Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Sci Adv ; 10(10): eadl1122, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38446892

ABSTRACT

Coxsackievirus B (CVB) infection of pancreatic ß cells is associated with ß cell autoimmunity and type 1 diabetes. We investigated how CVB affects human ß cells and anti-CVB T cell responses. ß cells were efficiently infected by CVB in vitro, down-regulated human leukocyte antigen (HLA) class I, and presented few, selected HLA-bound viral peptides. Circulating CD8+ T cells from CVB-seropositive individuals recognized a fraction of these peptides; only another subfraction was targeted by effector/memory T cells that expressed exhaustion marker PD-1. T cells recognizing a CVB epitope cross-reacted with ß cell antigen GAD. Infected ß cells, which formed filopodia to propagate infection, were more efficiently killed by CVB than by CVB-reactive T cells. Our in vitro and ex vivo data highlight limited CD8+ T cell responses to CVB, supporting the rationale for CVB vaccination trials for type 1 diabetes prevention. CD8+ T cells recognizing structural and nonstructural CVB epitopes provide biomarkers to differentially follow response to infection and vaccination.


Subject(s)
Coxsackievirus Infections , Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Humans , CD8-Positive T-Lymphocytes , Antibodies , Epitopes , Peptides , Antiviral Agents
2.
bioRxiv ; 2023 Aug 21.
Article in English | MEDLINE | ID: mdl-37662376

ABSTRACT

Coxsackievirus B (CVB) infection of pancreatic ß cells is associated with ß-cell autoimmunity. We investigated how CVB impacts human ß cells and anti-CVB T-cell responses. ß cells were efficiently infected by CVB in vitro, downregulated HLA Class I and presented few, selected HLA-bound viral peptides. Circulating CD8+ T cells from CVB-seropositive individuals recognized only a fraction of these peptides, and only another sub-fraction was targeted by effector/memory T cells that expressed the exhaustion marker PD-1. T cells recognizing a CVB epitope cross-reacted with the ß-cell antigen GAD. Infected ß cells, which formed filopodia to propagate infection, were more efficiently killed by CVB than by CVB-reactive T cells. Thus, our in-vitro and ex-vivo data highlight limited T-cell responses to CVB, supporting the rationale for CVB vaccination trials for type 1 diabetes prevention. CD8+ T cells recognizing structural and non-structural CVB epitopes provide biomarkers to differentially follow response to infection and vaccination.

3.
J Clin Endocrinol Metab ; 109(1): 183-196, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37474341

ABSTRACT

CONTEXT: Validated assays to measure autoantigen-specific T-cell frequency and phenotypes are needed for assessing the risk of developing diabetes, monitoring disease progression, evaluating responses to treatment, and personalizing antigen-based therapies. OBJECTIVE: Toward this end, we performed a technical validation of a tetramer assay for HLA-DRA-DRB1*04:01, a class II allele that is strongly associated with susceptibility to type 1 diabetes (T1D). METHODS: HLA-DRA-DRB1*04:01-restricted T cells specific for immunodominant epitopes from islet cell antigens GAD65, IGRP, preproinsulin, and ZnT8, and a reference influenza epitope, were enumerated and phenotyped in a single staining tube with a tetramer assay. Single and multicenter testing was performed, using a clone-spiked specimen and replicate samples from T1D patients, with a target coefficient of variation (CV) less than 30%. The same assay was applied to an exploratory cross-sectional sample set with 24 T1D patients to evaluate the utility of the assay. RESULTS: Influenza-specific T-cell measurements had mean CVs of 6% for the clone-spiked specimen and 11% for T1D samples in single-center testing, and 20% and 31%, respectively, for multicenter testing. Islet-specific T-cell measurements in these same samples had mean CVs of 14% and 23% for single-center and 23% and 41% for multicenter testing. The cross-sectional study identified relationships between T-cell frequencies and phenotype and disease duration, sex, and autoantibodies. A large fraction of the islet-specific T cells exhibited a naive phenotype. CONCLUSION: Our results demonstrate that the assay is reproducible and useful to characterize islet-specific T cells and identify correlations between T-cell measures and clinical traits.


Subject(s)
Diabetes Mellitus, Type 1 , Influenza, Human , Humans , Diabetes Mellitus, Type 1/diagnosis , Cross-Sectional Studies , HLA-DR alpha-Chains , T-Lymphocytes
4.
Diabetologia ; 64(12): 2731-2740, 2021 12.
Article in English | MEDLINE | ID: mdl-34522983

ABSTRACT

AIMS/HYPOTHESIS: We report the case of a woman who underwent a partial pancreatectomy for a serous cystadenoma when aged 56 years. She had been diagnosed with diabetes 6 years before and had Hashimoto's thyroiditis. Despite positive anti-GAD autoantibodies (GADA) and previous surgery, she was transiently weaned off long-acting insulin. Blood glucose levels remained well controlled with low-dose long-acting insulin. Insulin needs eventually increased 8 years after surgery, in conjunction with anti-zinc transporter 8 (ZnT8) seroconversion and decreasing residual C-peptide. We hypothesised that the surgical pancreas specimens and blood autoimmune T cell responses may provide correlates of this indolent clinical course. METHODS: Beta and alpha cell area and insulitis were quantified on pancreas head tissue sections obtained at surgery. Blood T cell responses against beta cell antigens were analysed by enzyme-linked immunospot. RESULTS: Pancreas sections displayed reduced beta cell and normal alpha cell area (0.27% and 0.85% of section area, respectively). High-grade insulitis was observed, mostly in insulin-containing islets, with a peri-insulitis pattern enriched in T cells positive for regulatory forkhead box protein 3 (FOXP3). In vitro challenge with beta cell antigens of circulating T cells collected 4 and 9 years after surgery revealed dominant and persistent IL-10 responses; IFN-γ responses increasing at 9 years, after anti-ZnT8 seroconversion, was observed. CONCLUSIONS/INTERPRETATION: Despite persistent GADA and the histopathological finding of insulitis and decreased beta cell area 6 years after diabetes diagnosis, glycaemic control was maintained with low-dose insulin up to 8 years after surgery. Regulated T cell responses towards beta cell antigens and FOXP3-positive peri-insulitis suggest spontaneous long-term regulation of islet autoimmunity after substantial beta cell loss, and eventual autoimmune progression upon anti-ZnT8 seroconversion.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans , Autoantibodies/metabolism , Female , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Middle Aged , Pancreas/metabolism , Pancreaticoduodenectomy
5.
Diabetes ; 70(12): 2879-2891, 2021 12.
Article in English | MEDLINE | ID: mdl-34561224

ABSTRACT

In type 1 diabetes, autoimmune ß-cell destruction may be favored by neoantigens harboring posttranslational modifications (PTMs) such as citrullination. We studied the recognition of native and citrullinated glucose-regulated protein (GRP)78 peptides by CD8+ T cells. Citrullination modulated T-cell recognition and, to a lesser extent, HLA-A2 binding. GRP78-reactive CD8+ T cells circulated at similar frequencies in healthy donors and donors with type 1 diabetes and preferentially recognized either native or citrullinated versions, without cross-reactivity. Rather, the preference for native GRP78 epitopes was associated with CD8+ T cells cross-reactive with bacterial mimotopes. In the pancreas, a dominant GRP78 peptide was instead preferentially recognized when citrullinated. To further clarify these recognition patterns, we considered the possibility of citrullination in the thymus. Citrullinating peptidylarginine deiminase (Padi) enzymes were expressed in murine and human medullary epithelial cells (mTECs), with citrullinated proteins detected in murine mTECs. However, Padi2 and Padi4 expression was diminished in mature mTECs from NOD mice versus C57BL/6 mice. We conclude that, on one hand, the CD8+ T cell preference for native GRP78 peptides may be shaped by cross-reactivity with bacterial mimotopes. On the other hand, PTMs may not invariably favor loss of tolerance because thymic citrullination, although impaired in NOD mice, may drive deletion of citrulline-reactive T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Citrullination/physiology , Diabetes Mellitus, Type 1/immunology , Endoplasmic Reticulum Chaperone BiP/immunology , Epitopes, T-Lymphocyte/metabolism , Adolescent , Adult , Animals , Child , Citrullination/immunology , Diabetes Mellitus, Type 1/metabolism , Endoplasmic Reticulum Chaperone BiP/chemistry , Endoplasmic Reticulum Chaperone BiP/metabolism , Epitopes, T-Lymphocyte/chemistry , Female , Humans , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Middle Aged , Protein Processing, Post-Translational/immunology , Protein Processing, Post-Translational/physiology , Young Adult
6.
Diabetes ; 69(12): 2678-2690, 2020 12.
Article in English | MEDLINE | ID: mdl-32928873

ABSTRACT

The antigenic peptides processed by ß-cells and presented through surface HLA class I molecules are poorly characterized. Each HLA variant (e.g., the most common being HLA-A2 and HLA-A3) carries some peptide-binding specificity. Hence, features that, despite these specificities, remain shared across variants may reveal factors favoring ß-cell immunogenicity. Building on our previous description of the HLA-A2/A3 peptidome of ß-cells, we analyzed the HLA-A3-restricted peptides targeted by circulating CD8+ T cells. Several peptides were recognized by CD8+ T cells within a narrow frequency (1-50/106), which was similar in donors with and without type 1 diabetes and harbored variable effector/memory fractions. These epitopes could be classified as conventional peptides or neoepitopes, generated either via peptide cis-splicing or mRNA splicing (e.g., secretogranin-5 [SCG5]-009). As reported for HLA-A2-restricted peptides, several epitopes originated from ß-cell granule proteins (e.g., SCG3, SCG5, and urocortin-3). Similarly, H-2Kd-restricted CD8+ T cells recognizing the murine orthologs of SCG5, urocortin-3, and proconvertase-2 infiltrated the islets of NOD mice and transferred diabetes into NOD/scid recipients. The finding of granule proteins targeted in both humans and NOD mice supports their disease relevance and identifies the insulin granule as a rich source of epitopes, possibly reflecting its impaired processing in type 1 diabetes.


Subject(s)
Chromogranins/metabolism , Diabetes Mellitus, Type 1/metabolism , Adult , Alternative Splicing , Animals , CD8-Positive T-Lymphocytes , Case-Control Studies , Chromogranins/genetics , Computer Simulation , Data Mining , Diabetes Mellitus, Type 1/genetics , Epitopes , Female , Gene Expression Regulation , HLA-A3 Antigen , Humans , Insulin , Male , Mice , Mice, Inbred NOD , Neuroendocrine Secretory Protein 7B2/genetics , Neuroendocrine Secretory Protein 7B2/metabolism , Protein Binding , RNA, Messenger/genetics , Urocortins/genetics , Urocortins/metabolism , Young Adult
7.
Cell Metab ; 28(6): 946-960.e6, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30078552

ABSTRACT

Although CD8+ T-cell-mediated autoimmune ß cell destruction occurs in type 1 diabetes (T1D), the target epitopes processed and presented by ß cells are unknown. To identify them, we combined peptidomics and transcriptomics strategies. Inflammatory cytokines increased peptide presentation in vitro, paralleling upregulation of human leukocyte antigen (HLA) class I expression. Peptide sources featured several insulin granule proteins and all known ß cell antigens, barring islet-specific glucose-6-phosphatase catalytic subunit-related protein. Preproinsulin yielded HLA-A2-restricted epitopes previously described. Secretogranin V and its mRNA splice isoform SCG5-009, proconvertase-2, urocortin-3, the insulin gene enhancer protein ISL-1, and an islet amyloid polypeptide transpeptidation product emerged as antigens processed into HLA-A2-restricted epitopes, which, as those already described, were recognized by circulating naive CD8+ T cells in T1D and healthy donors and by pancreas-infiltrating cells in T1D donors. This peptidome opens new avenues to understand antigen processing by ß cells and for the development of T cell biomarkers and tolerogenic vaccination strategies.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/immunology , Transcriptome/immunology , Animals , Biomarkers/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cell Line , Corticotropin-Releasing Hormone/metabolism , Cytokines/metabolism , HLA Antigens/metabolism , Humans , Insulin/metabolism , Islet Amyloid Polypeptide/metabolism , Mice , Neuroendocrine Secretory Protein 7B2/metabolism , Proprotein Convertase 2/metabolism , Protein Precursors/metabolism , Proteomics/methods , Urocortins/metabolism
8.
Sci Immunol ; 3(20)2018 02 02.
Article in English | MEDLINE | ID: mdl-29429978

ABSTRACT

The human leukocyte antigen-A2 (HLA-A2)-restricted zinc transporter 8186-194 (ZnT8186-194) and other islet epitopes elicit interferon-γ secretion by CD8+ T cells preferentially in type 1 diabetes (T1D) patients compared with controls. We show that clonal ZnT8186-194-reactive CD8+ T cells express private T cell receptors and display equivalent functional properties in T1D and healthy individuals. Ex vivo analyses further revealed that CD8+ T cells reactive to ZnT8186-194 and other islet epitopes circulate at similar frequencies and exhibit a predominantly naïve phenotype in age-matched T1D and healthy donors. Higher frequencies of ZnT8186-194-reactive CD8+ T cells with a more antigen-experienced phenotype were detected in children versus adults, irrespective of disease status. Moreover, some ZnT8186-194-reactive CD8+ T cell clonotypes were found to cross-recognize a Bacteroides stercoris mimotope. Whereas ZnT8 was poorly expressed in thymic medullary epithelial cells, variable thymic expression levels of islet antigens did not modulate the peripheral frequency of their cognate CD8+ T cells. In contrast, ZnT8186-194-reactive cells were enriched in the pancreata of T1D patients versus nondiabetic and type 2 diabetic individuals. Thus, islet-reactive CD8+ T cells circulate in most individuals but home to the pancreas preferentially in T1D patients. We conclude that the activation of this common islet-reactive T cell repertoire and progression to T1D likely require defective peripheral immunoregulation and/or a proinflammatory islet microenvironment.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Pancreas/cytology , Pancreas/immunology , Adult , Cell Line , Child , Female , HLA-A2 Antigen/immunology , Healthy Volunteers , Humans , Male
9.
J Immunol ; 196(1): 256-263, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26608912

ABSTRACT

The quality of Ag-specific CD8(+) T cell responses is central to immune efficacy in infectious and malignant settings. Inducing effector CD8(+) T cells with potent functional properties is therefore a priority in the field of immunotherapy. However, the optimal assessment of new treatment strategies in humans is limited by currently available testing platforms. In this study, we introduce an original model of in vitro CD8(+) T cell priming, based on an accelerated dendritic cell coculture system, which uses unfractionated human PBMCs as the starting material. This approach enables the rapid evaluation of adjuvant effects on the functional properties of human CD8(+) T cells primed from Ag-specific naive precursors. We demonstrate that a selective TLR8 agonist, in combination with FLT3L, primes high-quality CD8(+) T cell responses. TLR8L/FLT3L-primed CD8(+) T cells displayed enhanced cytotoxic activity, polyfunctionality, and Ag sensitivity. The acquisition of this superior functional profile was associated with increased T-bet expression induced via an IL-12-dependent mechanism. Collectively, these data validate an expedited route to vaccine delivery or optimal T cell expansion for adoptive cell transfer.


Subject(s)
Immunotherapy, Adoptive/methods , Lymphocyte Activation/immunology , Membrane Proteins/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 8/agonists , Cell Line, Tumor , Coculture Techniques , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-12 Subunit p35/immunology , Interleukin-4/pharmacology , Leukocytes, Mononuclear/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/transplantation , Toll-Like Receptor 8/immunology
10.
Aging Cell ; 15(1): 14-21, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26472076

ABSTRACT

Aging is associated with impaired vaccine efficacy and increased susceptibility to infectious and malignant diseases. CD8(+) T-cells are key players in the immune response against pathogens and tumors. In aged mice, the dwindling naïve CD8(+) T-cell compartment is thought to compromise the induction of de novo immune responses, but no experimental evidence is yet available in humans. Here, we used an original in vitro assay based on an accelerated dendritic cell coculture system in unfractioned peripheral blood mononuclear cells to examine CD8(+) T-cell priming efficacy in human volunteers. Using this approach, we report that old individuals consistently mount quantitatively and qualitatively impaired de novo CD8(+) T-cell responses specific for a model antigen. Reduced CD8(+) T-cell priming capacity in vitro was further associated with poor primary immune responsiveness in vivo. This immune deficit likely arises as a consequence of intrinsic cellular defects and a reduction in the size of the naïve CD8(+) T-cell pool. Collectively, these findings provide new insights into the cellular immune insufficiencies that accompany human aging.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/cytology , Leukocytes, Mononuclear/cytology , Lymphocyte Activation/immunology , Adult , Aged , Aged, 80 and over , Dendritic Cells/immunology , Female , Flow Cytometry/methods , Humans , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Young Adult
11.
Diabetes ; 64(10): 3532-42, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25918233

ABSTRACT

The first signs of autoimmune activation leading to ß-cell destruction in type 1 diabetes (T1D) appear during the first months of life. Thus, the perinatal period offers a suitable time window for disease prevention. Moreover, thymic selection of autoreactive T cells is most active during this period, providing a therapeutic opportunity not exploited to date. We therefore devised a strategy by which the T1D-triggering antigen preproinsulin fused with the immunoglobulin (Ig)G Fc fragment (PPI-Fc) is delivered to fetuses through the neonatal Fc receptor (FcRn) pathway, which physiologically transfers maternal IgGs through the placenta. PPI-Fc administered to pregnant PPIB15-23 T-cell receptor-transgenic mice efficiently accumulated in fetuses through the placental FcRn and protected them from subsequent diabetes development. Protection relied on ferrying of PPI-Fc to the thymus by migratory dendritic cells and resulted in a rise in thymic-derived CD4(+) regulatory T cells expressing transforming growth factor-ß and in increased effector CD8(+) T cells displaying impaired cytotoxicity. Moreover, polyclonal splenocytes from nonobese diabetic (NOD) mice transplacentally treated with PPI-Fc were less diabetogenic upon transfer into NOD.scid recipients. Transplacental antigen vaccination provides a novel strategy for early T1D prevention and, further, is applicable to other immune-mediated conditions.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Histocompatibility Antigens Class I/metabolism , Insulin/metabolism , Maternal-Fetal Exchange/physiology , Protein Precursors/metabolism , Receptors, Fc/metabolism , Animals , Autoimmunity , Cell Proliferation , Dendritic Cells/physiology , Female , Gene Expression Regulation, Developmental/physiology , Histocompatibility Antigens Class I/genetics , Humans , Insulin/administration & dosage , Mice , Mice, Inbred NOD , Mice, Transgenic , Placenta/metabolism , Pregnancy , Protein Precursors/administration & dosage , Receptors, Fc/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Specific Pathogen-Free Organisms , Thymus Gland/physiology
12.
J Autoimmun ; 58: 48-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25634360

ABSTRACT

Most autoimmune diseases (AID) are linked to an imbalance between autoreactive effector T cells (Teffs) and regulatory T cells (Tregs). While blocking Teffs with immunosuppression has long been the only therapeutic option, activating/expanding Tregs may achieve the same objective without the toxicity of immunosuppression. We showed that low-dose interleukin-2 (ld-IL-2) safely expands/activates Tregs in patients with AID, such HCV-induced vasculitis and Type 1 Diabetes (T1D). Here we analyzed the kinetics and dose-relationship of IL-2 effects on immune responses in T1D patients. Ld-IL-2 therapy induced a dose-dependent increase in CD4(+)Foxp3(+) and CD8(+)Foxp3(+) Treg numbers and proportions, the duration of which was markedly dose-dependent. Tregs expressed enhanced levels of activation markers, including CD25, GITR, CTLA-4 and basal pSTAT5, and retained a 20-fold higher sensitivity to IL-2 than Teff and NK cells. Plasma levels of regulatory cytokines were increased in a dose-dependent manner, while cytokines linked to Teff and Th17 inflammatory cells were mostly unchanged. Global transcriptome analyses showed a dose-dependent decrease in immune response signatures. At the highest dose, Teff responses against beta-cell antigens were suppressed in all 4 patients tested. These results inform of broader changes induced by ld-IL-2 beyond direct effects on Tregs, and relevant for further development of ld-IL-2 for therapy and prevention of T1D, and other autoimmune and inflammatory diseases.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Immunotherapy/methods , Interleukin-2/administration & dosage , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , CD8 Antigens/metabolism , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Type 1/immunology , Dose-Response Relationship, Drug , Drug Dosage Calculations , Female , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Humans , Immunosuppression Therapy , Insulin-Secreting Cells/immunology , Interleukin-2/adverse effects , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Male , Middle Aged , STAT5 Transcription Factor/metabolism , Transcriptome , Young Adult
13.
Diabetes ; 61(10): 2546-55, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22997432

ABSTRACT

The cartography of ß-cell epitopes targeted by CD8(+) T cells in type 1 diabetic (T1D) patients remains largely confined to the common HLA-A2 restriction. We aimed to identify ß-cell epitopes restricted by the HLA-B7 (B*07:02) molecule, which is associated with mild T1D protection. Using DNA immunization on HLA-B7-transgenic mice and prediction algorithms, we identified GAD and preproinsulin candidate epitopes. Interferon-γ (IFN-γ) enzyme-linked immunospot assays on peripheral blood mononuclear cells showed that most candidates were recognized by new-onset T1D patients, but not by type 2 diabetic and healthy subjects. Some epitopes were highly immunodominant and specific to either T1D children (GAD(530-538); 44% T cell-positive patients) or adults (GAD(311-320); 38%). All epitopes displayed weak binding affinity and stability for HLA-B7 compared with HLA-A2-restricted ones, a general feature of HLA-B7. Single-cell PCR analysis on ß-cell-specific (HLA-B7 tetramer-positive) T cells revealed uniform IFN-γ and transforming growth factor-ß (TGF-ß) mRNA expression, different from HLA-A2-restricted T cells. We conclude that HLA-B7-restricted islet epitopes display weak HLA-binding profiles, are different in T1D children and adults, and are recognized by IFN-γ(+)TGF-ß(+)CD8(+) T cells. These features may explain the T1D-protective effect of HLA-B7. The novel epitopes identified should find valuable applications for immune staging of HLA-B7(+) individuals.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Epitopes/genetics , HLA-B7 Antigen/genetics , Insulin-Secreting Cells/metabolism , Adolescent , Adult , Aged , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Child , Child, Preschool , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Epitopes/immunology , Epitopes/metabolism , Female , HLA-B7 Antigen/metabolism , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Mice , Middle Aged , Transforming Growth Factor beta/metabolism
14.
Blood ; 118(8): 2128-37, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21715316

ABSTRACT

Detection of human Ag-specific T cells is limited by sensitivity and blood requirements. As dendritic cells (DCs) can potently stimulate T cells, we hypothesized that their induction in PBMCs in situ could link Ag processing and presentation to Ag-specific T-cell activation. To this end, unfractionated PBMCs (fresh or frozen) or whole blood were incubated for 48 hours with protein or peptide Ag together with different DC-activating agents to rapidly and sequentially induce, pulse, and mature DCs. DC activation was therefore lined up with Ag recognition by neighboring T cells, thus telescoping the sequential steps of T-cell activation. Efficient processing of protein Ags made prior knowledge of epitopes and HLA restrictions dispensable. While reducing stimulation time, manipulation and blood requirements, in situ DC induction specifically amplified Ag-specific T-cell responses (cytokine secretion, proliferation, CD137/CD154 up-regulation, and binding of peptide-HLA multimers). IL-1ß, although released by DCs, was also secreted in an Ag-specific fashion, thus providing an indirect biomarker of T-cell responses. These accelerated cocultured DC (acDC) assays offered a sensitive means with which to evaluate T-cell responses to viral and melanoma Ag vaccination, and may therefore find application for immune monitoring in viral, tumor, autoimmune, and transplantation settings.


Subject(s)
Antigens , Dendritic Cells/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation , Antigens/administration & dosage , Cancer Vaccines/administration & dosage , Cell Proliferation , Coculture Techniques , Cytokines/biosynthesis , Cytokines/blood , Dendritic Cells/cytology , Dendritic Cells/drug effects , Epitopes/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HLA Antigens/administration & dosage , Humans , Interleukin-4/pharmacology , Lymphocyte Activation , Melanoma/immunology , Melanoma/therapy , Melanoma-Specific Antigens/administration & dosage , Mice , Recombinant Proteins , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Vaccination
15.
J Immunol Methods ; 359(1-2): 28-36, 2010 Jul 31.
Article in English | MEDLINE | ID: mdl-20641145

ABSTRACT

Assays detecting antigen (Ag)-specific T-cell responses in immune-mediated processes are increasingly employed to understand disease pathogenesis and immune staging. The quantity and quality of starting peripheral blood mononuclear cell (PBMC) preparations are important factors in the performance of such assays. We therefore compared final PBMC yield and function by modifying parameters at the blood drawing, storage and processing steps. While drawing blood in vacuum-driven tubes or syringes and separating PBMCs on density gradients using standard or membrane (Leucosep) tubesmade no difference, storing tubes for 18 h without any agitation led to PBMC preparations contaminated with granulocytes and decreased interferon (IFN)-gamma enzyme-linked immunospot (ELISpot) responses. Even agitated blood showed a trend towards reduced ELISpot responses and increased human leukocyte Ag (HLA) multimer readouts when stored for 18 h compared to 3 h. These changes were reduced by diluting blood prior to storage. Washing PBMCs with media containing 10% human serum increased PBMC yields by 40.5%, without affecting ELISpot responses and multimer counts. However, washes with > 10% human serum decreased multimer counts, with no additional improvement in PBMC yields. These findings may be relevant for optimizing and harmonizing PBMC processing procedures for T-cell assays.


Subject(s)
Blood Specimen Collection/methods , Cell Separation/methods , Immunoenzyme Techniques/methods , Leukocytes, Mononuclear/cytology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Adult , Blood Preservation/methods , Centrifugation, Density Gradient , Female , Granulocytes/cytology , Granulocytes/immunology , HLA-A2 Antigen/blood , HLA-A2 Antigen/immunology , Humans , Interferon-gamma/immunology , Leukocyte Count , Leukocytes, Mononuclear/immunology , Male
16.
Diabetes ; 56(3): 613-21, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17327428

ABSTRACT

Despite the understanding that type 1 diabetes pathogenesis is mediated by T-cells, detection of these rare lymphocytes remains largely elusive. Suitable T-cell assays are highly needed, since they could offer preclinical diagnoses and immune surrogate end points for clinical trials. Although CD4+ T-cell assays have met with limited success, CD8+ T-cells are increasingly recognized as key actors in the diabetes of the NOD mouse. CD8+ T-cells are likely to play a role also in humans and may provide new markers of beta-cell autoimmunity. Taking advantage of a panel of HLA-A2-restricted beta-cell epitopes derived from preproinsulin, GAD, and islet glucose-6-phosphatase catalytic subunit-related protein (IGRP), we have implemented an islet-specific CD8+ T-cell interferon-gamma enzyme-linked immunospot (ISL8Spot) assay. The ISL8Spot assay is capable of detecting and quantifying beta-cell-reactive CD8+ T-cells directly ex vivo, without any preliminary expansion, using either fresh or frozen samples. Positive ISL8Spot responses separate new-onset diabetic and healthy samples with high accuracy (86% sensitivity, 91% specificity), using as few as five immunodominant epitopes. Moreover, sensitivity reaches 100% when the ISL8Spot assay is complemented by antibody determinations. Combination of CD8+ T-cell measurements with immune intervention strategies may open new avenues toward type 1 diabetes prediction and prevention.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/metabolism , Insulin-Secreting Cells/metabolism , Adolescent , Adult , Enzyme-Linked Immunosorbent Assay/methods , Epitopes, T-Lymphocyte/metabolism , Female , HLA-A2 Antigen/metabolism , Humans , Male , Middle Aged , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL