Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1348131, 2024.
Article in English | MEDLINE | ID: mdl-38455055

ABSTRACT

There is accumulating evidence that pathogenic T cells in T1D recognize epitopes formed by post-translational modifications of ß-cell antigens, including hybrid insulin peptides (HIPs). The ligands for several CD4 T-cell clones derived from the NOD mouse are HIPs composed of a fragment of proinsulin joined to peptides from endogenous ß-cell granule proteins. The diabetogenic T-cell clone BDC-6.9 reacts to a fragment of C-peptide fused to a cleavage product of pro-islet amyloid polypeptide (6.9HIP). In this study, we used a monoclonal antibody (MAb) to the 6.9HIP to determine when and where HIP antigens are present in NOD islets during disease progression and with which immune cells they associate. Immunogold labeling of the 6.9HIP MAb and organelle-specific markers for electron microscopy were employed to map the subcellular compartment(s) in which the HIP is localized within ß-cells. While the insulin B9-23 peptide was present in nearly all islets, the 6.9HIP MAb stained infiltrated islets only in NOD mice at advanced stages of T1D development. Islets co-stained with the 6.9HIP MAb and antibodies to mark insulin, macrophages, and dendritic cells indicate that 6.9HIP co-localizes within insulin-positive ß-cells as well as intra-islet antigen-presenting cells (APCs). In electron micrographs, the 6.9HIP co-localized with granule structures containing insulin alone or both insulin and LAMP1 within ß-cells. Exposing NOD islets to the endoplasmic reticulum (ER) stress inducer tunicamycin significantly increased levels of 6.9HIP in subcellular fractions containing crinosomes and dense-core granules (DCGs). This work demonstrates that the 6.9HIP can be visualized in the infiltrated islets and suggests that intra-islet APCs may acquire and present HIP antigens within islets.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Islets of Langerhans , Animals , Mice , Mice, Inbred NOD , Peptides/metabolism , Insulin-Secreting Cells/metabolism , Antigens/metabolism
2.
Diabetes ; 73(5): 743-750, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38295386

ABSTRACT

Hybrid insulin peptides (HIPs) formed through covalent cross-linking of proinsulin fragments to secretory granule peptides are detectable within murine and human islets. The 2.5HIP (C-peptide-chromogranin A [CgA] HIP), recognized by the diabetogenic BDC-2.5 clone, is a major autoantigen in the nonobese diabetic mouse. However, the relevance of this epitope in human disease is currently unclear. A recent study probed T-cell reactivity toward HIPs in patients with type 1 diabetes, documenting responses in one-third of the patients and isolating several HIP-reactive T-cell clones. In this study, we isolated a novel T-cell clone and showed that it responds vigorously to the human equivalent of the 2.5HIP (designated HIP9). Although the responding patient carried the risk-associated DRB1*04:01/DQ8 haplotype, the response was restricted by DRB1*11:03 (DR11). HLA class II tetramer staining revealed higher frequencies of HIP9-reactive T cells in individuals with diabetes than in control participants. Furthermore, in DR11+ participants carrying the DRB4 allele, HIP9-reactive T-cell frequencies were higher than observed frequencies for the immunodominant proinsulin 9-28 epitope. Finally, there was a negative correlation between HIP9-reactive T-cell frequency and age at diagnosis. These results provide direct evidence that this C-peptide-CgA HIP is relevant in human type 1 diabetes and suggest a mechanism by which nonrisk HLA haplotypes may contribute to the development of ß-cell autoimmunity.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin , Humans , Animals , Mice , T-Lymphocytes , Proinsulin , C-Peptide , Chromogranin A , Peptides , Insulin, Regular, Human , Epitopes , Peptide Fragments
3.
Diagnostics (Basel) ; 13(17)2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37685398

ABSTRACT

Type 1 diabetes (T1D) is a chronic autoimmune disease that attacks the insulin-producing b cells of the pancreatic islets. Autoantibodies to b cell proteins typically appear in the circulation years before disease onset, and serve as the most accurate biomarkers of T1D risk. Our laboratory has recently discovered novel b cell proteins comprising hybrid proinsulin:islet amyloid polypeptide peptides (IAPP). T cells from a diabetic mouse model and T1D patients are activated by these hybrid peptides. In this study, we asked whether these hybrid molecules could serve as antigens for autoantibodies in T1D and prediabetic patients. We analyzed sera from T1D patients, prediabetics and healthy age-matched donors. Using a highly sensitive electrochemiluminescence assay, sera were screened for binding to recombinant proinsulin:IAPP probes or truncated derivatives. Our results show that sera from T1D patients contain antibodies that bind larger hybrid proinsulin:IAPP probes, but not proinsulin or insulin, at significantly increased frequencies compared to normal donors. Examination of sera from prediabetic patients confirms titers of antibodies to these hybrid probes in more than 80% of individuals, often before seroconversion. These results suggest that hybrid insulin peptides are common autoantigens in T1D and prediabetic patients, and that antibodies to these peptides may serve as valuable early biomarkers of the disease.

4.
J Biol Chem ; 299(11): 105264, 2023 11.
Article in English | MEDLINE | ID: mdl-37734557

ABSTRACT

Hybrid insulin peptides (HIPs) form in beta-cells when insulin fragments link to other peptides through a peptide bond. HIPs contain nongenomic amino acid sequences and have been identified as targets for autoreactive T cells in type 1 diabetes. A subgroup of HIPs, in which N-terminal amine groups of various peptides are linked to aspartic acid residues of insulin C-peptide, was detected through mass spectrometry in pancreatic islets. Here, we investigate a novel mechanism that leads to the formation of these HIPs in human and murine islets. Our research herein shows that these HIPs form spontaneously in beta-cells through a mechanism involving an aspartic anhydride intermediate. This mechanism leads to the formation of a regular HIP containing a standard peptide bond as well as a HIP-isomer containing an isopeptide bond by linkage to the carboxylic acid side chain of the aspartic acid residue. We used mass spectrometric analyses to confirm the presence of both HIP isomers in islets, thereby validating the occurrence of this novel reaction mechanism in beta-cells. The spontaneous formation of new peptide bonds within cells may lead to the development of neoepitopes that contribute to the pathogenesis of type 1 diabetes as well as other autoimmune diseases.


Subject(s)
Insulin-Secreting Cells , Insulin , Peptides , Animals , Humans , Mice , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Diabetes Mellitus, Type 1/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Peptides/analysis , Peptides/metabolism , In Vitro Techniques , Mass Spectrometry
5.
Diabetes ; 71(12): 2793-2803, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36041196

ABSTRACT

Hybrid insulin peptides (HIPs) form in pancreatic ß-cells through the formation of peptide bonds between proinsulin fragments and other peptides. HIPs have been identified in pancreatic islets by mass spectrometry and are targeted by CD4 T cells in patients with type 1 diabetes (T1D) as well as by pathogenic CD4 T-cell clones in nonobese diabetic (NOD) mice. The mechanism of HIP formation is currently poorly understood; however, it is well established that proteases can drive the formation of new peptide bonds in a side reaction during peptide bond hydrolysis. Here, we used a proteomic strategy on enriched insulin granules and identified cathepsin D (CatD) as the primary protease driving the specific formation of HIPs targeted by disease-relevant CD4 T cells in T1D. We also established that NOD islets deficient in cathepsin L (CatL), another protease implicated in the formation of disease-relevant HIPs, contain elevated levels of HIPs, indicating a role for CatL in the proteolytic degradation of HIPs. In summary, our data suggest that CatD may be a therapeutic target in efforts to prevent or slow the autoimmune destruction of ß-cells mediated by HIP-reactive CD4 T cells in T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Mice , Animals , Diabetes Mellitus, Type 1/metabolism , Insulin , Cathepsin D , Proteomics , Mice, Inbred NOD , Peptides , CD4-Positive T-Lymphocytes , Insulin, Regular, Human
6.
Front Immunol ; 13: 926650, 2022.
Article in English | MEDLINE | ID: mdl-36032090

ABSTRACT

Insulin is considered to be a key antigenic target of T cells in Type 1 Diabetes (T1D) and autoimmune diabetes in the NOD mouse with particular focus on the B-chain amino acid sequence B:9-23 as the primary epitope. Our lab previously discovered that hybrid insulin peptides (HIPs), comprised of insulin C-peptide fragments fused to other ß-cell granule peptides, are ligands for several pathogenic CD4 T cell clones derived from NOD mice and for autoreactive CD4 T cells from T1D patients. A subset of CD4 T cell clones from our panel react to insulin and B:9-23 but only at high concentrations of antigen. We hypothesized that HIPs might also be formed from insulin B-chain sequences covalently bound to other endogenously cleaved ß-cell proteins. We report here on the identification of a B-chain HIP, termed the 6.3HIP, containing a fragment of B:9-23 joined to an endogenously processed peptide of ProSAAS, as a strong neo-epitope for the insulin-reactive CD4 T cell clone BDC-6.3. Using an I-Ag7 tetramer loaded with the 6.3HIP, we demonstrate that T cells reactive to this B-chain HIP can be readily detected in NOD mouse islet infiltrates. This work suggests that some portion of autoreactive T cells stimulated by insulin B:9-23 may be responding to B-chain HIPs as peptide ligands.


Subject(s)
Diabetes Mellitus, Type 1 , Animals , CD4-Positive T-Lymphocytes , Epitopes , Mice , Mice, Inbred NOD , Peptide Fragments , Peptides
7.
Diabetes ; 71(3): 483-496, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35007324

ABSTRACT

The induction of antigen (Ag)-specific tolerance and replacement of islet ß-cells are major ongoing goals for the treatment of type 1 diabetes (T1D). Our group previously showed that a hybrid insulin peptide (2.5HIP) is a critical autoantigen for diabetogenic CD4+ T cells in the NOD mouse model. In this study, we investigated whether induction of Ag-specific tolerance using 2.5HIP-coupled tolerogenic nanoparticles (NPs) could protect diabetic NOD mice from disease recurrence upon syngeneic islet transplantation. Islet graft survival was significantly prolonged in mice treated with 2.5HIP NPs, but not NPs containing the insulin B chain peptide 9-23. Protection in 2.5HIP NP-treated mice was attributed both to the simultaneous induction of anergy in 2.5HIP-specific effector T cells and the expansion of Foxp3+ regulatory T cells specific for the same Ag. Notably, our results indicate that effector function of graft-infiltrating CD4+ and CD8+ T cells specific for other ß-cell epitopes was significantly impaired, suggesting a novel mechanism of therapeutically induced linked suppression. This work establishes that tolerance induction with an HIP can delay recurrent autoimmunity in NOD mice, which could inform the development of an Ag-specific therapy for T1D.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Graft Survival/drug effects , Insulin/administration & dosage , Islets of Langerhans Transplantation/methods , Peptide Fragments/administration & dosage , Animals , Autoantigens/immunology , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/prevention & control , Female , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Nanoparticles/administration & dosage , Recurrence
8.
Cells ; 10(12)2021 12 07.
Article in English | MEDLINE | ID: mdl-34943952

ABSTRACT

We have shown that PLG nanoparticles loaded with peptide antigen can reduce disease in animal models of autoimmunity and in a phase 1/2a clinical trial in celiac patients. Clarifying the mechanisms by which antigen-loaded nanoparticles establish tolerance is key to further adapting them to clinical use. The mechanisms underlying tolerance induction include the expansion of antigen-specific CD4+ regulatory T cells and sequestration of autoreactive cells in the spleen. In this study, we employed nanoparticles loaded with two model peptides, GP33-41 (a CD8 T cell epitope derived from lymphocytic choriomeningitis virus) and OVA323-339 (a CD4 T cell epitope derived from ovalbumin), to modulate the CD8+ and CD4+ T cells from two transgenic mouse strains, P14 and DO11.10, respectively. Firstly, it was found that the injection of P14 mice with particles bearing the MHC I-restricted GP33-41 peptide resulted in the expansion of CD8+ T cells with a regulatory cell phenotype. This correlated with reduced CD4+ T cell viability in ex vivo co-cultures. Secondly, both nanoparticle types were able to sequester transgenic T cells in secondary lymphoid tissue. Flow cytometric analyses showed a reduction in the surface expression of chemokine receptors. Such an effect was more prominently observed in the CD4+ cells rather than the CD8+ cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Celiac Disease/therapy , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/immunology , Antigens/pharmacology , Antigens, Viral/immunology , Antigens, Viral/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Celiac Disease/genetics , Celiac Disease/immunology , Cell Lineage/drug effects , Cell Lineage/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/pharmacology , Glycoproteins/immunology , Glycoproteins/pharmacology , Humans , Immune Tolerance/drug effects , Mice , Mice, Transgenic , Nanoparticles/chemistry , Ovalbumin/immunology , Ovalbumin/pharmacology , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Peptides/immunology , Peptides/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , T-Lymphocytes, Regulatory/drug effects , Viral Proteins/immunology , Viral Proteins/pharmacology
9.
Diabetes ; 70(12): 2860-2870, 2021 12.
Article in English | MEDLINE | ID: mdl-34497137

ABSTRACT

Recognition of ß-cell antigens by autoreactive T cells is a critical step in the initiation of autoimmune type1 diabetes. A complete protection from diabetes development in NOD mice harboring a point mutation in the insulin B-chain 9-23 epitope points to a dominant role of insulin in diabetogenesis. Generation of NOD mice lacking the chromogranin A protein (NOD.ChgA-/-) completely nullified the autoreactivity of the BDC2.5 T cell and conferred protection from diabetes onset. These results raised the issue concerning the dominant antigen that drives the autoimmune process. Here we revisited the NOD.ChgA-/- mice and found that their lack of diabetes development may not be solely explained by the absence of chromogranin A reactivity. NOD.ChgA-/- mice displayed reduced presentation of insulin peptides in the islets and periphery, which corresponded to impaired T-cell priming. Diabetes development in these mice was restored by antibody treatment targeting regulatory T cells or inhibiting transforming growth factor-ß and programmed death-1 pathways. Therefore, the global deficiency of chromogranin A impairs recognition of the major diabetogenic antigen insulin, leading to broadly impaired autoimmune responses controlled by multiple regulatory mechanisms.


Subject(s)
Autoimmunity/genetics , Chromogranin A/genetics , Diabetes Mellitus, Type 1/genetics , Animals , Antigen Presentation/genetics , Autoantigens/immunology , Autoantigens/metabolism , Cytoprotection/genetics , Cytoprotection/immunology , Diabetes Mellitus, Type 1/prevention & control , Epitopes, T-Lymphocyte/immunology , Female , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout
10.
Front Immunol ; 12: 668680, 2021.
Article in English | MEDLINE | ID: mdl-34113344

ABSTRACT

Hybrid Insulin Peptides (HIPs), which consist of insulin fragments fused to other peptides from ß-cell secretory granule proteins, are CD4 T cell autoantigens in type 1 diabetes (T1D). We have studied HIPs and HIP-reactive CD4 T cells extensively in the context of the non-obese diabetic (NOD) mouse model of autoimmune diabetes and have shown that CD4 T cells specific for HIPs are major contributors to disease pathogenesis. Additionally, in the human context, HIP-reactive CD4 T cells can be found in the islets and peripheral blood of T1D patients. Here, we performed an in-depth characterization of the CD4 T cell response to a C-peptide/C-peptide HIP (HIP11) in human T1D. We identified the TCR expressed by the previously-reported HIP11-reactive CD4 T cell clone E2, which was isolated from the peripheral blood of a T1D patient, and determined that it recognizes HIP11 in the context of HLA-DQ2. We also identified a HIP11-specific TCR directly in the islets of a T1D donor and demonstrated that this TCR recognizes a different minimal epitope of HIP11 presented by HLA-DQ8. We generated and tested an HLA-DQ2 tetramer loaded with HIP11 that will enable direct ex vivo interrogation of CD4 T cell responses to HIP11 in human patients and control subjects. Using mass spectrometric analysis, we confirmed that HIP11 is present in human islets. This work represents an important step in characterizing the role of CD4 T cell responses to HIPs in human T1D.


Subject(s)
Autoantigens/immunology , C-Peptide/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin/immunology , Islets of Langerhans/immunology , Receptors, Antigen, T-Cell/immunology , Autoantigens/metabolism , C-Peptide/metabolism , CD4-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/blood , Epitopes , Female , HLA-DQ Antigens/immunology , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , K562 Cells , Male , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
11.
Biomedicines ; 9(3)2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33673706

ABSTRACT

Autoreactive T cells are thought to orchestrate the onset and progression of autoimmune diabetes. Key cognate antigens of these diabetogenic T cells include hybrid insulin peptides, formed by the fusion of insulin fragments to cleavage products of other ß-cell granule proteins. Here we review initial work exploring tolerance induction to a hybrid insulin peptide using a biodegradable, nanoparticle delivery system in non-obese diabetic (NOD) mice. The immune phenotype(s) and possible mechanism(s) behind antigen-specific tolerance induction were dissected with a disease transfer model using transgenic autoreactive mouse T cells. Treatment of NOD mice with peptide-coupled nanoparticles appeared to have a dual function in preventing diabetes onset, inducing anergy in effector T cells and enhancing the activity of regulatory T cells. Importantly, the ratio of these two cell types in the pancreas was pushed toward tolerance. Antigen-specific tolerance induction to hybrid insulin peptides has the translational potential to preserve islet ß-cells in new-onset or at-risk patients and prevent recurrent autoimmunity in transplant patients.

12.
Immunol Lett ; 223: 62-70, 2020 07.
Article in English | MEDLINE | ID: mdl-32335144

ABSTRACT

Endogenous retrovirus (ERV) are remnants of ancient retroviruses that have been incorporated into the genome and evidence suggests that they may play a role in the etiology of T1D. We previously identified a murine leukemia retrovirus-like ERV whose Env and Gag antigens are involved in autoimmune responses in non-obese diabetic (NOD) mice. In this study, we show that the Gag antigen is present in the islet stromal cells. Although Gag gene transcripts were present, Gag protein was not detected in diabetes-resistant mice. Cloning and sequencing analysis of individual Gag genes revealed that NOD islets express Gag gene variants with complete open-reading frames (ORFs), in contrast to the diabetes-resistant mice, whose islet Gag gene transcripts are mostly non-ORFs. Importantly, the ORFs obtained from the NOD islets are extremely heterogenous, coding for various mutants that are absence in the genome. We further show that Gag antigens are stimulatory for autoreactive T cells and identified one islet-expressing Gag variant that contains an altered peptide ligand capable of inducing IFN-gamma release by the T cells. The data highlight a unique retrovirus-like factor in the islets of the NOD mouse strain, which may participate in key events triggering autoimmunity and T1D.


Subject(s)
Autoantigens/metabolism , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Endogenous Retroviruses/physiology , Gene Products, gag/metabolism , Islets of Langerhans/metabolism , T-Lymphocytes/immunology , Animals , Autoantigens/immunology , Cell-Derived Microparticles/metabolism , Cells, Cultured , Gene Products, gag/immunology , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred NOD
13.
Diabetes ; 69(7): 1492-1502, 2020 07.
Article in English | MEDLINE | ID: mdl-32291282

ABSTRACT

T cells isolated from the pancreatic infiltrates of nonobese diabetic mice have been shown to recognize epitopes formed by the covalent cross-linking of proinsulin and secretory granule peptides. Formation of such hybrid insulin peptides (HIPs) was confirmed through mass spectrometry, and responses to HIPs were observed among the islet-infiltrating T cells of pancreatic organ donors and in the peripheral blood of individuals with type 1 diabetes (T1D). However, questions remain about the prevalence of HIP-specific T cells in humans, the sequences they recognize, and their role in disease. We identified six novel HIPs that are recognized in the context of DRB1*04:01, discovered by using a library of theoretical HIP sequences derived from insulin fragments covalently linked to one another or to fragments of secretory granule proteins or other islet-derived proteins. We demonstrate that T cells that recognize these HIPs are detectable in the peripheral blood of subjects with T1D and exhibit an effector memory phenotype. HIP-reactive T-cell clones produced Th1-associated cytokines and proliferated in response to human islet preparations. These results support the relevance of HIPs in human disease, further establishing a novel posttranslational modification that may contribute to the loss of peripheral tolerance in T1D.


Subject(s)
HLA-DRB1 Chains/immunology , Insulin/immunology , Peptide Fragments/immunology , T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cross Reactions , Diabetes Mellitus, Type 1/immunology , Epitopes , Humans , Insulin/chemistry , Insulin-Secreting Cells/immunology , Peptide Fragments/chemistry
14.
Crit Rev Immunol ; 40(5): 441-446, 2020.
Article in English | MEDLINE | ID: mdl-33463955

ABSTRACT

Our investigations of antigens for pathogenic T cells in autoimmune diabetes led to the discovery of hybrid insulin peptides as T cell epitopes. T cells reactive to hybrid insulin peptides can be found at high frequency in the nonobese diabetic mouse model of type 1 diabetes and are also present in human patients. Hybrid insulin peptides can also be administered to mice in a tolerogenic form, thereby suppressing the autoimmune response.


Subject(s)
Diabetes Mellitus, Type 1 , Animals , Cell Differentiation , Epitopes, T-Lymphocyte , Humans , Mice , Mice, Inbred NOD , T-Lymphocytes
15.
Nat Metab ; 1(5): 509-518, 2019 05.
Article in English | MEDLINE | ID: mdl-31423480

ABSTRACT

Type 1 diabetes (T1D) is characterized by pancreatic islet infiltration by autoreactive immune cells and a near-total loss of ß-cells1. Restoration of insulin-producing ß-cells coupled with immunomodulation to suppress the autoimmune attack has emerged as a potential approach to counter T1D2-4. Here we report that enhancing ß-cell mass early in life, in two models of female NOD mice, results in immunomodulation of T-cells, reduced islet infiltration and lower ß-cell apoptosis, that together protect them from developing T1D. The animals displayed altered ß-cell antigens, and islet transplantation studies showed prolonged graft survival in the NOD-LIRKO model. Adoptive transfer of splenocytes from the NOD-LIRKOs prevented development of diabetes in pre-diabetic NOD mice. A significant increase in the splenic CD4+CD25+FoxP3+ regulatory T-cell (Treg) population was observed to underlie the protected phenotype since Treg depletion rendered NOD-LIRKO mice diabetic. The increase in Tregs coupled with activation of TGF-ß/SMAD3 signaling pathway in pathogenic T-cells favored reduced ability to kill ß-cells. These data support a previously unidentified observation that initiating ß-cell proliferation, alone, prior to islet infiltration by immune cells alters the identity of ß-cells, decreases pathologic self-reactivity of effector cells and increases Tregs to prevent progression of T1D.


Subject(s)
Cell Proliferation , Diabetes Mellitus, Type 1/pathology , Immune System/immunology , Insulin-Secreting Cells/pathology , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Disease Progression , Humans , Mice
16.
Diabetes ; 68(9): 1830-1840, 2019 09.
Article in English | MEDLINE | ID: mdl-31175101

ABSTRACT

We recently established that hybrid insulin peptides (HIPs) are present in human islets and that T cells reactive to HIPs are found in the residual islets of organ donors with type 1 diabetes (T1D). Here, we investigate whether HIP-reactive T cells are indicative of ongoing autoimmunity in patients with T1D. We used interferon-γ enzyme-linked immune absorbent spot analyses on peripheral blood mononuclear cells (PBMCs) to determine whether patients with new-onset T1D or control subjects displayed T-cell reactivity to a panel of 16 HIPs. We observed that nearly one-half of the patients responded to one or more HIPs. Responses to four HIPs were significantly elevated in patients with T1D but not in control subjects. To characterize the T cells reactive to HIPs, we used a carboxyfluorescein succinimidyl ester-based assay to clone T cells from PBMCs. We isolated six nonredundant, antigen-specific T-cell clones, most of which reacting to their target HIPs in the low nanomolar range. One T-cell clone was isolated from the same patient on two different blood draws, indicating persistence of this T-cell clone in the peripheral blood. This work suggests that HIPs are important target antigens in human subjects with T1D and may play a critical role in disease.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Insulin/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Autoimmunity/immunology , Child , Female , Humans , Islets of Langerhans/immunology , Leukocytes, Mononuclear/immunology , Male , Young Adult
17.
Curr Opin Endocrinol Diabetes Obes ; 26(4): 195-200, 2019 08.
Article in English | MEDLINE | ID: mdl-31166225

ABSTRACT

PURPOSE OF REVIEW: The current review covers recent advances in our knowledge of the newest autoantigen neo-epitopes in type 1 diabetes (T1D): hybrid insulin peptides or HIPs. These ligands for autoreactive T cells are formed by peptide fusion, a novel posttranslational modification process that we first reported in 2016. RECENT FINDINGS: Two major HIPs in the nonobese diabetic mouse model, ligands for diabetogenic CD4 T-cell clones, have been incorporated into tetramers and used to track HIP-reactive T cells during progression of disease. HIPs have also been used in strategies for induction of antigen-specific tolerance and show promise for delaying or reversing disease in the nonobese diabetic mouse. Importantly, CD4 T cells reactive to various HIPs have been detected in the islets and peripheral blood mononuclear cell of T1D patients and newly developed human T-cell clones are being employed to gather more data on the phenotype and function of HIP-reactive T cells in patients. SUMMARY: These new hybrid insulin peptide epitopes may provide the basis for establishing autoreactive T cells as biomarkers of disease and as potential tolerogens for treatment of T1D.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/immunology , Insulin/immunology , Animals , Humans , Mice , Peptides/immunology
18.
J Immunol ; 203(1): 48-57, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31109955

ABSTRACT

CD4 T cells play a critical role in promoting the development of autoimmunity in type 1 diabetes. The diabetogenic CD4 T cell clone BDC-2.5, originally isolated from a NOD mouse, has been widely used to study the contribution of autoreactive CD4 T cells and relevant Ags to autoimmune diabetes. Recent work from our laboratory has shown that the Ag for BDC-2.5 T cells is a hybrid insulin peptide (2.5HIP) consisting of an insulin C-peptide fragment fused to a peptide from chromogranin A (ChgA) and that endogenous 2.5HIP-reactive T cells are major contributors to autoimmune pathology in NOD mice. The objective of this study was to determine if poly(lactide-co-glycolide) (PLG) nanoparticles (NPs) loaded with the 2.5HIP Ag (2.5HIP-coupled PLG NPs) can tolerize BDC-2.5 T cells. Infusion of 2.5HIP-coupled PLG NPs was found to prevent diabetes in an adoptive transfer model by impairing the ability of BDC-2.5 T cells to produce proinflammatory cytokines through induction of anergy, leading to an increase in the ratio of Foxp3+ regulatory T cells to IFN-γ+ effector T cells. To our knowledge, this work is the first to use a hybrid insulin peptide, or any neoepitope, to re-educate diabetogenic T cells and may have significant implications for the development of an Ag-specific therapy for type 1 diabetes patients.


Subject(s)
Chromogranin A/metabolism , Diabetes Mellitus, Type 1/therapy , Immunotherapy/methods , Insulin/metabolism , Nanoparticles/therapeutic use , Peptides/metabolism , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Animals, Genetically Modified , Cells, Cultured , Chromogranin A/genetics , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Insulin/genetics , Interferon-gamma/metabolism , Mice , Mice, Inbred NOD , Nanoparticles/metabolism , Peptides/genetics , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics
19.
Front Immunol ; 10: 99, 2019.
Article in English | MEDLINE | ID: mdl-30766536

ABSTRACT

Type 1 diabetes (T1D) is a T cell mediated autoimmune disease that affects more than 19 million people with incidence increasing rapidly worldwide. For T cells to effectively drive T1D, they must first traffic to the islets and extravasate through the islet vasculature. Understanding the cues that lead to T cell entry into inflamed islets is important because diagnosed T1D patients already have established immune infiltration of their islets. Here we show that CD11c+ cells are a key mediator of T cell trafficking to infiltrated islets in non-obese diabetic (NOD) mice. Using intravital 2-photon islet imaging we show that T cell extravasation into the islets is an extended process, with T cells arresting in the islet vasculature in close proximity to perivascular CD11c+ cells. Antigen is not required for T cell trafficking to infiltrated islets, but T cell chemokine receptor signaling is necessary. Using RNAseq, we show that islet CD11c+ cells express over 20 different chemokines that bind chemokine receptors expressed on islet T cells. One highly expressed chemokine-receptor pair is CXCL16-CXCR6. However, NOD. CXCR6-/- mice progressed normally to T1D and CXCR6 deficient T cells trafficked normally to the islets. Even with CXCR3 and CXCR6 dual deficiency, T cells trafficked to infiltrated islets. These data reinforce that chemokine receptor signaling is highly redundant for T cell trafficking to inflamed islets. Importantly, depletion of CD11c+ cells strongly inhibited T cell trafficking to infiltrated islets of NOD mice. We suggest that targeted depletion of CD11c+ cells associated with the islet vasculature may yield a therapeutic target to inhibit T cell trafficking to inflamed islets to prevent progression of T1D.


Subject(s)
CD11c Antigen/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , T-Lymphocytes/immunology , Animals , Female , Mice, Inbred NOD , Mice, Knockout
20.
J Proteome Res ; 18(3): 814-825, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30585061

ABSTRACT

We recently discovered hybrid insulin peptides (HIPs) as a novel class of post-translationally modified peptides in murine-derived beta cell tumors, and we demonstrated that these molecules are autoantigens in type 1 diabetes (T1D). A HIP consists of an insulin fragment linked to another secretory granule peptide via a peptide bond. We verified that autoreactive CD4 T cells in both mouse and human autoimmune diabetes recognize these modified peptides. Here, we use mass spectrometric analyses to confirm the presence of HIPs in both mouse and human pancreatic islets. We also present criteria for the confident identification of these peptides. This work supports the hypothesis that HIPs are autoantigens in human T1D and provides a foundation for future efforts to interrogate this previously unknown component of the beta cell proteome.


Subject(s)
Autoantigens/analysis , Insulin/chemistry , Islets of Langerhans/chemistry , Mass Spectrometry/methods , Animals , Autoantigens/blood , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Humans , Mice , Peptides/analysis , Peptides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...