Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 128(8): 3460-3474, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29851415

ABSTRACT

In type 1 diabetes, cytotoxic CD8+ T cells with specificity for ß cell autoantigens are found in the pancreatic islets, where they are implicated in the destruction of insulin-secreting ß cells. In contrast, the disease relevance of ß cell-reactive CD8+ T cells that are detectable in the circulation, and their relationship to ß cell function, are not known. Here, we tracked multiple, circulating ß cell-reactive CD8+ T cell subsets and measured ß cell function longitudinally for 2 years, starting immediately after diagnosis of type 1 diabetes. We found that change in ß cell-specific effector memory CD8+ T cells expressing CD57 was positively correlated with C-peptide change in subjects below 12 years of age. Autoreactive CD57+ effector memory CD8+ T cells bore the signature of enhanced effector function (higher expression of granzyme B, killer-specific protein of 37 kDa, and CD16, and reduced expression of CD28) compared with their CD57- counterparts, and network association modeling indicated that the dynamics of ß cell-reactive CD57+ effector memory CD8+ T cell subsets were strongly linked. Thus, coordinated changes in circulating ß cell-specific CD8+ T cells within the CD57+ effector memory subset calibrate to functional insulin reserve in type 1 diabetes, providing a tool for immune monitoring and a mechanism-based target for immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Immunologic Memory , Insulin-Secreting Cells/immunology , Adult , CD8-Positive T-Lymphocytes/pathology , Child , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Female , Humans , Insulin-Secreting Cells/pathology , Male
2.
Diabetologia ; 61(3): 658-670, 2018 03.
Article in English | MEDLINE | ID: mdl-29196783

ABSTRACT

AIMS/HYPOTHESIS: Validated biomarkers are needed to monitor the effects of immune intervention in individuals with type 1 diabetes. Despite their importance, few options exist for monitoring antigen-specific T cells. Previous reports described a combinatorial approach that enables the simultaneous detection and quantification of multiple islet-specific CD8+ T cell populations. Here, we set out to evaluate the performance of a combinatorial HLA-A2 multimer assay in a multi-centre setting. METHODS: The combinatorial HLA-A2 multimer assay was applied in five participating centres using centralised reagents and blinded replicate samples. In preliminary experiments, samples from healthy donors were analysed using recall antigen multimers. In subsequent experiments, samples from healthy donors and individuals with type 1 diabetes were analysed using beta cell antigen and recall antigen multimers. RESULTS: The combinatorial assay was successfully implemented in each participating centre, with CVs between replicate samples that indicated good reproducibility for viral epitopes (mean %CV = 33.8). For beta cell epitopes, the assay was very effective in a single-centre setting (mean %CV = 18.4), but showed sixfold greater variability across multi-centre replicates (mean %CV = 119). In general, beta cell antigen-specific CD8+ T cells were detected more commonly in individuals with type 1 diabetes than in healthy donors. Furthermore, CD8+ T cells recognising HLA-A2-restricted insulin and glutamate decarboxylase epitopes were found to occur at higher frequencies in individuals with type 1 diabetes than in healthy donors. CONCLUSIONS/INTERPRETATION: Our results suggest that, although combinatorial multimer assays are challenging, they can be implemented in multiple laboratories, providing relevant T cell frequency measurements. Assay reproducibility was notably higher in the single-centre setting, suggesting that biomarker analysis of clinical trial samples would be most successful when assays are performed in a single laboratory. Technical improvements, including further standardisation of cytometry platforms, will likely be necessary to reduce assay variability in the multi-centre setting.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , HLA-A2 Antigen/metabolism , Adult , Biomarkers/metabolism , Diabetes Mellitus, Type 1/metabolism , Female , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Young Adult
3.
Sci Rep ; 6: 35332, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27748447

ABSTRACT

CD8+ T-cells play a role in the pathogenesis of autoimmune diseases such as multiple sclerosis and type 1 diabetes. However, drugs that target the entire CD8+ T-cell population are not desirable because the associated lack of specificity can lead to unwanted consequences, most notably an enhanced susceptibility to infection. Here, we show that autoreactive CD8+ T-cells are highly dependent on CD8 for ligand-induced activation via the T-cell receptor (TCR). In contrast, pathogen-specific CD8+ T-cells are relatively CD8-independent. These generic differences relate to an intrinsic dichotomy that segregates self-derived and exogenous antigen-specific TCRs according to the monomeric interaction affinity with cognate peptide-major histocompatibility complex class I (pMHCI). As a consequence, "blocking" anti-CD8 antibodies can suppress autoreactive CD8+ T-cell activation in a relatively selective manner. These findings provide a rational basis for the development and in vivo assessment of novel therapeutic strategies that preferentially target disease-relevant autoimmune responses within the CD8+ T-cell compartment.


Subject(s)
Antibodies/immunology , CD8-Positive T-Lymphocytes/cytology , Histocompatibility Antigens Class I/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Autoimmune Diseases/immunology , CD8 Antigens/immunology , Cell Line , Epitopes/metabolism , Humans , Immunosuppression Therapy , Islets of Langerhans/metabolism , Ligands , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, Transgenic , Peptides/metabolism
5.
J Clin Invest ; 126(6): 2191-204, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183389

ABSTRACT

The cross-reactivity of T cells with pathogen- and self-derived peptides has been implicated as a pathway involved in the development of autoimmunity. However, the mechanisms that allow the clonal T cell antigen receptor (TCR) to functionally engage multiple peptide-major histocompatibility complexes (pMHC) are unclear. Here, we studied multiligand discrimination by a human, preproinsulin reactive, MHC class-I-restricted CD8+ T cell clone (1E6) that can recognize over 1 million different peptides. We generated high-resolution structures of the 1E6 TCR bound to 7 altered peptide ligands, including a pathogen-derived peptide that was an order of magnitude more potent than the natural self-peptide. Evaluation of these structures demonstrated that binding was stabilized through a conserved lock-and-key-like minimal binding footprint that enables 1E6 TCR to tolerate vast numbers of substitutions outside of this so-called hotspot. Highly potent antigens of the 1E6 TCR engaged with a strong antipathogen-like binding affinity; this engagement was governed though an energetic switch from an enthalpically to entropically driven interaction compared with the natural autoimmune ligand. Together, these data highlight how T cell cross-reactivity with pathogen-derived antigens might break self-tolerance to induce autoimmune disease.


Subject(s)
Insulin/immunology , Insulin/metabolism , Protein Precursors/immunology , Protein Precursors/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Amino Acid Sequence , Autoimmunity , Clone Cells , Cross Reactions , HLA-A Antigens/chemistry , HLA-A Antigens/metabolism , Humans , Insulin/genetics , Kinetics , Ligands , Models, Molecular , Oligopeptides/genetics , Oligopeptides/immunology , Oligopeptides/metabolism , Protein Binding , Protein Precursors/genetics , Receptors, Antigen, T-Cell/chemistry
6.
Diabetes ; 64(3): 916-925, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25249579

ABSTRACT

Autoreactive CD8 T cells play a central role in the destruction of pancreatic islet ß-cells that leads to type 1 diabetes, yet the key features of this immune-mediated process remain poorly defined. In this study, we combined high-definition polychromatic flow cytometry with ultrasensitive peptide-human leukocyte antigen class I tetramer staining to quantify and characterize ß-cell-specific CD8 T cell populations in patients with recent-onset type 1 diabetes and healthy control subjects. Remarkably, we found that ß-cell-specific CD8 T cell frequencies in peripheral blood were similar between subject groups. In contrast to healthy control subjects, however, patients with newly diagnosed type 1 diabetes displayed hallmarks of antigen-driven expansion uniquely within the ß-cell-specific CD8 T cell compartment. Molecular analysis of selected ß-cell-specific CD8 T cell populations further revealed highly skewed oligoclonal T cell receptor repertoires comprising exclusively private clonotypes. Collectively, these data identify novel and distinctive features of disease-relevant CD8 T cells that inform the immunopathogenesis of type 1 diabetes.


Subject(s)
Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Adult , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/physiology , Female , Flow Cytometry , Glutamate Decarboxylase/immunology , Humans , Insulin-Secreting Cells/cytology , Male , Receptor-Like Protein Tyrosine Phosphatases, Class 8/immunology
7.
J Immunol ; 192(2): 572-80, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24337380

ABSTRACT

CD8(+) T cells are critical in human type 1 diabetes and in the NOD mouse. In this study, we elucidated the natural history of islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-specific CD8(+) T cells in NOD diabetes using MHC-tetramer technology. IGRP206-214-specific T cells in the peripheral lymphoid tissue increased with age, and their numbers correlated with insulitis progression. IGRP206-214-specific T cells in the peripheral lymphoid tissue expressed markers of chronic Ag stimulation, and their numbers were stable after diagnosis of diabetes, consistent with their memory phenotype. IGRP206-214-specific T cells in NOD mice expand, acquire the phenotype of effector-memory T cells in the islets, and emigrate to the peripheral lymphoid tissue. Our observations suggest that enumeration of effector-memory T cells of multiple autoantigen specificities in the periphery of type 1 diabetic subjects could be a reliable reporter for progression of islet pathology.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Immunologic Memory/immunology , Islets of Langerhans/immunology , Animals , Autoantigens/immunology , Diabetes Mellitus, Type 1/pathology , Glucose-6-Phosphatase/immunology , Islets of Langerhans/pathology , Lymphocytes/immunology , Mice , Mice, Inbred NOD
8.
J Immunol ; 190(9): 4542-52, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23547116

ABSTRACT

A major goal of immunotherapy remains the control of pathogenic T cell responses that drive autoimmunity and allograft rejection. Adherent progenitor cells, including mesenchymal stromal cells (MSCs) and multipotent adult progenitor cells (MAPCs), represent attractive immunomodulatory cell therapy candidates currently active in clinical trials. MAPCs can be distinguished from MSCs on the basis of cellular phenotype, size, transcriptional profile, and expansion capacity. However, despite their ongoing evaluation in autoimmune and allogeneic solid organ transplantation settings, data supporting the immune regulatory potential of clinical-grade MAPCs are limited. In this study, we used allogeneic islet transplantation as a model indication to assess the ability of clinical-grade MAPCs to control T cell responses that drive immunopathology in human autoimmune disease and allograft rejection. MAPCs suppressed T cell proliferation and Th1 and Th17 cytokine production while increasing secretion of IL-10 and were able to suppress effector functions of bona fide autoreactive T cells from individuals with type 1 diabetes mellitus, including killing of human islets. Furthermore, MAPCs favored the proliferation of regulatory T cells during homeostatic expansion driven by γ-chain cytokines and exerted a durable, yet reversible, control of T cell function. MAPC suppression required licensing and proceeded via IDO-mediated tryptophan catabolism. Therefore, the common immune modulatory characteristics of clinical-grade MAPCs shown in this study suggest that they can be regarded as an alternative source of adult progenitor cells with similar clinical usefulness to MSCs. Taken collectively, these findings may guide the successful deployment of both MSCs and MAPCs for the amelioration of human autoimmunity and allograft rejection.


Subject(s)
Autoimmunity/immunology , Islets of Langerhans Transplantation/immunology , Lymphocyte Activation/immunology , Stem Cells/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adult , Adult Stem Cells/immunology , Cell Proliferation , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Graft Rejection/immunology , Humans , Immunomodulation/immunology , Interleukin-10/immunology , Male , Tryptophan/immunology , Young Adult
9.
Diabetes ; 62(1): 205-13, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22936177

ABSTRACT

The end-stage immunopathology of type 1 diabetes resulting in ß-cell destruction appears to be strongly dominated by cytotoxic CD8 T lymphocytes (CD8 T cells). However, the mechanism of cytotoxicity used by autoreactive CD8 T cells in the human setting remains unknown. Using type 1 diabetes patient-derived preproinsulin-specific CD8 T-cell clones recognizing either an HLA-A2 (A*0201) or HLA-A24 (A*2402)-restricted epitope (peptide of preproinsulin [PPI](15-24), ALWGPDPAAA; or PPI(3-11), LWMRLLPLL), we assessed the use of conventional mediators of cytotoxicity in the destruction of human ß-cells in vitro compared with virus-specific cytotoxic CD8 T-cell clones. We show that PPI-specific CD8 T-cell clones are mainly reliant upon cytotoxic degranulation for inducing ß-cell death. Furthermore, we find that in comparison with virus-specific CD8 T cells, there are differences in the killing potency of PPI-specific CD8 T cells that are not due to cell-intrinsic differences, but rather are mediated by differences in strength of signaling by peptide-HLA ligands. The study highlights the regulation of ß-cell killing as a potential point for therapeutic control, including the possibility of blocking autoreactive CD8 T-cell function without impacting upon general immune competence.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Degranulation , Cytotoxicity, Immunologic , Insulin-Secreting Cells/pathology , Insulin/immunology , Protein Precursors/immunology , Receptors, Antigen, T-Cell/physiology , Cell Line , Fas Ligand Protein/physiology , Humans , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology
10.
Blood ; 121(7): 1112-23, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23255554

ABSTRACT

αß-TCRs expressed at the CD8(+) T-cell surface interact with short peptide fragments (p) bound to MHC class I molecules (pMHCI). The TCR/pMHCI interaction is pivotal in all aspects of CD8(+) T-cell immunity. However, the rules that govern the outcome of TCR/pMHCI engagement are not entirely understood, and this is a major barrier to understanding the requirements for both effective immunity and vaccination. In the present study, we discovered an unexpected feature of the TCR/pMHCI interaction by showing that any given TCR exhibits an explicit preference for a single MHCI-peptide length. Agonists of nonpreferred length were extremely rare, suboptimal, and often entirely distinct in sequence. Structural analysis indicated that alterations in peptide length have a major impact on antigenic complexity, to which individual TCRs are unable to adapt. This novel finding demonstrates that the outcome of TCR/pMHCI engagement is determined by peptide length in addition to the sequence identity of the MHCI-bound peptide. Accordingly, the effective recognition of pMHCI Ag, which is a prerequisite for successful CD8(+) T-cell immunity and protective vaccination, can only be achieved by length-matched Ag-specific CD8(+) T-cell clonotypes.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Peptide Fragments/chemistry , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Amino Acid Sequence , Antigen Presentation , Antigens/chemistry , Antigens/genetics , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Clone Cells , Humans , Immunity, Cellular , Models, Molecular , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/immunology , Peptide Fragments/genetics , Peptide Library
11.
J Exp Med ; 209(5): 935-45, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22547651

ABSTRACT

Human skin immune homeostasis, and its regulation by specialized subsets of tissue-residing immune sentinels, is poorly understood. In this study, we identify an immunoregulatory tissue-resident dendritic cell (DC) in the dermis of human skin that is characterized by surface expression of CD141, CD14, and constitutive IL-10 secretion (CD141(+) DDCs). CD141(+) DDCs possess lymph node migratory capacity, induce T cell hyporesponsiveness, cross-present self-antigens to autoreactive T cells, and induce potent regulatory T cells that inhibit skin inflammation. Vitamin D(3) (VitD3) promotes certain phenotypic and functional properties of tissue-resident CD141(+) DDCs from human blood DCs. These CD141(+) DDC-like cells can be generated in vitro and, once transferred in vivo, have the capacity to inhibit xeno-graft versus host disease and tumor alloimmunity. These findings suggest that CD141(+) DDCs play an essential role in the maintenance of skin homeostasis and in the regulation of both systemic and tumor alloimmunity. Finally, VitD3-induced CD141(+) DDC-like cells have potential clinical use for their capacity to induce immune tolerance.


Subject(s)
Antigens, Surface/metabolism , Cell- and Tissue-Based Therapy/methods , Dermatitis/immunology , Graft vs Host Disease/prevention & control , Homeostasis/immunology , Interleukin-10/immunology , Langerhans Cells/immunology , T-Lymphocytes, Regulatory/immunology , Analysis of Variance , Animals , Cholecalciferol/pharmacology , Female , Humans , Indoles , Interleukin Receptor Common gamma Subunit/genetics , Langerhans Cells/drug effects , Langerhans Cells/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Thrombomodulin
12.
Diabetes ; 61(7): 1752-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22522618

ABSTRACT

Type 1 diabetes results from T cell-mediated ß-cell destruction. The HLA-A*24 class I gene confers significant risk of disease and early onset. We tested the hypothesis that HLA-A24 molecules on islet cells present preproinsulin (PPI) peptide epitopes to CD8 cytotoxic T cells (CTLs). Surrogate ß-cell lines secreting proinsulin and expressing HLA-A24 were generated and their peptide ligandome examined by mass spectrometry to discover naturally processed and HLA-A24-presented PPI epitopes. A novel PPI epitope was identified and used to generate HLA-A24 tetramers and examine the frequency of PPI-specific T cells in new-onset HLA-A*24(+) patients and control subjects. We identified a novel naturally processed and HLA-A24-presented PPI signal peptide epitope (PPI(3-11); LWMRLLPLL). HLA-A24 tetramer analysis reveals a significant expansion of PPI(3-11)-specific CD8 T cells in the blood of HLA-A*24(+) recent-onset patients compared with HLA-matched control subjects. Moreover, a patient-derived PPI(3-11)-specific CD8 T-cell clone shows a proinflammatory phenotype and kills surrogate ß-cells and human HLA-A*24(+) islet cells in vitro. These results indicate that the type 1 diabetes susceptibility molecule HLA-A24 presents a naturally processed PPI signal peptide epitope. PPI-specific, HLA-A24-restricted CD8 T cells are expanded in patients with recent-onset disease. Human islet cells process and present PPI(3-11), rendering themselves targets for CTL-mediated killing.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , HLA-A24 Antigen/immunology , Insulin-Secreting Cells/immunology , Insulin/immunology , Protein Precursors/immunology , Protein Sorting Signals , Adult , Autoantibodies/blood , Autoantibodies/immunology , Cell Death/immunology , Cell Line , Epitopes, T-Lymphocyte/immunology , Female , Glutamate Decarboxylase/immunology , Humans , Insulin/blood , Male , Middle Aged , Protein Precursors/blood , Young Adult
13.
Nat Immunol ; 13(3): 283-9, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22245737

ABSTRACT

The structural characteristics of the engagement of major histocompatibility complex (MHC) class II-restricted self antigens by autoreactive T cell antigen receptors (TCRs) is established, but how autoimmune TCRs interact with complexes of self peptide and MHC class I has been unclear. Here we examined how CD8(+) T cells kill human islet beta cells in type 1 diabetes via recognition of a human leukocyte antigen HLA-A*0201-restricted glucose-sensitive preproinsulin peptide by the autoreactive TCR 1E6. Rigid 'lock-and-key' binding underpinned the 1E6-HLA-A*0201-peptide interaction, whereby 1E6 docked similarly to most MHC class I-restricted TCRs. However, this interaction was extraordinarily weak because of limited contacts with MHC class I. TCR binding was highly peptide centric, dominated by two residues of the complementarity-determining region 3 (CDR3) loops that acted as an 'aromatic-cap' over the complex of peptide and MHC class I (pMHCI). Thus, highly focused peptide-centric interactions associated with suboptimal TCR-pMHCI binding affinities might lead to thymic escape and potential CD8(+) T cell-mediated autoreactivity.


Subject(s)
Apoptosis , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , CD8-Positive T-Lymphocytes/chemistry , Histocompatibility Antigens/immunology , Humans , Insulin-Secreting Cells/pathology , Models, Molecular , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology
14.
J Immunol ; 185(4): 2600-10, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20639478

ABSTRACT

Improving T cell Ags by altering MHC anchor residues is a common strategy used to enhance peptide vaccines, but there has been little assessment of how such modifications affect TCR binding and T cell recognition. In this study, we use surface plasmon resonance and peptide-MHC tetramer binding at the cell surface to demonstrate that changes in primary peptide anchor residues can substantially and unpredictably alter TCR binding. We also demonstrate that the ability of TCRs to differentiate between natural and anchor-modified heteroclitic peptides distinguishes T cells that exhibit a strong preference for either type of Ag. Furthermore, we show that anchor-modified heteroclitic peptides prime T cells with different TCRs compared with those primed with natural Ag. Thus, vaccination with heteroclitic peptides may elicit T cells that exhibit suboptimal recognition of the intended natural Ag and, consequently, impaired functional attributes in vivo. Heteroclitic peptide-based immune interventions therefore require careful evaluation to ensure efficacy in the clinic.


Subject(s)
HLA-A Antigens/immunology , Oligopeptides/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Flow Cytometry , HLA-A Antigens/genetics , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Mutation , Oligopeptides/genetics , Oligopeptides/metabolism , Peptide Library , Protein Binding/immunology , Receptors, Antigen, T-Cell/metabolism , Surface Plasmon Resonance , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
15.
Diabetes ; 59(6): 1451-60, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20299476

ABSTRACT

OBJECTIVE: Regulatory T-cells (Tregs) recognizing islet autoantigens are proposed as a key mechanism in the maintenance of self-tolerance and protection from type 1 diabetes. To date, however, detailed information on such cells in humans, and insight into their mechanisms of action, has been lacking. We previously reported that a subset of CD4 T-cells secreting high levels of the immunosuppressive cytokine interleukin-10 (IL-10) is significantly associated with late onset of type 1 diabetes and is constitutively present in a majority of nondiabetic individuals. Here, we test the hypothesis that these T-cells represent a naturally generated population of Tregs capable of suppressing proinflammatory T-cell responses. RESEARCH DESIGN AND METHODS: We isolated and cloned islet-specific IL-10-secreting CD4(+) T-cells from nondiabetic individuals after brief ex vivo exposure to islet autoantigens using cytokine capture technology and examined their phenotype and regulatory potential. RESULTS: Islet-specific IL-10(+) CD4 T-cells are potent suppressors of Th1 effector cells, operating through a linked suppression mechanism in which there is an absolute requirement for the cognate antigen of both the regulatory and effector T-cells to be presented by the same antigen-presenting cell (APC). The regulatory T-cells secrete perforin and granzymes, and suppression is associated with the specific killing of APCs presenting antigen to effector T-cells. CONCLUSIONS: This hitherto undescribed population of islet autoantigen-specific Tregs displays unique characteristics that offer exquisite specificity and control over the potential for pathological autoreactivity and may provide a suitable target with which to strengthen beta-cell-specific tolerance.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Interleukin-10/metabolism , Islets of Langerhans/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Survival , Cytokines/genetics , Cytokines/immunology , Humans , Immunosuppression Therapy , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/cytology
16.
Diabetes ; 58(1): 138-45, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18835928

ABSTRACT

OBJECTIVE: Immune-mediated destruction of beta-cells resulting in type 1 diabetes involves activation of proinflammatory, islet autoreactive T-cells, a process under the control of dendritic cells of the innate immune system. We tested the hypothesis that type 1 diabetes development is associated with disturbance of blood dendritic cell subsets that could enhance islet-specific autoimmunity. RESEARCH DESIGN AND METHODS: We examined blood dendritic cells (plasmacytoid and myeloid) in 40 patients with recent-onset diabetes (median duration 28 days) and matched control subjects. We also examined the relative ability of different dendritic cell subsets to process and present soluble or immune complexed islet cell autoantigen (the islet tyrosine phosphatase IA-2) to responder CD4 T-cells. RESULTS: The balance of blood dendritic cells was profoundly disturbed at diabetes diagnosis, with a significantly elevated proportion of plasmacytoid and reduction of myeloid cells compared with control subjects. Dendritic cell subset distribution was normal in long-standing disease and in patients with type 2 diabetes. Both dendritic cell subsets processed and presented soluble IA-2 to CD4 T-cells after short-term culture, but only plasmacytoid dendritic cells enhanced (by as much as 100%) autoantigen presentation in the presence of IA-2(+) autoantibody patient serum. CONCLUSIONS: The plasmacytoid subset of dendritic cells is overrepresented in the blood close to diabetes onset and shows a distinctive ability to capture islet autoantigenic immune complexes and enhance autoantigen-driven CD4 T-cell activation. This suggests a synergistic proinflammatory role for plasmacytoid dendritic cells and islet cell autoantibodies in type 1 diabetes.


Subject(s)
Autoantigens/immunology , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Antigen Presentation/immunology , Autoimmunity/immunology , Chemokines/metabolism , Child , Child, Preschool , Cytokines/metabolism , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/metabolism , Female , Flow Cytometry , Humans , Male , Middle Aged , T-Lymphocytes/metabolism , Young Adult
17.
J Immunol Methods ; 340(1): 11-24, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18929568

ABSTRACT

Flow cytometry with fluorochrome-conjugated peptide-major histocompatibility complex (pMHC) tetramers has transformed the study of antigen-specific T-cells by enabling their visualization, enumeration, phenotypic characterization and isolation from ex vivo samples. Here, we demonstrate that the reversible protein kinase inhibitor (PKI) dasatinib improves the staining intensity of human (CD8+ and CD4+) and murine T-cells without concomitant increases in background staining. Dasatinib enhances the capture of cognate pMHC tetramers from solution and produces higher intensity staining at lower pMHC concentrations. Furthermore, dasatinib reduces pMHC tetramer-induced cell death and substantially lowers the T-cell receptor (TCR)/pMHC interaction affinity threshold required for cell staining. Accordingly, dasatinib permits the identification of T-cells with very low affinity TCR/pMHC interactions, such as those that typically predominate in tumour-specific responses and autoimmune conditions that are not amenable to detection by current technology.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Flow Cytometry/methods , HLA-A2 Antigen/immunology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Adult , Animals , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Dasatinib , HLA-DR Antigens/immunology , Humans , Mice , Mice, Transgenic , Models, Immunological , Staining and Labeling/methods
18.
J Clin Invest ; 118(10): 3390-402, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18802479

ABSTRACT

The final pathway of beta cell destruction leading to insulin deficiency, hyperglycemia, and clinical type 1 diabetes is unknown. Here we show that circulating CTLs can kill beta cells via recognition of a glucose-regulated epitope. First, we identified 2 naturally processed epitopes from the human preproinsulin signal peptide by elution from HLA-A2 (specifically, the protein encoded by the A*0201 allele) molecules. Processing of these was unconventional, requiring neither the proteasome nor transporter associated with processing (TAP). However, both epitopes were major targets for circulating effector CD8+ T cells from HLA-A2+ patients with type 1 diabetes. Moreover, cloned preproinsulin signal peptide-specific CD8+ T cells killed human beta cells in vitro. Critically, at high glucose concentration, beta cell presentation of preproinsulin signal epitope increased, as did CTL killing. This study provides direct evidence that autoreactive CTLs are present in the circulation of patients with type 1 diabetes and that they can kill human beta cells. These results also identify a mechanism of self-antigen presentation that is under pathophysiological regulation and could expose insulin-producing beta cells to increasing cytotoxicity at the later stages of the development of clinical diabetes. Our findings suggest that autoreactive CTLs are important targets for immune-based interventions in type 1 diabetes and argue for early, aggressive insulin therapy to preserve remaining beta cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Epitopes, T-Lymphocyte/immunology , Glucose/metabolism , Insulin-Secreting Cells/pathology , Insulin/immunology , Protein Precursors/immunology , Adolescent , Adult , Epitopes, T-Lymphocyte/drug effects , Female , Glucose/pharmacology , Humans , Insulin/metabolism , K562 Cells , Male , Phenotype , Protein Precursors/metabolism , Protein Sorting Signals
19.
Clin Infect Dis ; 43(1): 1-7, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16758411

ABSTRACT

BACKGROUND: Medical countermeasures against biological warfare include the use of vaccines for anthrax and plague, which require repeated dosing and adjuvant to achieve adequate protection from threats such as inhalational anthrax and pneumonic plague. Despite the widespread use of these measures in preparation for recent military deployments, little is known about the cell-mediated immune response that is induced by these vaccines, in comparison with conventional vaccines, such as pertussis or tetanus-diphtheria vaccines. METHODS: To examine this question, we used cytokine enzyme-linked immunospot assays to measure interferon-gamma, interleukin (IL)-2, IL-4, and IL-13-producing cells in military service personnel vaccinated during the Gulf War of 1990-1991. RESULTS: Our data indicate that 12-15 years after vaccination against anthrax and plague, antigen-specific T cell recall responses are present in the circulation and are comparable in magnitude to those for tetanus-diphtheria toxoids. Recall responses to anthrax were an approximately equal mixture of type 1 T helper cell (interferon-gamma and IL-2) and type 2 T helper cell (predominantly IL-13) responses, whereas plague cellular immunity was more polarized toward type 1 T helper cell responses. Responder cell frequency and type were similar to that against conventional tetanus-diphtheria (mixed type 1 and type 2 T helper cells) vaccine. When veterans were divided according to whether or not they reported multisymptom illness, there was no difference in the frequency or type of cellular response, although the number of cases in each group was small, and these data should be interpreted as preliminary. CONCLUSIONS: This study shows that, despite any putative limitations of vaccines for anthrax and plague in terms of achieving protective host immunity, long-lasting cell-mediated responses are generated with these agents.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/immunology , Biological Warfare , Plague Vaccine/immunology , Plague/immunology , T-Lymphocytes/immunology , Cytokines , Humans , Immunity, Cellular , Th1 Cells , Th2 Cells
20.
Philos Trans R Soc Lond B Biol Sci ; 361(1468): 681-7, 2006 Apr 29.
Article in English | MEDLINE | ID: mdl-16687270

ABSTRACT

One candidate cause of Gulf War illness is vaccination against infectious diseases including medical counter-measures against biological weapons. One influential theory has suggested that such mass-vaccination caused a shift in immune response to a Type 2 cytokine pattern (Th2), which it was suggested was accompanied by a chronic fatigue syndrome-like illness. This article critically appraises this theory. We start by examining epidemiological evidence, which indicates that single vaccines are unlikely to be a substantial cause of Gulf War illness, but that there was a modest relationship with multiple vaccines, which was strongest in those vaccinated while deployed to the Gulf. These relationships may be affected by recall bias. We conclude by examining the results of immunological studies carried out in veterans or in a relevant setting in vitro. The balance of evidence from immunological studies on veterans returning from the War, including those developing multi-symptom illness, is that the immune response has not become polarized towards Th2. In summary, the epidemiological evidence for a multiple vaccine effect on Gulf War-related illness remains a potentially important aetiological lead, but mechanistic studies available at this stage do not identify any immunological basis for it.


Subject(s)
Immune System Diseases/etiology , Immune System Diseases/immunology , Persian Gulf Syndrome/etiology , Persian Gulf Syndrome/immunology , Vaccination/adverse effects , Cytokines/metabolism , Gulf War , History, 20th Century , Humans , Immune System Diseases/physiopathology , Persian Gulf Syndrome/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...