Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 260
Filter
1.
PLoS One ; 19(5): e0287877, 2024.
Article in English | MEDLINE | ID: mdl-38787820

ABSTRACT

Type 1 diabetes (T1D) is characterized by HLA class I-mediated presentation of autoantigens on the surface of pancreatic ß-cells. Recognition of these autoantigens by CD8+ T cells results in the destruction of pancreatic ß-cells and, consequently, insulin deficiency. Most epitopes presented at the surface of ß-cells derive from the insulin precursor molecule proinsulin. The intracellular processing pathway(s) involved in the generation of these peptides are poorly defined. In this study, we show that a proinsulin B-chain antigen (PPIB5-14) originates from proinsulin molecules that are processed by ER-associated protein degradation (ERAD) and thus originate from ER-resident proteins. Furthermore, screening genes encoding for E2 ubiquitin conjugating enzymes, we identified UBE2G2 to be involved in proinsulin degradation and subsequent presentation of the PPIB10-18 autoantigen. These insights into the pathway involved in the generation of insulin-derived peptides emphasize the importance of proinsulin processing in the ER to T1D pathogenesis and identify novel targets for future T1D therapies.


Subject(s)
Autoantigens , Endoplasmic Reticulum-Associated Degradation , Proinsulin , Proteolysis , Ubiquitin-Conjugating Enzymes , Proinsulin/metabolism , Proinsulin/immunology , Proinsulin/genetics , Autoantigens/metabolism , Autoantigens/immunology , Humans , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Antigen Presentation/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/immunology
2.
Proc Natl Acad Sci U S A ; 119(15): e2110987119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35385352

ABSTRACT

Antigen-specific immunotherapy involves the delivery of self-antigens as proteins or peptides (or using nucleic acids encoding them) to reestablish tolerance. The Endotope platform supports the optimal presentation of endogenously expressed epitopes on appropriate major histocompatibility complex (MHC) class I and II molecules. Using specific epitopes that are disease-relevant (including neoepitopes and mimotopes) and restricted to the subject's MHC haplotypes provides a more focused and tailored way of targeting autoreactive T cells. We evaluated the efficacy of an Endotope DNA vaccine tailored to the nonobese diabetic (NOD) mouse in parallel to one expressing the Proinsulin protein, a central autoantigen in NOD mice, and assessed the influence of several parameters (e.g., route, dosing frequency, disease stage) on diabetes prevention. Secretion of encoded peptides and intradermal delivery of DNA offered more effective disease prevention. Long-term weekly treatments were needed to achieve protection that can persist after discontinuation, likely mediated by regulatory T cells induced by at least one epitope. Although epitopes were presented for at least 2 wk, weekly treatments were needed, at least initially, to achieve significant protection. While Endotope and Proinsulin DNA vaccines were effective at both the prediabetic normoglycemic and dysglycemic stages of disease, Proinsulin provided better protection in the latter stage, particularly in animals with slower progression of disease, and Endotope limited insulitis the most in the earlier stage. Thus, our data support the possibility of applying a precision medicine approach based on tailored epitopes for the treatment of tissue-specific autoimmune diseases with DNA vaccines.


Subject(s)
Diabetes Mellitus, Type 1 , Proinsulin , Vaccines, DNA , Animals , Diabetes Mellitus, Type 1/prevention & control , Epitopes, T-Lymphocyte/immunology , Mice , Mice, Inbred NOD , Precision Medicine , Proinsulin/genetics , Proinsulin/immunology , Vaccination , Vaccines, DNA/immunology
3.
Front Immunol ; 12: 648021, 2021.
Article in English | MEDLINE | ID: mdl-33889155

ABSTRACT

Introduction: Insulin and proinsulin autoantibodies (IAA/PAA) are usually the first markers to appear in patients with type 1 Diabetes Mellitus (T1DM) and their prevalence ranges from 10 to 60% in the child-adolescent population. The reference method for IAA/PAA detection is the Radioligand Binding Assay (RBA), a highly specific and sensitive technique, but expensive and polluting. The aim of this work was to develop a novel flow cytometric microsphere-based immunoassay (FloCMIA) for PAA detection, employing recombinant human proinsulin (PI), as an alternative method to RBA, less expensive and harmful to the environment. Materials and Methods: Human PI was expressed as Thioredoxin fusion protein (TrxPI) in E. coli and a fraction was biotinylated. A double paratope model was used in which samples were incubated with TrxPI-biotin and microspheres adsorbed with TrxPI. The immune complexes were revealed using Streptavidin-Phycoerythrin. The geometric mean of the signals was analyzed, and the results were expressed as Standard Deviation scores (SDs). Sera from 100 normal human control and from 111 type 1 diabetic patients were evaluated by FloCMIA. To correlate the novel assay with RBA, 51 diabetic patients were selected, spanning a wide range of PAA reactivity by RBA. Results: The study of ROC curves allowed choosing a cut-off value of 3.0 SDs and the AUC was 0.705, indicating that FloCMIA has fair ability to distinguish between samples from each group. A prevalence of 50% for PAA was obtained in the population of diabetic patients studied. The specificity was 96% and the analytical sensitivity (percentage of patients RBA positive, also positive by FloCMIA) was 69%. There was a substantial agreement between methods (kappa statistic=0.700). Conclusions: A novel immunoassay based on flow cytometry that uses easy-to produce recombinant PI was developed. This assay constitutes an innovative and cost-effective alternative to RBA for the determination of PAA in patients' sera. The method developed here, presents good performance and a wide dynamic range together with a small required sample volume. Furthermore, these results make it possible to develop multiplex immunoassays that allow the combined detection of autoantibodies present in T1DM and other related autoimmune diseases.


Subject(s)
Autoantibodies/immunology , Autoantigens/metabolism , Diabetes Mellitus, Type 1/immunology , Escherichia coli/metabolism , Flow Cytometry/methods , Proinsulin/immunology , Proinsulin/metabolism , Adolescent , Adult , Autoantibodies/blood , Autoantigens/genetics , Biomarkers/blood , Case-Control Studies , Child , Child, Preschool , Diabetes Mellitus, Type 1/blood , Escherichia coli/genetics , Female , Humans , Immunoassay/methods , Infant , Male , Microspheres , Middle Aged , Proinsulin/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Young Adult
4.
Front Immunol ; 12: 645817, 2021.
Article in English | MEDLINE | ID: mdl-33841427

ABSTRACT

T-cell responses to insulin and its precursor proinsulin are central to islet autoimmunity in humans and non-obese diabetic (NOD) mice that spontaneously develop autoimmune diabetes. Mice have two proinsulin genes proinsulin -1 and 2 that are differentially expressed, with predominant proinsulin-2 expression in the thymus and proinsulin-1 in islet beta-cells. In contrast to proinsulin-2, proinsulin-1 knockout NOD mice are protected from autoimmune diabetes. This indicates that proinsulin-1 epitopes in beta-cells maybe preferentially targeted by autoreactive T cells. To study the contribution of proinsulin-1 reactive T cells in autoimmune diabetes, we generated transgenic NOD mice with tetracycline-regulated expression of proinsulin-1 in antigen presenting cells (TIP-1 mice) with an aim to induce immune tolerance. TIP-1 mice displayed a significantly reduced incidence of spontaneous diabetes, which was associated with reduced severity of insulitis and insulin autoantibody development. Antigen experienced proinsulin specific T cells were significantly reduced in in TIP-1 mice indicating immune tolerance. Moreover, T cells from TIP-1 mice expressing proinsulin-1 transferred diabetes at a significantly reduced frequency. However, proinsulin-1 expression in APCs had minimal impact on the immune responses to the downstream antigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) and did not prevent diabetes in NOD 8.3 mice with a pre-existing repertoire of IGRP reactive T cells. Thus, boosting immune tolerance to proinsulin-1 partially prevents islet-autoimmunity. This study further extends the previously established role of proinsulin-1 epitopes in autoimmune diabetes in NOD mice.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Immune Tolerance , Proinsulin/physiology , Animals , Autoantibodies/analysis , Glucose-6-Phosphatase/physiology , Insulin/immunology , Mice , Mice, Inbred NOD , Proinsulin/genetics , Proinsulin/immunology
5.
Biochem Biophys Res Commun ; 534: 680-686, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33208230

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease caused by destruction of insulin-producing ß cells. The response of autoreactive T cells to ß cell antigens plays a central role in the development of T1D. Recently, fusion peptides composed by insulin C-peptide fragments and other proteins were reported as ß cell target antigens for diabetogenic CD4+ T cells in non-obese diabetic (NOD) mice. In this study, we generated a T cell-receptor (TCR)-like monoclonal antibody (mAb) against a fusion peptide bound to major histocompatibility complex (MHC) class II component to elucidate the function of the fusion peptides in T1D. In addition, we developed a novel NFAT-GFP TCR reporter system to evaluate the TCR-like mAb. The NFAT-GFP reporter T cells expressing the diabetogenic TCR were specifically activated by the fusion peptide presented on the MHC class II molecules. By using the NFAT-GFP reporter T cells, we showed that the TCR-like mAb blocks the diabetogenic T cell response against the fusion peptide presented on the MHC class II molecules. Furthermore, the development of T1D was ameliorated when pre-diabetic NOD mice were treated with this mAb. These findings suggest that NFAT-GFP reporter T cells are useful to assess the function of specific TCR and the recognition of fusion peptides by T cells is crucial for the pathogenesis of T1D.


Subject(s)
Antibodies, Monoclonal/pharmacology , Diabetes Mellitus, Type 1/prevention & control , Proinsulin/antagonists & inhibitors , Proinsulin/immunology , Receptors, Antigen, T-Cell/immunology , Animals , C-Peptide/antagonists & inhibitors , C-Peptide/genetics , C-Peptide/immunology , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Disease Progression , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Mice , Mice, Inbred NOD , Proinsulin/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology
6.
J Immunol ; 204(9): 2349-2359, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32229538

ABSTRACT

Type 1 diabetes (T1D) is a T cell-mediated autoimmune disease in which the insulin-producing ß cells within the pancreas are destroyed. Identification of target Ags and epitopes of the ß cell-reactive T cells is important both for understanding T1D pathogenesis and for the rational development of Ag-specific immunotherapies for the disease. Several studies suggest that proinsulin is an early and integral target autoantigen in T1D. However, proinsulin epitopes recognized by human CD4+ T cells have not been comprehensively characterized. Using a dye dilution-based T cell cloning method, we generated and characterized 24 unique proinsulin-specific CD4+ T cell clones from the peripheral blood of 17 individuals who carry the high-risk DR3-DQ2 and/or DR4-DQ8 HLA class II haplotypes. Some of the clones recognized previously reported DR4-restricted epitopes within the C-peptide (C25-35) or A-chain (A1-15) of proinsulin. However, we also characterized DR3-restricted epitopes within both the B-chain (B16-27 and B22-C3) and C-peptide (C25-35). Moreover, we identified DQ2-restricted epitopes within the B-chain and several DQ2- or DQ8-restricted epitopes within the C-terminal region of C-peptide that partially overlap with previously reported DQ-restricted epitopes. Two of the DQ2-restricted epitopes, B18-26 and C22-33, were shown to be naturally processed from whole human proinsulin. Finally, we observed a higher frequency of CDR3 sequences matching the TCR sequences of the proinsulin-specific T cell clones in pancreatic lymph node samples compared with spleen samples. In conclusion, we confirmed several previously reported epitopes but also identified novel (to our knowledge) epitopes within proinsulin, which are presented by HLA class II molecules associated with T1D risk.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/immunology , HLA-DQ Antigens/immunology , Proinsulin/immunology , Adolescent , Amino Acid Sequence , Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Child , Child, Preschool , Humans , Infant , Insulin/immunology , Insulin-Secreting Cells/immunology , Spleen/immunology
7.
Diabetologia ; 63(6): 1174-1185, 2020 06.
Article in English | MEDLINE | ID: mdl-32157332

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to characterise islet autoantibody profiles and immune cell phenotypes in slow progressors to type 1 diabetes. METHODS: Immunological variables were compared across peripheral blood samples obtained from slow progressors to type 1 diabetes, individuals with newly diagnosed or long-standing type 1 diabetes, and healthy individuals. Polychromatic flow cytometry was used to characterise the phenotypic attributes of B and T cells. Islet autoantigen-specific B cells were quantified using an enzyme-linked immunospot (ELISpot) assay and islet autoantigen-specific CD8+ T cells were quantified using peptide-HLA class I tetramers. Radioimmunoassays were used to detect islet autoantibodies. Sera were assayed for various chemokines, cytokines and soluble receptors via ELISAs. RESULTS: Islet autoantibodies were lost over time in slow progressors. Various B cell subsets expressed higher levels of CD95 in slow progressors, especially after polyclonal stimulation, compared with the corresponding B cell subsets in healthy donors (p < 0.05). The phenotypic characteristics of CD4+ and CD8+ T cells were similar in slow progressors and healthy donors. Lower frequencies of CD4+ T cells with a central memory phenotype (CD27int, CD127+, CD95int) were observed in slow progressors compared with healthy donors (mean percentage of total CD4+ T cells was 3.00% in slow progressors vs 4.67% in healthy donors, p < 0.05). Autoreactive B cell responses to proinsulin were detected at higher frequencies in slow progressors compared with healthy donors (median no. of spots was 0 in healthy donors vs 24.34 in slow progressors, p < 0.05) in an ELISpot assay. Islet autoantigen-specific CD8+ T cell responses were largely absent in slow progressors and healthy donors. Serum levels of DcR3, the decoy receptor for CD95L, were elevated in slow progressors compared with healthy donors (median was 1087 pg/ml in slow progressors vs 651 pg/ml in healthy donors, p = 0.06). CONCLUSIONS/INTERPRETATION: In this study, we found that slow progression to type 1 diabetes was associated with a loss of islet autoantibodies and a distinct B cell phenotype, consistent with enhanced apoptotic regulation of peripheral autoreactivity via CD95. These phenotypic changes warrant further studies in larger cohorts to determine their functional implications.


Subject(s)
Autoantibodies/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , fas Receptor/immunology , Autoantibodies/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/metabolism , Flow Cytometry , Humans , Proinsulin/immunology , Proinsulin/metabolism , fas Receptor/metabolism
8.
Article in English | MEDLINE | ID: mdl-32098895

ABSTRACT

OBJECTIVE: Here we looked for possible mechanisms regulating the progression of type 1 diabetes mellitus (T1DM). In this disease, autoaggressive T cells (T conventional cells, Tconvs) not properly controlled by T regulatory cells (Tregs) destroy pancreatic islets. RESEARCH DESIGN AND METHODS: We compared the T-cell compartment of patients with newly diagnosed T1DM (NDT1DM) with long-duration T1DM (LDT1DM) ones. The third group consisted of patients with LDT1DM treated previously with polyclonal Tregs (LDT1DM with Tregs). We have also looked if the differences might be dependent on the antigen specificity of Tregs expanded for clinical use and autologous sentinel Tconvs. RESULTS: Patients with LDT1DM were characterized by T-cell immunosenescence-like changes and expansion of similar vß/T-cell receptor (TCR) clones in Tconvs and Tregs. The treatment with Tregs was associated with some inhibition of these effects. Patients with LDT1DM possessed an increased percentage of various proinsulin-specific T cells but not GAD65-specific ones. The percentages of all antigen-specific subsets were higher in the expansion cultures than in the peripheral blood. The proliferation was more intense in proinsulin-specific Tconvs than in specific Tregs but the levels of some proinsulin-specific Tregs were exceptionally high at baseline and remained higher in the expanded clinical product than the levels of respective Tconvs in sentinel cultures. CONCLUSIONS: T1DM is associated with immunosenescence-like changes and reduced diversity of T-cell clones. Preferential expansion of the same TCR families in both Tconvs and Tregs suggests a common trigger/autoantigen responsible. Interestingly, the therapy with polyclonal Tregs was associated with some inhibition of these effects. Proinsulin-specific Tregs appeared to be dominant in the immune responses in patients with T1DM and probably associated with better control over respective autoimmune Tconvs. TRIAL REGISTRATION NUMBER: EudraCT 2014-004319-35.


Subject(s)
Adoptive Transfer/methods , Autoantigens/immunology , Autoimmunity , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Proinsulin/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Autoantibodies/immunology , Body Mass Index , Cellular Senescence/immunology , Child , Cohort Studies , Diabetes Mellitus, Type 1/diagnosis , Female , Glutamate Decarboxylase/immunology , Humans , Islets of Langerhans/immunology , Male , Phenotype , Receptors, Antigen, T-Cell/immunology , Treatment Outcome
9.
J Autoimmun ; 107: 102361, 2020 02.
Article in English | MEDLINE | ID: mdl-31776056

ABSTRACT

Induction of antigen-specific regulatory T cells (Tregs) in vivo is the holy grail of current immune-regulating therapies in autoimmune diseases, such as type 1 diabetes. Tolerogenic dendritic cells (tolDCs) generated from monocytes by a combined treatment with vitamin D and dexamethasone (marked by CD52hi and CD86lo expression) induce antigen-specific Tregs. We evaluated the phenotypes of these Tregs using high-dimensional mass cytometry to identify a surface-based T cell signature of tolerogenic modulation. Naïve CD4+ T cells were stimulated with tolDCs or mature inflammatory DCs pulsed with proinsulin peptide, after which the suppressive capacity, cytokine production and phenotype of stimulated T cells were analysed. TolDCs induced suppressive T cell lines that were dominated by a naïve phenotype (CD45RA+CCR7+). These naïve T cells, however, did not show suppressive capacity, but were arrested in their naïve status. T cell cultures stimulated by tolDC further contained memory-like (CD45RA-CCR7-) T cells expressing regulatory markers Lag-3, CD161 and ICOS. T cells expressing CD25lo or CD25hi were most prominent and suppressed CD4+ proliferation, while CD25hi Tregs also effectively supressed effector CD8+ T cells. We conclude that tolDCs induce antigen-specific Tregs with various phenotypes. This extends our earlier findings pointing to a functionally diverse pool of antigen-induced and specific Tregs and provides the basis for immune-monitoring in clinical trials with tolDC.


Subject(s)
Autoimmunity , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune Tolerance , Peptides/immunology , Proinsulin/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers , Cytokines/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Humans , Immunophenotyping , Monocytes/immunology , Monocytes/metabolism
10.
J Autoimmun ; 105: 102304, 2019 12.
Article in English | MEDLINE | ID: mdl-31327552

ABSTRACT

It is still an enigma why T cell autoreactivity in type 1 diabetes targets few beta cell antigens only. Among these, one primary autoantigen is pro(insulin). Autoimmune T cells preferentially recognise three epitopes on the proinsulin molecule, of which the peptide region B:11-23 is the dominant one. Interestingly, the three regions superimpose with binding sites of the chaperone hsp70, the region B:11-23 being the strongest binding one. Absence of an intact core region B:15-17 prevents autoimmune diabetes in NOD as well as binding of hsp70. A role of hsp70 in selecting autoimmune epitopes is supported by the ability of this and other chaperones to deliver bound peptides to MHC class I and II molecules for efficient antigen presentation. Binding of hsp70 to receptors on antigen presenting cells such as TLR4 results in costimulatory signals for T cell activation. Strongest effects are seen for the mixture of hsp70 with the peptide B:11-23. Thus, hsp70 may assist in proinsulin epitope selection and efficient presentation to autoreactive T cells. The concept of chaperone guided immune reactivity may also apply to other autoimmune diseases.


Subject(s)
Autoimmunity/immunology , Diabetes Mellitus, Type 1/immunology , Molecular Chaperones/immunology , Peptides/immunology , Proinsulin/immunology , Animals , Humans , Insulin/immunology , T-Lymphocytes/immunology
11.
Pediatr Diabetes ; 20(3): 271-281, 2019 05.
Article in English | MEDLINE | ID: mdl-30635950

ABSTRACT

BACKGROUND AND OBJECTIVES: Regulatory T cells (Tregs) play an important role in maintaining tolerance to self-antigens. Defects in the frequency and function of polyclonal Tregs have been reported in type 1 diabetes (T1D). However, characteristics of proinsulin (PI)-specific Tregs in human T1D have not yet been explored. Therefore, we aimed to characterize PI-specific Tregs in two distinct pathophysiological subtypes of T1D, juvenile-onset T1D (JOT1D) and adult-onset T1D (AOT1D), distinguished by the age of onset. METHODS: Peripheral blood mononuclear cells of the recruited subjects were stimulated in vitro with PI-derived peptides. PI-specific Tregs were characterized by flow cytometry using the combination of markers CD25, CD137, FOXP3 and CD45RA. RESULTS: Firstly, we observed similar frequencies of polyclonal Tregs in the T1D (n = 25) and healthy control (HC) (n = 20) subjects (P = 0.96), with a positive correlation between age and frequency of polyclonal Tregs (r = +0.35, P = 0.04). While the frequency of polyclonal Tregs was higher in AOT1D group (P = 0.02), both JOT1D (n = 14) and AOT1D groups (n = 11) had a comparable frequency of PI-specific Tregs in their peripheral blood. The frequency of PI-specific memory Tregs was significantly high in both the JOT1D (P = 0.02) and AOT1D (P = 0.009) groups compared to their respective HC groups (n = 10). Finally, we observed no significant difference in the expression of FOXP3 and IL-2 receptor in PI-specific Tregs in all the groups. CONCLUSIONS: Unlike polyclonal Tregs, both T1D subtypes harbor comparable frequencies of PI-specific Tregs. Chronic antigen presentation results in a distinct memory-like phenotype of PI-specific Tregs in these subjects irrespective of the age of disease onset.


Subject(s)
Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/immunology , Proinsulin/immunology , T-Lymphocytes, Regulatory/pathology , Adolescent , Adult , Age Factors , Age of Onset , Autoantibodies/analysis , Autoantibodies/blood , Biomarkers/analysis , Biomarkers/blood , Case-Control Studies , Child , Diabetes Mellitus, Type 1/pathology , Female , Humans , Immunophenotyping , Male
12.
Cell Immunol ; 335: 68-75, 2019 01.
Article in English | MEDLINE | ID: mdl-30428974

ABSTRACT

CD4+ T cell responses are thought to play a role in type 1 diabetes (T1D). However, detection and characterization of T cells that respond to beta cell epitopes in subjects with T1D has been limited by technical obstacles, including the inherently low frequencies in peripheral blood and variable responsiveness of individual subjects to single epitopes. We implemented a multicolor staining approach that allows direct ex vivo characterization of multiple CD4+ T cell specificities in a single sample. Here we demonstrate and apply that multicolor approach to directly measure the frequency and phenotype of beta cell specific CD4+ T cells in T1D patients and HLA matched controls. For this work we utilized five DR0401 restricted peptides from proinsulin, GAD65, IA-2, and IGRP, which were previously reported as disease relevant epitopes. Surprisingly, although responses to each of these peptides can be readily detected after in vitro expansion, our results indicated that only proinsulin specific T cells were consistently detectable at moderate frequencies in subjects with T1D. Characterization of beta cell specific CD4+ T cells revealed only modest differences between subjects with T1D and healthy controls. Subjects with T1D did have higher proportions of CD45RA negative epitope specific T cells than controls. In patients epitope specific T cells were often CXCR3 positive and a substantial proportion were CCR7 negative, suggesting a Th1-like effector phenotype. Finally, we demonstrated that our multicolor staining approach is compatible with class I multimer analysis, facilitating the characterization of self-reactive CD4+ and CD8+ T cells using a single sample.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Adolescent , Adult , Aged , CD4-Positive T-Lymphocytes/metabolism , Epitopes , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunohistochemistry/methods , Insulin-Secreting Cells/metabolism , Male , Middle Aged , Proinsulin/immunology , Proinsulin/metabolism
13.
Proc Natl Acad Sci U S A ; 115(42): 10732-10737, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30275329

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease in which insulin-producing beta cells, found within the islets of Langerhans in the pancreas, are destroyed by islet-infiltrating T cells. Identifying the antigenic targets of beta-cell reactive T cells is critical to gain insight into the pathogenesis of T1D and develop antigen-specific immunotherapies. Several lines of evidence indicate that insulin is an important target of T cells in T1D. Because many human islet-infiltrating CD4+ T cells recognize C-peptide-derived epitopes, we hypothesized that full-length C-peptide (PI33-63), the peptide excised from proinsulin as it is converted to insulin, is a target of CD4+ T cells in people with T1D. CD4+ T cell responses to full-length C-peptide were detected in the blood of: 14 of 23 (>60%) people with recent-onset T1D, 2 of 15 (>13%) people with long-standing T1D, and 1 of 13 (<8%) HLA-matched people without T1D. C-peptide-specific CD4+ T cell clones, isolated from six people with T1D, recognized epitopes from the entire 31 amino acids of C-peptide. Eighty-six percent (19 of 22) of the C-peptide-specific clones were restricted by HLA-DQ8, HLA-DQ2, HLA-DQ8trans, or HLA-DQ2trans, HLA alleles strongly associated with risk of T1D. We also found that full-length C-peptide was a much more potent agonist of some CD4+ T cell clones than an 18mer peptide encompassing the cognate epitope. Collectively, our findings indicate that proinsulin C-peptide is a key target of autoreactive CD4+ T cells in T1D. Hence, full-length C-peptide is a promising candidate for antigen-specific immunotherapy in T1D.


Subject(s)
Autoantigens/immunology , C-Peptide/immunology , C-Peptide/metabolism , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/diagnosis , HLA Antigens/immunology , Islets of Langerhans/immunology , Proinsulin/immunology , Adolescent , Adult , Cells, Cultured , Child , Child, Preschool , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Humans , Middle Aged , Young Adult
14.
Acta Diabetol ; 55(3): 263-270, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29305766

ABSTRACT

AIMS: Insulin autoantibodies (IAA) are often the first marker of autoimmunity detected in children in the preclinical phase of type 1 diabetes (T1D). Currently, the vast majority of laboratories adopt the radiobinding micro-assay (RBA) for measuring IAA. Our aim was to replace RBA with a novel non-radioactive IAA Luciferase Immuno Precipitation System (LIPS) assay with improved performance. METHODS: We developed (pro)insulin antigens with alternative placements of a NanoLuc™ luciferase reporter (NLuc). Performance in LIPS was evaluated by testing sera from new onset T1D (n = 80), blood donors (n = 123), schoolchildren (n = 186), first-degree relatives (FDRs) from the Bart's Oxford family study (n = 53) and from the Belgian Diabetes Registry (n = 136), coded sera from the Islet Autoantibody Standardization Program (IASP) (T1D n = 50, blood donors n = 90). RESULTS: IAA LIPS based on B chain-NLuc proinsulin or B chain-NLuc insulin, in which NLuc was fused at the C-terminus of the insulin B chain, required only 2 µL of serum and a short incubation time, showed high concordance with RBA (Spearman r = 0.866 and 0.833, respectively), high assay performance (B chain-NLuc proinsulin ROC-AUC = 0.894 and B chain-NLuc insulin ROC-AUC = 0.916), and an adjusted sensitivity at 95% specificity ranking on par with the best assays submitted to the two most recent IASP workshops. In FDRs, the IAA LIPS showed improved discrimination of progressors to T1D compared to RBA. CONCLUSIONS: We established a novel high-performance non-radioactive IAA LIPS that might replace the current gold standard RBA and find wide application in the study of the IAA response in T1D.


Subject(s)
Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/diagnosis , Fluorescent Dyes/metabolism , Immunoprecipitation/methods , Insulin Antibodies/analysis , Luciferases/metabolism , Autoantibodies/blood , Biomarkers/analysis , Biomarkers/metabolism , Case-Control Studies , Child , Fluorescent Dyes/analysis , HEK293 Cells , Humans , Insulin/immunology , Insulin Antibodies/blood , Predictive Value of Tests , Prognosis , Proinsulin/immunology , Sensitivity and Specificity
15.
Clin Immunol ; 188: 23-30, 2018 03.
Article in English | MEDLINE | ID: mdl-29229565

ABSTRACT

The phenotype of autoreactive T cells in type 1 diabetes is described as Th1, Th17 and/or Th21, but is largely uncharacterized. We combined multi-parameter cytokine profiling and proliferation, and identified GM-CSF producing cells as a component of the response to beta cell autoantigens proinsulin and GAD65. Overall cytokine profiles of CD4+ T cell were not altered in type 1 diabetes. In contrast, patients with recent onset type 1 diabetes had increased frequencies of proinsulin-responsive CD4+CD45RA- T cells producing GM-CSF (p=0.002), IFNγ (p=0.004), IL-17A (p=0.008), IL-21 (p=0.011), and IL-22 (p=0.007), and GAD65-responsive CD4+CD45RA- T cells producing IL-21 (p=0.039). CD4+ T cells with a GM-CSF+IFNγ-IL-17A-IL-21-IL-22- phenotype were increased in patients for responses to both proinsulin (p=0.006) and GAD65 (p=0.037). GM-CSF producing T cells are a novel phenotype in the repertoire of T helper cells in type 1 diabetes and consolidate a Th1/Th17 pro-inflammatory pathogenesis in the disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Diabetes Mellitus, Type 1/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Autoantigens/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Type 1/metabolism , Gene Expression/immunology , Glutamate Decarboxylase , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Proinsulin/immunology , Proinsulin/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
16.
Cell Metab ; 26(4): 595-597, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978425

ABSTRACT

Antigen-specific immunotherapy has long been hailed as the ideal disease-modifying approach for type 1 diabetes, both for disease prevention and reversal. A small phase 1 trial now demonstrates safety of a peptide-based treatment in recently diagnosed adults.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Immunotherapy/methods , Proinsulin/therapeutic use , Adult , Autoimmunity , Clinical Trials, Phase I as Topic , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , HLA-DR4 Antigen/immunology , Humans , Immunotherapy/adverse effects , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Proinsulin/administration & dosage , Proinsulin/adverse effects , Proinsulin/immunology
17.
Cell Immunol ; 319: 43-52, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28864263

ABSTRACT

Cholera toxin B subunit fusion to autoantigens such as proinsulin (CTB-INS) down regulate dendritic cell (DC) activation and stimulate synthesis of DC immunosuppressive cytokines. Recent studies of CTB-INS induction of immune tolerance in human DCs indicate that increased biosynthesis of indoleamine 2,3-dioxygenase (IDO1) may play an important role in CTB-INS vaccine suppression of DC activation. Studies in murine models suggest a role for transforming growth factor beta (TGF-ß) in the stimulation of IDO1 biosynthesis, for the induction of tolerance in DCs. Here, we investigated the contribution of TGF-ß superfamily proteins to CTB-INS induction of IDO1 biosynthesis in human monocyte-derived DCs (moDCs). We show that CTB-INS upregulates the level of TGF-ß1, activin-A and the TGF-ß activator, integrin αvß8 in human DCs. However, inhibition of endogenous TGF-ß, activin-A or addition of biologically active TGF-ß1, and activin-A, did not inhibit or stimulate IDO1 biosynthesis in human DCs treated with CTB-INS. While inhibition with the kinase inhibitor, RepSox, blocked SMAD2/3 phosphorylation and diminished IDO1 biosynthesis in a concentration dependent manner. Specific blocking of the TGF-ß type 1 kinase receptor with SB-431542 did not arrest IDO1 biosynthesis, suggesting the involvement of a different kinase pathway other than TGF-ß type 1 receptor kinase in CTB-INS induction of IDO1 in human moDCs. Together, our experimental findings identify additional immunoregulatory proteins induced by the CTB-INS fusion protein, suggesting CTB-INS may utilize multiple mechanisms in the induction of tolerance in human moDCs.


Subject(s)
Dendritic Cells/drug effects , Gene Expression Regulation/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Recombinant Fusion Proteins/pharmacology , Transforming Growth Factor beta1/genetics , Activins/genetics , Activins/immunology , Animals , Cell Differentiation , Cholera Toxin/genetics , Cholera Toxin/immunology , Cloning, Molecular , Dendritic Cells/cytology , Dendritic Cells/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Integrins/genetics , Integrins/immunology , Mice , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Primary Cell Culture , Proinsulin/genetics , Proinsulin/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/immunology , Smad3 Protein/genetics , Smad3 Protein/immunology , Transforming Growth Factor beta1/immunology
18.
Eur J Immunol ; 47(9): 1550-1561, 2017 09.
Article in English | MEDLINE | ID: mdl-28665492

ABSTRACT

Type 1 diabetes (T1D) results from autoimmune destruction of insulin-producing pancreatic ß cells. Therapies need to incorporate strategies to overcome the genetic defects that impair induction or maintenance of peripheral T-cell tolerance and contribute to disease development. We tested whether the enforced expression of an islet autoantigen in antigen-presenting cells (APC) counteracted peripheral T-cell tolerance defects in autoimmune-prone NOD mice. We observed that insulin-specific CD8+ T cells transferred to mice in which proinsulin was transgenically expressed in APCs underwent several rounds of division and the majority were deleted. Residual insulin-specific CD8+ T cells were rendered unresponsive and this was associated with TCR downregulation, loss of tetramer binding and expression of a range of co-inhibitory molecules. Notably, accumulation and effector differentiation of insulin-specific CD8+ T cells in pancreatic lymph nodes was prominent in non-transgenic recipients but blocked by transgenic proinsulin expression. This shift from T-cell priming to T-cell tolerance exemplifies the tolerogenic capacity of autoantigen expression by APC and the capacity to overcome genetic tolerance defects.


Subject(s)
Antigen-Presenting Cells/immunology , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Proinsulin/immunology , Animals , Autoimmunity , Cells, Cultured , Humans , Immune Tolerance , Lymphocyte Activation , Male , Mice , Mice, Inbred NOD
19.
Immunol Cell Biol ; 95(9): 765-774, 2017 10.
Article in English | MEDLINE | ID: mdl-28611473

ABSTRACT

Type 1 diabetes (T1D) results from T-cell-mediated autoimmune destruction of pancreatic ß cells. Effector T-cell responses emerge early in disease development and expand as disease progresses. Following ß-cell destruction, a long-lived T-cell memory is generated that represents a barrier to islet transplantation and other cellular insulin-replacement therapies. Development of effective immunotherapies that control or ablate ß-cell destructive effector and memory T-cell responses has the potential to prevent disease progression and recurrence. Targeting antigen expression to antigen-presenting cells inactivates cognate CD8+ effector and memory T-cell responses and has therapeutic potential. Here we investigated this in the context of insulin-specific responses in the non-obese diabetic mouse where genetic immune tolerance defects could impact on therapeutic tolerance induction. Insulin-specific CD8+ memory T cells transferred to mice expressing proinsulin in antigen-presenting cells proliferated in response to transgenically expressed proinsulin and the majority were rapidly deleted. A small proportion of transferred insulin-specific Tmem remained undeleted and these were antigen-unresponsive, exhibited reduced T cell receptor (TCR) expression and H-2Kd/insB15-23 tetramer binding and expressed co-inhibitory molecules. Expression of proinsulin in antigen-presenting cells also abolished the diabetogenic capacity of CD8+ effector T cells. Therefore, destructive insulin-specific CD8+ T cells are effectively inactivated by enforced proinsulin expression despite tolerance defects that exist in diabetes-prone NOD mice. These findings have important implications in developing immunotherapeutic approaches to T1D and other T-cell-mediated autoimmune diseases.


Subject(s)
Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/physiology , Proinsulin/metabolism , Adoptive Transfer , Animals , Autoantigens/immunology , Cells, Cultured , Humans , Immune Tolerance , Immunologic Memory , Insulin/immunology , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Proinsulin/genetics , Proinsulin/immunology , Receptors, Antigen, T-Cell/metabolism
20.
PLoS One ; 12(5): e0176584, 2017.
Article in English | MEDLINE | ID: mdl-28472070

ABSTRACT

Latent Autoimmune Diabetes in Adults (LADA) is a slowly progressing form of immune-mediated diabetes that combines phenotypical features of type 2 diabetes (T2D) with the presence of islet cell antigens detected in type 1 diabetes (T1D). Heterogeneous clinical picture have led to the classification of patients based on the levels of antibodies against glutamic acid decarboxylase 65 (GADA) that correlate with clinical phenotypes closer to T1D or T2D when GADA titers are high or low, respectively. To date, LADA etiology remains elusive despite numerous studies investigating on genetic predisposition and environmental risk factors. To our knowledge, this is the first study aimed at evaluation of a putative role played by Mycobacterium avium subsp. paratuberculosis (MAP) as an infective agent in LADA pathogenesis. MAP is known to cause chronic enteritis in ruminants and has been associated with autoimmune disorders in humans. We analyzed seroreactivity of 223 Sardinian LADA subjects and 182 healthy volunteers against MAP-derived peptides and their human homologs of proinsulin and zinc transporter 8 protein. A significantly elevated positivity for MAP/proinsulin was detected among patients, with the highest prevalence in the 32-41-year-old T1D-like LADA subgroup, supporting our hypothesis of a possible MAP contribution in the development of autoimmunity.


Subject(s)
Bacterial Proteins/immunology , Latent Autoimmune Diabetes in Adults/immunology , Mycobacterium avium subsp. paratuberculosis/immunology , Peptides/immunology , Proinsulin/immunology , Adult , Aged , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL