , Syndrome
10.
Immunol Cell Biol ; 97(3): 326-339, 2019 03.
Article in English | MEDLINE | ID: mdl-30537346

ABSTRACT

Class Ib major histocompatibility complex (MHC) is an extended family of molecules, which demonstrate tissue-specific expression and presentation of monomorphic antigens. These characteristics tend to imbue class Ib MHC with unique functions. H2-Q10 is potentially one such molecule that is overexpressed in the liver but its immunological function is not known. We have previously shown that H2-Q10 is a ligand for the natural killer cell receptor Ly49C and now, using H2-Q10-deficient mice, we demonstrate that H2-Q10 can also stabilize the expression of Qa-1b. In the absence of H2-Q10, the development and maturation of conventional hepatic natural killer cells is disrupted. We also provide evidence that H2-Q10 is a new high affinity ligand for CD8αα and controls the development of liver-resident CD8αα γδT cells. These data demonstrate that H2-Q10 has multiple roles in the development of immune subsets and identify an overlap of recognition within the class Ib MHC that is likely to be relevant to the regulation of immunity.


Subject(s)
H-2 Antigens/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Immunologic/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Biomarkers , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , H-2 Antigens/genetics , H-2 Antigens/metabolism , Immunomodulation/genetics , Immunophenotyping , Killer Cells, Natural/cytology , Ligands , Liver/immunology , Liver/metabolism , Mice , Protein Binding , T-Lymphocyte Subsets/cytology
11.
Immunol Rev ; 267(1): 148-66, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26284476

ABSTRACT

The surveillance of target cells by natural killer (NK) cells utilizes an ensemble of inhibitory and activating receptors, many of which interact with major histocompatibility complex (MHC) class I molecules. NK cell recognition of MHC class I proteins is important developmentally for the acquisition of full NK cell effector capacity and during target cell recognition, where the engagement of inhibitory receptors and MHC class I molecules attenuates NK cell activation. Human NK cells have evolved two broad strategies for recognition of human leukocyte antigen (HLA) class I molecules: (i) direct recognition of polymorphic classical HLA class I proteins by diverse receptor families such as the killer cell immunoglobulin-like receptors (KIRs), and (ii) indirect recognition of conserved sets of HLA class I-derived peptides displayed on the non-classical HLA-E for recognition by CD94-NKG2 receptors. In this review, we assess the structural basis for the interaction between these NK receptors and their HLA class I ligands and, using the suite of published KIR and CD94-NKG2 ternary complexes, highlight the features that allow NK cells to orchestrate the recognition of a range of different HLA class I proteins.


Subject(s)
HLA Antigens/immunology , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Receptors, Natural Killer Cell/immunology , Animals , HLA Antigens/chemistry , HLA Antigens/metabolism , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/metabolism , Ligands , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Receptors, Natural Killer Cell/chemistry , Receptors, Natural Killer Cell/metabolism
12.
J Biol Chem ; 292(51): 21149-21158, 2017 12 22.
Article in English | MEDLINE | ID: mdl-28972140

ABSTRACT

αß T cell receptors (TCRs) interact with peptides bound to the polymorphic major histocompatibility complex class Ia (MHC-Ia) and class II (MHC-II) molecules as well as the essentially monomorphic MHC class Ib (MHC-Ib) molecules. Although there is a large amount of information on how TCRs engage with MHC-Ia and MHC-II, our understanding of TCR/MHC-Ib interactions is very limited. Infection with cytomegalovirus (CMV) can elicit a CD8+ T cell response restricted by the human MHC-Ib molecule human leukocyte antigen (HLA)-E and specific for an epitope from UL40 (VMAPRTLIL), which is characterized by biased TRBV14 gene usage. Here we describe an HLA-E-restricted CD8+ T cell able to recognize an allotypic variant of the UL40 peptide with a modification at position 8 (P8) of the peptide (VMAPRTLVL) that uses the TRBV9 gene segment. We report the structures of a TRBV9+ TCR in complex with the HLA-E molecule presenting the two peptides. Our data revealed that the TRBV9+ TCR adopts a different docking mode and molecular footprint atop HLA-E when compared with the TRBV14+ TCR-HLA-E ternary complex. Additionally, despite their differing V gene segment usage and different docking mechanisms, mutational analyses showed that the TCRs shared a conserved energetic footprint on the HLA-E molecule, focused around the peptide-binding groove. Hence, we provide new insights into how monomorphic MHC molecules interact with T cells.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Energy Metabolism , Histocompatibility Antigens Class I/metabolism , Models, Molecular , Receptors, Antigen, T-Cell, alpha-beta/agonists , Amino Acid Sequence , Binding Sites , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Clone Cells , Conserved Sequence , Crystallography, X-Ray , Epitope Mapping , Epitopes, T-Lymphocyte , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Humans , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , HLA-E Antigens
13.
Immunity ; 30(2): 193-203, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19167249

ABSTRACT

During selection of the T cell repertoire, the immune system navigates the subtle distinction between self-restriction and self-tolerance, yet how this is achieved is unclear. Here we describe how self-tolerance toward a trans-HLA (human leukocyte antigen) allotype shapes T cell receptor (TCR) recognition of an Epstein-Barr virus (EBV) determinant (FLRGRAYGL). The recognition of HLA-B8-FLRGRAYGL by two archetypal TCRs was compared. One was a publicly selected TCR, LC13, that is alloreactive with HLA-B44; the other, CF34, lacks HLA-B44 reactivity because it arises when HLA-B44 is coinherited in trans with HLA-B8. Whereas the alloreactive LC13 TCR docked at the C terminus of HLA-B8-FLRGRAYGL, the CF34 TCR docked at the N terminus of HLA-B8-FLRGRAYGL, which coincided with a polymorphic region between HLA-B8 and HLA-B44. The markedly contrasting footprints of the LC13 and CF34 TCRs provided a portrait of how self-tolerance shapes the specificity of TCRs selected into the immune repertoire.


Subject(s)
Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Self Tolerance/immunology , Amino Acid Sequence , Antigens, Viral/chemistry , Antigens, Viral/immunology , Crystallography, X-Ray , HLA-B8 Antigen/chemistry , HLA-B8 Antigen/immunology , Herpesvirus 4, Human/chemistry , Herpesvirus 4, Human/immunology , Humans , Models, Molecular , Peptides/chemistry , Peptides/immunology , Protein Structure, Quaternary , Structural Homology, Protein , Surface Plasmon Resonance
14.
Immunity ; 31(1): 47-59, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19592275

ABSTRACT

The semi-invariant natural killer T cell receptor (NKT TCR) recognizes CD1d-lipid antigens. Although the TCR alpha chain is typically invariant, the beta chain expression is more diverse, where three V beta chains are commonly expressed in mice. We report the structures of V alpha 14-V beta 8.2 and V alpha 14-V beta 7 NKT TCRs in complex with CD1d-alpha-galactosylceramide (alpha-GalCer) and the 2.5 A structure of the human NKT TCR-CD1d-alpha-GalCer complex. Both V beta 8.2 and V beta 7 NKT TCRs and the human NKT TCR ligated CD1d-alpha-GalCer in a similar manner, highlighting the evolutionarily conserved interaction. However, differences within the V beta domains of the V beta 8.2 and V beta 7 NKT TCR-CD1d complexes resulted in altered TCR beta-CD1d-mediated contacts and modulated recognition mediated by the invariant alpha chain. Mutagenesis studies revealed the differing contributions of V beta 8.2 and V beta 7 residues within the CDR2 beta loop in mediating contacts with CD1d. Collectively we provide a structural basis for the differential NKT TCR V beta usage in NKT cells.


Subject(s)
Antigens, CD1d/immunology , Galactosylceramides/immunology , Natural Killer T-Cells/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , Antigens, CD1d/chemistry , Cloning, Molecular , Crystallization , Galactosylceramides/chemistry , Humans , Mice , Mutagenesis , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Conformation , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics
16.
J Biol Chem ; 291(36): 18740-52, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27385590

ABSTRACT

Murine natural killer (NK) cells are regulated by the interaction of Ly49 receptors with major histocompatibility complex class I molecules (MHC-I). Although the ligands for inhibitory Ly49 were considered to be restricted to classical MHC (MHC-Ia), we have shown that the non-classical MHC molecule (MHC-Ib) H2-M3 was a ligand for the inhibitory Ly49A. Here we establish that another MHC-Ib, H2-Q10, is a bona fide ligand for the inhibitory Ly49C receptor. H2-Q10 bound to Ly49C with a marginally lower affinity (∼5 µm) than that observed between Ly49C and MHC-Ia (H-2K(b)/H-2D(d), both ∼1 µm), and this recognition could be prevented by cis interactions with H-2K in situ To understand the molecular details underpinning Ly49·MHC-Ib recognition, we determined the crystal structures of H2-Q10 and Ly49C bound H2-Q10. Unliganded H2-Q10 adopted a classical MHC-I fold and possessed a peptide-binding groove that exhibited features similar to those found in MHC-Ia, explaining the diverse peptide binding repertoire of H2-Q10. Ly49C bound to H2-Q10 underneath the peptide binding platform to a region that encompassed residues from the α1, α2, and α3 domains, as well as the associated ß2-microglobulin subunit. This docking mode was conserved with that previously observed for Ly49C·H-2K(b) Indeed, structure-guided mutation of Ly49C indicated that Ly49C·H2-Q10 and Ly49C·H-2K(b) possess similar energetic footprints focused around residues located within the Ly49C ß4-stand and L5 loop, which contact the underside of the peptide-binding platform floor. Our data provide a structural basis for Ly49·MHC-Ib recognition and demonstrate that MHC-Ib represent an extended family of ligands for Ly49 molecules.


Subject(s)
Histocompatibility Antigen H-2D/chemistry , Killer Cells, Natural/chemistry , NK Cell Lectin-Like Receptor Subfamily A/chemistry , Animals , Crystallography, X-Ray , H-2 Antigens/chemistry , H-2 Antigens/genetics , H-2 Antigens/immunology , Histocompatibility Antigen H-2D/genetics , Histocompatibility Antigen H-2D/immunology , Killer Cells, Natural/immunology , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/immunology , Protein Domains , Protein Structure, Quaternary
17.
Amino Acids ; 49(10): 1691-1704, 2017 10.
Article in English | MEDLINE | ID: mdl-28718065

ABSTRACT

The lipopeptide, R4Pam2Cys, associates electrostatically with soluble protein antigens and significantly enhances their ability to induce protective humoral and cell-mediated responses. We demonstrate that antibody titers elicited by the antigen ovalbumin (OVA) associated with R4Pam2Cys are higher than those elicited by OVA in the presence of alum and comparable to those elicited by OVA formulated with complete Freund's adjuvant (CFA). The hierarchy of anti-OVA antibody avidities was CFA > R4Pam2Cys = alum. Each of the three adjuvants facilitated IgG class-switching with significantly more IgG1 elicited by OVA when formulated with R4Pam2Cys. The effects of substituting naturally occurring L-stereoisomers of the cationic residues within R4Pam2Cys with D-stereoisomers revealed that substitution did not affect the ability of R4Pam2Cys to stimulate dendritic cell maturation or its ability to elicit antibody production when used as an adjuvant. Minor detrimental effects were, however, observed in the ensuing CD8+ T cell responses suggesting that the use of D-amino acids affects antigen processing and presentation pathways involved in generation of cell-mediated immunity at least when facilitated through TLR2. Both D- and L-forms were found to be resistant to digestion by trypsin, indicating resistance of the branched structure to protease activity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lipopeptides , Ovalbumin , Toll-Like Receptor 2/agonists , Animals , Female , Immunoglobulin G/immunology , Lipopeptides/chemical synthesis , Lipopeptides/chemistry , Lipopeptides/immunology , Lipopeptides/pharmacology , Mice , Mice, Inbred BALB C , Ovalbumin/chemistry , Ovalbumin/immunology , Ovalbumin/pharmacology , Static Electricity , Structure-Activity Relationship , Toll-Like Receptor 2/immunology
18.
Eur J Immunol ; 45(4): 1069-81, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25631937

ABSTRACT

CD8 T cells contribute to protective immunity against Mycobacterium tuberculosis. In humans, M. tuberculosis reactive CD8 T cells typically recognize peptides associated to classical MHC class Ia molecules, but little information is available on CD8 T cells recognizing M. tuberculosis Ags presented by nonclassical MHC class Ib molecules. We show here that CD8 T cells from tuberculosis (TB) patients recognize HLA-E-binding M. tuberculosis peptides in a CD3/TCR αß mediated and CD8-dependent manner, and represent an additional type of effector cells playing a role in immune response to M. tuberculosis during active infection. HLA-E-restricted recognition of M. tuberculosis peptides is detectable by a significant enhanced ex vivo frequency of tetramer-specific circulating CD8 T cells during active TB. These CD8 T cells produce type 2 cytokines upon antigenic in vitro stimulation, help B cells for Ab production, and mediate limited TRAIL-dependent cytolytic and microbicidal activity toward M. tuberculosis infected target cells. Our results, together with the finding that HLA-E/M. tuberculosis peptide specific CD8 T cells are detected in TB patients with or without HIV coinfection, suggest that this is a new human T-cell population that participates in immune response in TB.


Subject(s)
Cytokines/biosynthesis , Histocompatibility Antigens Class I/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes, Cytotoxic/immunology , Tuberculosis/immunology , Adult , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Cells, Cultured , Coinfection/immunology , Coinfection/microbiology , Coinfection/virology , Cytokines/immunology , Epitopes, T-Lymphocyte/immunology , Female , HIV Infections/immunology , HIV Infections/virology , Humans , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily D/immunology , Protein Binding/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Tuberculosis/microbiology , HLA-E Antigens
19.
Eur J Immunol ; 45(3): 854-64, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25487143

ABSTRACT

Targeting antigens to dendritic cell (DC) surface receptors using antibodies has been successfully used to generate strong immune responses and is currently in clinical trials for cancer immunotherapy. Whilst cancer immunotherapy focuses on the induction of CD8(+) T-cell responses, many successful vaccines to pathogens or their toxins utilize humoral immunity as the primary effector mechanism. Universally, these approaches have used adjuvants or pathogen material that augment humoral responses. However, adjuvants are associated with safety issues. One approach, successfully used in the mouse, to generate strong humoral responses in the absence of adjuvant is to target antigen to Clec9A, also known as DNGR-1, a receptor on CD8α(+) DCs. Here, we address two issues relating to clinical application. First, we address the issue of variable adjuvant-dependence for different antibodies targeting mouse Clec9A. We show that multiple sites on Clec9A can be successfully targeted, but that strong in vivo binding and provision of suitable helper T cell determinants was essential for efficacy. Second, we show that induction of humoral immunity to CLEC9A-targeted antigens is extremely effective in nonhuman primates, in an adjuvant-free setting. Our findings support extending this vaccination approach to humans and offer important insights into targeting design.


Subject(s)
Antibodies/pharmacology , Dendritic Cells/immunology , Immunity, Humoral/drug effects , Lectins, C-Type/immunology , Adjuvants, Immunologic , Animals , Binding Sites, Antibody , CD8 Antigens/immunology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Dendritic Cells/pathology , Humans , Macaca nemestrina , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/immunology
SELECTION OF CITATIONS
SEARCH DETAIL