Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Nature ; 602(7895): 156-161, 2022 02.
Article in English | MEDLINE | ID: mdl-34847567

ABSTRACT

CD8 T cell-mediated autoimmune diseases result from the breakdown of self-tolerance mechanisms in autoreactive CD8 T cells1. How autoimmune T cell populations arise and are sustained, and the molecular programmes defining the autoimmune T cell state, are unknown. In type 1 diabetes, ß-cell-specific CD8 T cells destroy insulin-producing ß-cells. Here we followed the fate of ß-cell-specific CD8 T cells in non-obese diabetic mice throughout the course of type 1 diabetes. We identified a stem-like autoimmune progenitor population in the pancreatic draining lymph node (pLN), which self-renews and gives rise to pLN autoimmune mediators. pLN autoimmune mediators migrate to the pancreas, where they differentiate further and destroy ß-cells. Whereas transplantation of as few as 20 autoimmune progenitors induced type 1 diabetes, as many as 100,000 pancreatic autoimmune mediators did not. Pancreatic autoimmune mediators are short-lived, and stem-like autoimmune progenitors must continuously seed the pancreas to sustain ß-cell destruction. Single-cell RNA sequencing and clonal analysis revealed that autoimmune CD8 T cells represent unique T cell differentiation states and identified features driving the transition from autoimmune progenitor to autoimmune mediator. Strategies aimed at targeting the stem-like autoimmune progenitor pool could emerge as novel and powerful immunotherapeutic interventions for type 1 diabetes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/immunology , Stem Cells/pathology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Self Renewal , Clone Cells/immunology , Clone Cells/metabolism , Clone Cells/pathology , Disease Models, Animal , Female , Glucose-6-Phosphatase/immunology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Insulin-Secreting Cells/pathology , Lymph Nodes/immunology , Male , Mice , Receptors, Antigen, T-Cell/metabolism , Single-Cell Analysis , Stem Cell Transplantation , Stem Cells/immunology , Stem Cells/metabolism , Transcriptome
2.
Clin Immunol ; 233: 108893, 2021 12.
Article in English | MEDLINE | ID: mdl-34808330

ABSTRACT

The role of cellular autoimmunity in the pathogenesis of fulminant type 1 diabetes (FT1D) remains largely unknown. In this study, we performed an integrated assay using peripheral blood mononuclear cells to determine the islet antigen-specific CD8+ T cell responses in FT1D and compare the responses among acute-onset T1D (AT1D) and slowly progressive T1D (SP1D). IGRP- and ZnT8-specific IL-6, G-CSF, and TNF-α responses were significantly upregulated in patients with FT1D, while IGRP- and ZnT8-specific IP-10 responses were significantly upregulated in patients with AT1D than in non-diabetics (ND). Furthermore, the frequencies of IGRP-specific type 1 CD8+ cytotoxic T (Tc1) cells were significantly higher in the FT1D group than in the ND, SP1D, and AT1D groups. Additionally, IGRP-specific Tc1 cells were more abundant in the FT1D with HLA-A2 group than in the FT1D without A2 group. In conclusion, our study suggests that IGRP-specific CD8+ T cells significantly contribute to the pathogenesis of FT1D.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Glucose-6-Phosphatase/immunology , Adult , Female , Humans , Male , Middle Aged
3.
Diabetes ; 67(5): 936-945, 2018 05.
Article in English | MEDLINE | ID: mdl-29506985

ABSTRACT

Stem memory T cells (Tscm) constitute the earliest developmental stage of memory T cells, displaying stem cell-like properties, such as self-renewal capacity. Their superior immune reconstitution potential has sparked interest in cancer immune therapy, vaccine development, and immune reconstitution, whereas their role in autoimmunity is largely unexplored. Here we show that autoreactive CD8+ Tscm specific for ß-cell antigens GAD65, insulin, and IGRP are present in patients with type 1 diabetes (T1D). In vitro, the generation of autoreactive Tscm from naive precursors required the presence of the homeostatic cytokine interleukin-7 (IL-7). IL-7 promotes glucose uptake via overexpression of GLUT1 and upregulation of the glycolytic enzyme hexokinase 2. Even though metabolism depends on glucose uptake, the subsequent oxidation of pyruvate in the mitochondria was necessary for Tscm generation from naive precursors. In patients with T1D, high expression of GLUT1 was a hallmark of circulating Tscm, and targeting glucose uptake via GLUT1 using the selective inhibitor WZB117 resulted in inhibition of Tscm generation and expansion. Our results suggest that autoreactive Tscm are present in patients with T1D and can be selectively targeted by inhibition of glucose metabolism.


Subject(s)
Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Lymphoid Progenitor Cells/immunology , T-Lymphocyte Subsets/immunology , Adolescent , Adult , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Child , Diabetes Mellitus, Type 1/metabolism , Female , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Glucose-6-Phosphatase/immunology , Glutamate Decarboxylase/immunology , Hexokinase/metabolism , Humans , Hydroxybenzoates/pharmacology , Immunologic Memory/immunology , In Vitro Techniques , Insulin/immunology , Interleukin-7/immunology , Lymphopoiesis/drug effects , Male , Middle Aged , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism , Up-Regulation
4.
J Autoimmun ; 89: 112-124, 2018 05.
Article in English | MEDLINE | ID: mdl-29258717

ABSTRACT

Type 1 diabetes (T1D) is mediated by destruction of pancreatic ß cells by autoantigen-specific CD4+ and CD8+ T cells, thus the ideal solution for T1D is the restoration of immune tolerance to ß cell antigens. We demonstrate the ability of carboxylated 500 nm biodegradable poly(lactide-co-glycolide) (PLG) nanoparticles PLG nanoparticles (either surface coupled with or encapsulating the cognate diabetogenic peptides) to rapidly and efficiently restore tolerance in NOD.SCID recipients of both activated diabetogenic CD4+ BDC2.5 chromagranin A-specific and CD8+ NY8.3 islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-specific TCR transgenic T cells in an antigen-specific manner. Further, initiation and maintenance of Ag-PLG tolerance operates via several overlapping, but independent, pathways including regulation via negative-co-stimulatory molecules (CTLA-4 and PD-1) and the systemic induction of peptide-specific Tregs which were critical for long-term maintenance of tolerance by controlling both trafficking of effector T cells to, and their release of pro-inflammatory cytokines within the pancreas, concomitant with selective retention of effector cells in the spleens of recipient mice.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/pathology , Nanoparticles/therapeutic use , Animals , Autoantigens/chemistry , Autoantigens/immunology , Cells, Cultured , Diabetes Mellitus, Type 1/therapy , Disease Models, Animal , Female , Glucose-6-Phosphatase/chemistry , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/immunology , Immune Tolerance , Mice , Mice, Inbred NOD , Mice, Transgenic , Nanoparticles/chemistry , Peptides/chemistry , Peptides/immunology , Polylactic Acid-Polyglycolic Acid Copolymer , Receptors, Antigen, T-Cell, alpha-beta/genetics
5.
Cell ; 171(3): 655-667.e17, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29053971

ABSTRACT

The gut microbiota contributes to the development of normal immunity but, when dysregulated, can promote autoimmunity through various non-antigen-specific effects on pathogenic and regulatory lymphocytes. Here, we show that an integrase expressed by several species of the gut microbial genus Bacteroides encodes a low-avidity mimotope of the pancreatic ß cell autoantigen islet-specific glucose-6-phosphatase-catalytic-subunit-related protein (IGRP206-214). Studies in germ-free mice monocolonized with integrase-competent, integrase-deficient, and integrase-transgenic Bacteroides demonstrate that the microbial epitope promotes the recruitment of diabetogenic CD8+ T cells to the gut. There, these effectors suppress colitis by targeting microbial antigen-loaded, antigen-presenting cells in an integrin ß7-, perforin-, and major histocompatibility complex class I-dependent manner. Like their murine counterparts, human peripheral blood T cells also recognize Bacteroides integrase. These data suggest that gut microbial antigen-specific cytotoxic T cells may have therapeutic value in inflammatory bowel disease and unearth molecular mimicry as a novel mechanism by which the gut microbiota can regulate normal immune homeostasis. PAPERCLIP.


Subject(s)
Autoantigens/immunology , Bacteroides/immunology , Colitis/immunology , Gastrointestinal Microbiome , Glucose-6-Phosphatase/immunology , Adult , Animals , Bacteroides/classification , Bacteroides/enzymology , Colitis/microbiology , Female , Glucose-6-Phosphatase/genetics , Humans , Lymphoid Tissue/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Middle Aged , Molecular Mimicry , T-Lymphocytes/immunology
6.
Clin Immunol ; 161(2): 270-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26341315

ABSTRACT

The breakdown of immune tolerance against islet antigens causes type 1 diabetes (T1D). The antigens associated with adult-onset T1D (AT1D) remain largely undefined. It is possible that AT1D patients display a unique type of CD4(+) T cells specific for a certain islet antigen. Here we analyzed the cytokine production profiles of CD4(+) helper T (Th) cells that are specific for three islet antigens; GAD65, preproinsulin, and IGRP in patients with AT1D, juvenile-onset T1D (JT1D), and age-, gender- and human leukocyte antigen (HLA)-matched control adults. While IGRP-specific Th cells in AT1D patients were dominantly Th1 cells, IGRP-specific Th cells in control adults and JT1D patients were dominantly Th2 and T regulatory type 1 (Tr1) cells. Notably, the frequency of IGRP-specific Tr1 cells was significantly lower in AT1D patients than in control adults and JT1D patients. In conclusion, our study suggests that IGRP-specific Th cells play a unique pathogenic role in AT1D.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Glucose-6-Phosphatase/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Age of Onset , Amino Acid Sequence , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Diabetes Mellitus, Type 1/blood , Female , Glucose-6-Phosphatase/metabolism , Glutamate Decarboxylase/immunology , Glutamate Decarboxylase/metabolism , Humans , Insulin/immunology , Insulin/metabolism , Islets of Langerhans/immunology , Male , Middle Aged , Molecular Sequence Data , Protein Precursors/immunology , Protein Precursors/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism
7.
Immunol Cell Biol ; 93(6): 548-57, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25601275

ABSTRACT

Cross-presentation by CD8(+) conventional dendritic cells (cDCs) is involved in the maintenance of peripheral tolerance and this process is termed cross-tolerance. Previous reports showed that non-obese diabetic (NOD) mice have reduced number of splenic CD8(+) cDCs compared with non-diabetic strains, and that the administration of Flt3L to enhance DC development resulted in reduced diabetes incidence. As CD8(+) cDCs are the most efficient antigen cross-presenting cells, it was assumed that reduced cross-presentation by non-activated, tolerogenic CD8(+) cDC predisposes to autoimmune diabetogenesis. Here we show for the first time that indeed NOD mice have a defect in autoantigen cross-presentation capacity. First, we showed that NOD CD8(+) cDCs were less sensitive to iatrogenic cytochrome c, which had previously been shown to selectively deplete CD8(+) cDCs that functionally cross-present. Second, we found that proliferation of islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-specific CD8(+) T cells was impaired in NOD compared with non-obese diabetes resistant mice after immunization with cell associated recombinant fusion protein containing the cognate IGRP peptide. This study, therefore, suggests that the reduced number of CD8(+) cDCs in NOD mice, coupled with the reduced capacity to cross-present self-antigens, reduces the overall capacity to maintain peripheral tolerance in the spontaneous autoimmune type 1 diabetes mice.


Subject(s)
Antigen Presentation/immunology , Cross-Priming/immunology , Animals , Autoantigens/immunology , CD8 Antigens/metabolism , Cell Count , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/immunology , Glucose-6-Phosphatase/chemistry , Glucose-6-Phosphatase/immunology , Histocompatibility Antigens Class I/immunology , Immunophenotyping , Mice , Mice, Inbred NOD , Peptides/immunology , Spleen/cytology , Spleen/immunology
8.
Clin Exp Immunol ; 179(3): 398-413, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25302633

ABSTRACT

Several ß cell antigens recognized by T cells in the non-obese diabetic (NOD) mouse model of type 1 diabetes (T1D) are also T cell targets in the human disease. While numerous antigen-specific therapies prevent diabetes in NOD mice, successful translation of rodent findings to patients has been difficult. A human leucocyte antigen (HLA)-transgenic mouse model incorporating human ß cell-specific T cells might provide a better platform for evaluating antigen-specific therapies. The ability to study such T cells is limited by their low frequency in peripheral blood and the difficulty in obtaining islet-infiltrating T cells from patients. We have worked to overcome this limitation by using lentiviral transduction to 'reprogram' primary human CD8 T cells to express three T cell receptors (TCRs) specific for a peptide derived from the ß cell antigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP265-273 ) and recognized in the context of the human class I major histocompatibility complex (MHC) molecule HLA-A2. The TCRs bound peptide/MHC multimers with a range of avidities, but all bound with at least 10-fold lower avidity than the anti-viral TCR used for comparison. One exhibited antigenic recognition promiscuity. The ß cell-specific human CD8 T cells generated by lentiviral transduction with one of the TCRs released interferon (IFN)-γ in response to antigen and exhibited cytotoxic activity against peptide-pulsed target cells. The cells engrafted in HLA-A2-transgenic NOD-scid IL2rγ(null) mice and could be detected in the blood, spleen and pancreas up to 5 weeks post-transfer, suggesting the utility of this approach for the evaluation of T cell-modulatory therapies for T1D and other T cell-mediated autoimmune diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Genetic Vectors/genetics , Immunotherapy, Adoptive/methods , Insulin-Secreting Cells/immunology , Lentivirus/genetics , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Survival , Glucose-6-Phosphatase/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Jurkat Cells , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin-2/genetics , T-Lymphocytes, Cytotoxic/transplantation
9.
J Immunol ; 193(7): 3296-307, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25165150

ABSTRACT

We investigated whether a prevalent epitope of the ß-cell-specific autoantigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP206-214) reaches regional Ag-presentation pathways via unprocessed polypeptide chains, as free IGRP206-214 peptide or via preformed IGRP206-214/K(d) complexes. This was accomplished by expressing bacterial artificial chromosome transgenes encoding wild-type (stable) or ubiquitinated (unstable) forms of IGRP in IGRP-deficient NOD mice carrying MHC class I-deficient ß-cells, dendritic cells, or B cells. We investigated the ability of the pancreatic lymph nodes of these mice to prime naive IGRP206-214-reactive CD8(+) T cells in vivo, either in response to spontaneous Ag shedding, or to synchronized forms of ß-cell necrosis or apoptosis. When IGRP was made unstable by targeting it for proteasomal degradation within ß-cells, the cross-priming, autoimmune-initiating potential of this autoantigen (designated autoantigenicity) was impaired. Yet at the same time, the direct presentation, CTL-targeting potential of IGRP (designated pathogenicity) was enhanced. The appearance of IGRP206-214 in regional Ag-presentation pathways was dissociated from transfer of IGRP206-214 or IGRP206-214/K(d) from ß cells to dendritic cells. These results indicate that autoantigenicity and pathogenicity are separable and inversely related properties and suggest that pathogenic autoantigens, capable of efficiently priming CTLs while marking target cells for CTL-induced killing, may have a critical balance of these two properties.


Subject(s)
Antigen Presentation , Autoantigens/immunology , Cross-Priming , Dendritic Cells/immunology , Glucose-6-Phosphatase/immunology , Insulin-Secreting Cells/immunology , Animals , Apoptosis/immunology , Autoantigens/genetics , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Glucose-6-Phosphatase/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Necrosis
10.
Immunol Cell Biol ; 92(7): 640-4, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24777314

ABSTRACT

CD8(+) T cells are prominent in autoimmune diabetes of both humans and non-obese diabetic (NOD) mice. For example, CD8(+) T cells against islet-specific glucose 6-phosphatase catalytic subunit-related protein (IGRP) can be detected readily in older NOD mice. It has been suggested that the enumeration of islet-specific CD8(+) T cells in the peripheral blood may be a predictive biomarker for autoimmune type 1 diabetes (T1D). Here, we determined the natural history of the functional endogenous IGRP(206-214)-specific cytotoxic T lymphocytes (CTLs) in NOD mice with regard to age (3- to 15-week-old pre-diabetic mice and diabetic mice) and sex. We demonstrated that in vivo IGRP(206-214)-specific CTLs significantly increased after 12 weeks of age and in vivo cytotoxicity in female NOD mice was significantly higher than in male NOD mice. To determine the in vivo IGRP(206-214)-specific CTL frequency without killing the mice, we performed splenectomies on a cohort of mice after injecting IGRP(206-214)-coated targets and then followed their diabetes progression. We found that CTL frequency correlated with future of disease onset. Thus, our data support that IGRP(206-214)-specific CTLs may be a potent biomarker for T1D.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Epitopes, T-Lymphocyte/immunology , Glucose-6-Phosphatase/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Age Factors , Animals , Cytotoxicity, Immunologic , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/chemistry , Female , Glucose-6-Phosphatase/chemistry , Male , Mice , Mice, Inbred NOD , Peptides/chemistry , Prognosis
11.
Clin Exp Immunol ; 176(2): 199-206, 2014 May.
Article in English | MEDLINE | ID: mdl-24387268

ABSTRACT

Autoimmune diabetes is characterized by autoantigen-specific T cell-mediated destruction of pancreatic islet beta cells, and CD8(+) T cells are key players during this process. We assessed whether the bitransgenic RIP-CD80 x RIP-LCMV-GP (RIP-CD80GP) mice may be a versatile antigen-specific model of inducible CD8(+) T cell-mediated autoimmune diabetes. Antigen-encoding DNA, peptide-loaded dendritic cells and antigen plus incomplete Freund's adjuvant were used for vaccination. Of 14 pancreatic proteins tested by DNA vaccination, murine pre-proinsulin 2 (100% of mice; median time after vaccination, 60 days) and islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) (77%, 58 days) could induce diabetes. Vaccination with DNA encoding for zinc transporter 8, Ia-2, Ia-2ß, glutamic acid decarboxylase 67 (Gad67), chromogranin A, insulinoma amyloid polypeptide and homeobox protein Nkx-2.2 induced diabetes development in 25-33% of mice. Vaccination with DNA encoding for Gad65, secretogranin 5, pancreas/duodenum homeobox protein 1 (Pdx1), carboxyl ester lipase, glucagon and control hepatitis B surface antigen (HBsAg) induced diabetes in <20% of mice. Diabetes induction efficiency could be increased by DNA vaccination with a vector encoding a ubiquitin-antigen fusion construct. Diabetic mice had florid T cell islet infiltration. CD8(+) T cell targets of IGRP were identified with a peptide library-based enzyme-linked immunospot assay, and diabetes could also be induced by vaccination with major histocompatibility complex (MHC) class I-restricted IGRP peptides loaded on mature dendritic cells. Vaccination with antigen plus incomplete Freund's adjuvant, which can prevent diabetes in other models, led to rapid diabetes development in the RIP-CD80GP mouse. We conclude that RIP-CD80GP mice are a versatile model of antigen specific autoimmune diabetes and may complement existing mouse models of autoimmune diabetes for evaluating CD8(+) T cell-targeted prevention strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Glucose-6-Phosphatase/immunology , Insulin/immunology , Protein Precursors/immunology , Vaccination/methods , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , CD8-Positive T-Lymphocytes/metabolism , DNA/genetics , DNA/immunology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/genetics , Freund's Adjuvant/immunology , Glucose-6-Phosphatase/genetics , Glycoproteins/genetics , Glycoproteins/immunology , Insulin/genetics , Islets of Langerhans/immunology , Kaplan-Meier Estimate , Lipids/immunology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Protein Precursors/genetics , Rats , Time Factors , Vaccination/adverse effects , Vaccines, DNA/immunology , Viral Proteins/genetics , Viral Proteins/immunology
12.
Eur J Immunol ; 44(4): 983-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24435807

ABSTRACT

Targeting the BAFF/APRIL system has shown to be effective in preventing T-cell dependent autoimmune disease in the NOD mouse, a spontaneous model of type 1 diabetes. In this study we generated BAFF-deficient NOD mice to examine how BAFF availability would influence T-cell responses in vivo and the development of spontaneous diabetes. BAFF-deficient NOD mice which lack mature B cells, were protected from diabetes and showed delayed rejection of an allogeneic islet graft. Diabetes protection correlated with a failure to expand pathogenic IGRP-reactive CD8(+) T cells, which were maintained in the periphery at correspondingly low levels. Adoptive transfer of IGRP-reactive CD8(+) T cells with B cells into BAFF-deficient NOD mice enhanced IGRP-reactive CD8(+) T-cell expansion. Furthermore, when provoked with cyclophosphamide, or transferred to a secondary lymphopenic host, the latent pool of self-reactive T cells resident in BAFF-deficient NOD mice could elicit beta cell destruction. We conclude that lack of BAFF prevents the procurement of B-cell-dependent help necessary for the emergence of destructive diabetes. Indeed, treatment of NOD mice with the BAFF-blocking compound, BR3-Fc, resulted in a delayed onset and reduced incidence of diabetes.


Subject(s)
Autoimmunity/immunology , B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Autoimmunity/genetics , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Female , Flow Cytometry , Glucose-6-Phosphatase/immunology , Glucose-6-Phosphatase/metabolism , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/genetics , Graft Survival/immunology , Immunophenotyping , Islets of Langerhans Transplantation/methods , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , T-Lymphocytes/metabolism , Time Factors
13.
J Immunol ; 192(2): 572-80, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24337380

ABSTRACT

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


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Immunologic Memory/immunology , Islets of Langerhans/immunology , Animals , Autoantigens/immunology , Diabetes Mellitus, Type 1/pathology , Glucose-6-Phosphatase/immunology , Islets of Langerhans/pathology , Lymphocytes/immunology , Mice , Mice, Inbred NOD
14.
PLoS One ; 8(10): e77296, 2013.
Article in English | MEDLINE | ID: mdl-24204793

ABSTRACT

In the present study, we investigated the therapeutic potential of a selective S1P1 receptor modulator, ponesimod, to protect and reverse autoimmune diabetes in non-obese diabetic (NOD) mice. Ponesimod was administered orally to NOD mice starting at 6, 10, 13 and 16 weeks of age up to 35 weeks of age or to NOD mice showing recent onset diabetes. Peripheral blood and spleen B and T cell counts were significantly reduced after ponesimod administration. In pancreatic lymph nodes, B lymphocytes were increased and expressed a transitional 1-like phenotype. Chronic oral ponesimod treatment efficiently prevented autoimmune diabetes in 6, 10 and 16 week-old pre-diabetic NOD mice. Treatment withdrawal led to synchronized disease relapse. Ponesimod did not inhibit the differentiation of autoreactive T cells as assessed by adoptive transfer of lymphocytes from treated disease-free NOD mice. In addition, it did not affect the migration, proliferation and activation of transgenic BDC2.5 cells into the target tissue. However, ponesimod inhibited spreading of the T cell responses to islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP). Treatment of diabetic NOD mice with ponesimod induced disease remission. However, here again, upon treatment cessation, the disease rapidly recurred. This recurrence was effectively prevented by combination treatment with a CD3 antibody leading to the restoration of self-tolerance. In conclusion, treatment with a selective S1P1 modulator in combination with CD3 antibody represents a promising therapeutic approach for the treatment of autoimmune diabetes.


Subject(s)
Antibodies/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/pharmacology , Receptors, Lysosphingolipid/immunology , Thiazoles/pharmacology , Administration, Oral , Adoptive Transfer , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD3 Complex/genetics , CD3 Complex/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Drug Administration Schedule , Gene Expression , Glucose-6-Phosphatase/antagonists & inhibitors , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Count , Mice , Mice, Inbred NOD , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/genetics , Recurrence , Self Tolerance , Spleen/drug effects , Spleen/immunology , Spleen/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
15.
Curr Opin Allergy Clin Immunol ; 13(6): 596-606, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24145314

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to summarize pathogenic mechanisms and clinical implications of the most illustrative genetic entities of congenital neutropenia syndromes. RECENT FINDINGS: Congenital neutropenia comprise monogenetic entities with or without additional immunologic and extrahaematopoietic syndromatic features. Continuous careful explorations of known entities such as ELANE, GFI1, HAX1, G6PC3 deficiency and XLN help to define principles controlling differentiation and function of neutrophil granulocytes. Furthermore, the identification of novel genetic defects associated with congenital neutropenia, such as VPS45 deficiency, broadens our understanding of neutrophil biology. Pathogenic mechanisms imply protein and vesicle mistrafficking, endoplasmic reticulum stress, the unfolded protein response, destabilization of the mitochondrial membrane potential, disturbed energy metabolism, dysglycosylation and deregulated actin polymerization. SUMMARY: Advanced genetic and biochemical techniques have helped to expand our knowledge of congenital neutropenia syndromes. Known and novel genetic entities shed light on fundamental biological processes important for the homeostatis and functioning not only of the neutrophil granulocyte but as well of the entire haematopoietic system. Furthermore, treatment decisions become more tailored and might pave the road towards personalized molecular medicine.


Subject(s)
Neutropenia/congenital , Neutrophils , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Biological Transport, Active/genetics , Biological Transport, Active/immunology , Congenital Bone Marrow Failure Syndromes , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/immunology , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/immunology , Glucose-6-Phosphatase/metabolism , Humans , Membrane Potential, Mitochondrial/genetics , Membrane Potential, Mitochondrial/immunology , Neutropenia/genetics , Neutropenia/immunology , Neutropenia/metabolism , Neutropenia/pathology , Neutropenia/physiopathology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism , Unfolded Protein Response/genetics , Unfolded Protein Response/immunology , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/immunology , Vesicular Transport Proteins/metabolism
16.
Diabetologia ; 56(12): 2651-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24030068

ABSTRACT

AIMS/HYPOTHESIS: Thymic expression of self-antigens during T-lymphocyte development is believed to be crucial for preventing autoimmunity. It has been suggested that G6PC2, the gene encoding islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP), is differentially spliced between pancreatic beta cells and the thymus. This may contribute to incomplete elimination of IGRP-specific T lymphocytes in the thymus, predisposing individuals to type 1 diabetes. We tested whether specific splice variation in islets vs thymus correlates with loss of tolerance to IGRP in type 1 diabetes. METHODS: Expression of G6PC2 splice variants was compared among thymus, purified medullary thymic epithelial cells and pancreatic islets by RT-PCR. Differential immunogenicity of IGRP splice variants was tested in patients and healthy individuals for autoantibodies and specific cytotoxic T lymphocytes using radiobinding assays and HLA class I multimers, respectively. RESULTS: Previously reported G6PC2 splice variants, including full-length G6PC2, were confirmed, albeit that they occurred in both pancreas and thymus, rather than islets alone. Yet, their expression levels were profoundly greater in islets than in thymus. Moreover, three novel G6PC2 variants were discovered that occur in islets only, leading to protein truncations, frame shifts and neo-sequences prone to immunogenicity. However, autoantibodies to novel or known IGRP splice variants did not differ between patients and healthy individuals, and similar frequencies of IGRP-specific cytotoxic T lymphocytes could be detected in both patients with type 1 diabetes and healthy individuals. CONCLUSIONS/INTERPRETATION: We propose that post-transcriptional variation of tissue-specific self-proteins may affect negative thymic selection, although this need not necessarily lead to disease.


Subject(s)
Alternative Splicing , Diabetes Mellitus, Type 1/immunology , Glucose-6-Phosphatase/immunology , Islets of Langerhans/immunology , Pancreas/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Antibody Formation/genetics , Antibody Formation/immunology , Autoantigens/immunology , Autoantigens/metabolism , Base Sequence , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/genetics , Female , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , Glucose-6-Phosphatase/genetics , Humans , Islets of Langerhans/metabolism , Male , Pancreas/enzymology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Thymus Gland/enzymology , Transcription, Genetic
17.
J Immunol ; 190(7): 3109-20, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23427248

ABSTRACT

In the NOD mouse model of type 1 diabetes, insulin-dependent diabetes (Idd) loci control the development of insulitis and diabetes. Independently, protective alleles of Idd3/Il2 or Idd5 are able to partially protect congenic NOD mice from insulitis and diabetes, and to partially tolerize islet-specific CD8(+) T cells. However, when the two regions are combined, mice are almost completely protected, strongly suggesting the existence of genetic interactions between the two loci. Idd5 contains at least three protective subregions/causative gene candidates, Idd5.1/Ctla4, Idd5.2/Slc11a1, and Idd5.3/Acadl, yet it is unknown which of them interacts with Idd3/Il2. Through the use of a series of novel congenic strains containing the Idd3/Il2 region and different combinations of Idd5 subregion(s), we defined these genetic interactions. The combination of Idd3/Il2 and Idd5.3/Acadl was able to provide nearly complete protection from type 1 diabetes, but all three Idd5 subregions were required to protect from insulitis and fully restore self-tolerance. By backcrossing a Slc11a1 knockout allele onto the NOD genetic background, we have demonstrated that Slc11a1 is responsible for the diabetes protection resulting from Idd5.2. We also used Slc11a1 knockout-SCID and Idd5.2-SCID mice to show that both loss-of-function alleles provide protection from insulitis when expressed on the SCID host alone. These results lend further support to the hypothesis that Slc11a1 is Idd5.2.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Epistasis, Genetic , Quantitative Trait Loci , Alleles , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Diabetes Mellitus, Type 1/immunology , Female , Genetic Predisposition to Disease , Glucose-6-Phosphatase/immunology , Immune Tolerance/genetics , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Proteins/immunology
18.
Immunol Res ; 55(1-3): 91-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22983906

ABSTRACT

G6PC2, also known as islet-specific glucose 6-phosphatase catalytic subunit-related protein (IGRP), is a major target of autoreactive CD8(+) T cells in both diabetic human subjects and the non-obese diabetic (NOD) mouse. However, in contrast to the abundant literature regarding the CD8(+) response to this antigen, much less is known about the potential involvement of IGRP-reactive CD4(+) T cells in diabetogenesis. The single previous study that examined this question in NOD mice was based upon a candidate epitope approach and identified three I-A(g7)-restricted epitopes that each elicited spontaneous responses in these animals. However, given the known inaccuracies of MHC class II epitope prediction algorithms, we hypothesized that additional specificities might also be targeted. To address this issue, we immunized NOD mice with membranes from insect cells overexpressing full-length recombinant mouse IGRP and measured recall responses of purified CD4(+) T cells using a library of overlapping peptides encompassing the entire 355-aa primary sequence. Nine peptides representing 8 epitopes gave recall responses, only 1 of which corresponded to any of the previously reported sequences. In each case proliferation was blocked by a monoclonal antibody to I-A(g7), but not the appropriate isotype control. Consistent with a role in diabetogenesis, proliferative responses to 4 of the 9 peptides (3 epitopes) were also detected in CD4(+) T cells purified from the pancreatic draining lymph nodes of pre-diabetic female animals, but not from peripheral lymph nodes or spleens of the same animals. Intriguingly, one of the newly identified spontaneously reactive epitopes (P8 [IGRP(55-72)]) is highly conserved between mice and man, suggesting that it might also be a target of HLA-DQ8-restricted T cells in diabetic human subjects, an hypothesis that we are currently testing.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Glucose-6-Phosphatase/immunology , Histocompatibility Antigens Class II/immunology , Proteins/immunology , Animals , Drosophila , Epitopes/immunology , Female , Lymph Nodes/immunology , Male , Mice , Mice, Inbred NOD , Peptides/immunology
19.
J Clin Immunol ; 33(3): 520-5, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23180359

ABSTRACT

PURPOSE: G6PC3 deficiency presents as a complex and heterogeneous syndrome that classically associates severe congenital neutropenia with cardiac and urogenital developmental defects. Here we investigate the findings of T cell lymphopenia and inflammatory bowel disease in a child with G6PC3 deficiency due to compound heterozygous mutations in intron 3 (c.IVS3-1 G>A) and exon 6 (c.G778G/C; p.Gly260/Arg). METHODS: Histological examination was conducted on all biopsy specimens. Immunophenotyping and lymphocyte proliferation assays were performed. Immunoglobulin levels and vaccine responses were measured. RESULTS: The patient showed persistent global T cell lymphopenia, with only 8 to 13 % of thymic naive CD31(+)CD45RA(+) cells among CD4 T cells (normal range 27-60 %). Proliferation assays and vaccine responses were within normal limits. The gastrointestinal inflammatory lesions were very closely related to those of glycogen storage disease type 1b, with a Crohn's-like appearance but without granuloma or increased cryptic abscesses. The gastrointestinal disease responded to infliximab therapy. These findings were associated with a polyclonal hypergammaglobuliemia G. CONCLUSION: G6PC3 deficiency may present with inflammatory bowel disease and T cell lymphopenia. The diagnosis should thus be considered in a patient with chronic congenital neutropenia and gastrointestinal symptoms. Patients with confirmed disease should also undergo T cell phenotyping to rule out cellular immunodeficiency.


Subject(s)
Glucose-6-Phosphatase/genetics , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/genetics , Lymphopenia/complications , Lymphopenia/genetics , Adolescent , Child , Gastric Mucosa/pathology , Glucose-6-Phosphatase/immunology , Humans , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/pathology , Lymphocyte Count , Lymphopenia/immunology , Mutation
20.
J Autoimmun ; 39(4): 347-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22647732

ABSTRACT

Type 1 diabetes (T1D) is mediated by destruction of pancreatic ß-cells by CD4 and CD8 T cells specific for epitopes on numerous diabetogenic autoantigens resulting in loss of glucose homeostasis. Employing antigen-specific tolerance induced by i.v. administration of syngeneic splenocytes ECDI cross-linked to various diabetogenic antigens/epitopes (Ag-SP), we show that epitope spreading plays a functional role in the pathogenesis of T1D in NOD mice. Specifically, Ag-SP coupled with intact insulin, Ins B(9-23) or Ins B(15-23), but not GAD65(509-528), GAD65(524-543) or IGRP(206-214), protected 4-6 week old NOD mice from the eventual development of clinical disease; infiltration of immune cells to the pancreatic islets; and blocked the induction of DTH responses in a Treg-dependent, antigen-specific manner. However, tolerance induction in 19-21 week old NOD mice was effectively accomplished only by Ins-SP, suggesting Ins B(9-23) is a dominant initiating epitope, but autoimmune responses to insulin epitope(s) distinct from Ins B(9-23) emerge during disease progression.


Subject(s)
Autoantigens/immunology , Autoimmunity , Diabetes Mellitus, Type 1/immunology , Epitopes/immunology , Insulin-Secreting Cells/immunology , Insulin/immunology , Peptide Fragments/immunology , Animals , Autoantigens/genetics , Cell Movement , Cross-Linking Reagents/chemistry , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Disease Progression , Epitopes/genetics , Female , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/immunology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/immunology , Immune Tolerance , Injections, Intravenous , Insulin/genetics , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred NOD , Peptide Fragments/genetics , Proteins/genetics , Proteins/immunology , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...