Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Neuroimmunol ; 327: 1-9, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30685070

ABSTRACT

Type 1 Diabetes (T1D) is an autoimmune disease in which insulin producing beta cells of the pancreas are selectively destroyed. Glial Fibrillary Acidic Protein (GFAP) expressed in peri-islet Schwann cells (pSCs) and in the ductal cells of the pancreas is one of the candidate autoantigens for T1D. Immune responses to GFAP expressing cell types precede the islet autoimmunity in Non-Obese Diabetic (NOD) mice. By removing MHC class I from GFAP expressing cell types, we tested the role of autoantigens presented by these cell types in the development of invasive insulitis. Our findings indicate that antigens expressed by pancreatic ductal cells are important in the development of invasive insulitis in NOD mice.


Subject(s)
Autoantigens/immunology , Autoimmunity/immunology , Islets of Langerhans/immunology , Animals , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Mice, Knockout
2.
Islets ; 10(1): 10-24, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29157116

ABSTRACT

The contribution of environmental factors to pancreatic islet damage in type 1 diabetes remains poorly understood. In this study, we crossed mice susceptible to type 1 diabetes, where parental male (CD8+ T cells specific for IGRP206-214; NOD8.3) and female (NOD/ShiLt) mice were randomized to a diet either low or high in AGE content and maintained on this diet throughout pregnancy and lactation. After weaning, NOD8.3+ female offspring were identified and maintained on the same parental feeding regimen for until day 28 of life. A low AGE diet, from conception to early postnatal life, decreased circulating AGE concentrations in the female offspring when compared to a high AGE diet. Insulin, proinsulin and glucagon secretion were greater in islets isolated from offspring in the low AGE diet group, which was akin to age matched non-diabetic C57BL/6 mice. Pancreatic islet expression of Ins2 gene was also higher in offspring from the low AGE diet group. Islet expression of glucagon, AGEs and the AGE receptor RAGE, were each reduced in low AGE fed offspring. Islet immune cell infiltration was also decreased in offspring exposed to a low AGE diet. Within pancreatic lymph nodes and spleen, the proportions of CD4+ and CD8+ T cells did not differ between groups. There were no significant changes in body weight, fasting glucose or glycemic hormones. This study demonstrates that reducing exposure to dietary AGEs throughout gestation, lactation and early postnatal life may benefit pancreatic islet secretion and immune infiltration in the type 1 diabetic susceptible mouse strain, NOD8.3.


Subject(s)
Diet , Glycation End Products, Advanced/adverse effects , Islets of Langerhans/drug effects , Lactation , Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects , Animals , Animals, Newborn , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/physiopathology , Female , Glycation End Products, Advanced/administration & dosage , Islets of Langerhans/physiopathology , Lactation/drug effects , Lactation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/physiopathology
3.
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
4.
Front Immunol ; 8: 115, 2017.
Article in English | MEDLINE | ID: mdl-28261200

ABSTRACT

Many pathogens, including the malaria parasite Plasmodium falciparum, display high levels of polymorphism within T-cell epitope regions of proteins associated with protective immunity. The T-cell epitope variants are often non-cross-reactive. Herein, we show in a murine model, which modifies a protective CD8 T-cell epitope from the circumsporozoite protein (CS) of Plasmodium berghei (SYIPSAEKI), that simultaneous or sequential co-stimulation with two of its putative similarly non-cross-reactive altered peptide ligand (APL) epitopes (SYIPSAEDI or SYIPSAEAI) has radically different effects on immunity. Hence, co-immunization or sequential stimulation in vivo of SYIPSAEKI with its APL antagonist SYIPSAEDI decreases immunity to both epitopes. By contrast, co-immunization with SYIPSAEAI has no apparent initial effect, but it renders the immune response to SYIPSAEKI resistant to being turned off by subsequent immunization with SYIPSAEDI. These results suggest a novel strategy for vaccines that target polymorphic epitopes potentially capable of mutual immune interference in the field, by initiating an immune response by co-immunization with the desired index epitope, together with a carefully selected "potentiator" APL peptide.

5.
J Autoimmun ; 70: 52-62, 2016 06.
Article in English | MEDLINE | ID: mdl-27068879

ABSTRACT

The properties of CD4(+) regulatory T cell (Treg) subsets are dictated by distinct patterns of gene expression determined by FOXP3 and different combinations of various transcription factors. Here we show the NF-κB transcription factor RelA is constitutively active in naïve and effector Tregs. The conditional inactivation of Rela in murine FOXP3(+) cells induces a rapid onset, multi-focal autoimmune disease that depends on RelA being expressed in conventional T cells. In addition to promoting Treg lineage stability, RelA determines the size of the effector Treg population, a function influenced by the presence or absence of RelA in conventional T cells. These findings showing that RelA controls Treg stability and promotes the competitive fitness of effector Tregs highlight the importance of RelA activity in peripheral Treg induced tolerance.


Subject(s)
Immune Tolerance , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factor RelA/metabolism , Animals , Antibodies/blood , Antibodies/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Autoimmunity , Biomarkers , Cluster Analysis , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Immune Tolerance/genetics , Immunomodulation , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Count , Male , Mice , Mice, Transgenic , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcription Factor RelA/genetics
6.
Immunol Cell Biol ; 94(4): 334-41, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26446877

ABSTRACT

In type 1 diabetes, cytotoxic CD8(+) T lymphocytes (CTLs) directly interact with pancreatic beta cells through major histocompatibility complex class I. An immune synapse facilitates delivery of cytotoxic granules, comprised mainly of granzymes and perforin. Perforin deficiency protects the majority of non-obese diabetic (NOD) mice from autoimmune diabetes. Intriguingly perforin deficiency does not prevent diabetes in CD8(+) T-cell receptor transgenic NOD8.3 mice. We therefore investigated the importance of perforin-dependent killing via CTL-beta cell contact in autoimmune diabetes. Perforin-deficient CTL from NOD mice or from NOD8.3 mice were significantly less efficient at adoptive transfer of autoimmune diabetes into NODRag1(-/-) mice, confirming that perforin is essential to facilitate beta cell destruction. However, increasing the number of transferred in vitro-activated perforin-deficient 8.3 T cells reversed the phenotype and resulted in diabetes. Perforin-deficient NOD8.3 T cells were present in increased proportion in islets, and proliferated more in response to antigen in vivo indicating that perforin may regulate the activation of CTLs, possibly by controlling cytokine production. This was confirmed when we examined the requirement for direct interaction between beta cells and CD8(+) T cells in NOD8.3 mice, in which beta cells specifically lack major histocompatibility complex (MHC) class I through conditional deletion of ß2-microglobulin. Although diabetes was significantly reduced, 40% of these mice developed diabetes, indicating that NOD8.3 T cells can kill beta cells in the absence of direct interaction. Our data indicate that although perforin delivery is the main mechanism that CTL use to destroy beta cells, they can employ alternative mechanisms to induce diabetes in a perforin-independent manner.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Perforin/metabolism , Animals , Autoantigens/immunology , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Disease Models, Animal , Humans , Lymphocyte Activation/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Paracrine Communication , Perforin/genetics , Perforin/immunology
7.
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
8.
Clin Sci (Lond) ; 126(1): 1-18, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24020444

ABSTRACT

T1D (Type 1 diabetes) is an autoimmune disease caused by the immune-mediated destruction of pancreatic ß-cells. Studies in T1D patients have been limited by the availability of pancreatic samples, a protracted pre-diabetic phase and limitations in markers that reflect ß-cell mass and function. The NOD (non-obese diabetic) mouse is currently the best available animal model of T1D, since it develops disease spontaneously and shares many genetic and immunopathogenic features with human T1D. Consequently, the NOD mouse has been extensively studied and has made a tremendous contribution to our understanding of human T1D. The present review summarizes the key lessons from NOD mouse studies concerning the genetic susceptibility, aetiology and immunopathogenic mechanisms that contribute to autoimmune destruction of ß-cells. Finally, we summarize the potential and limitations of immunotherapeutic strategies, successful in NOD mice, now being trialled in T1D patients and individuals at risk of developing T1D.


Subject(s)
Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 1/etiology , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Genetic Predisposition to Disease , Humans , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Mice , Mice, Inbred NOD
9.
Diabetologia ; 56(12): 2659-68, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24013782

ABSTRACT

AIMS/HYPOTHESIS: Type 1 diabetes is characterised by early peri-islet insulitis and insulin autoantibodies, followed by invasive insulitis and beta cell destruction. The immunological events that precipitate invasive insulitis are not well understood. We tested the hypothesis that B cells in diabetes-prone NOD mice drive invasive insulitis through elevated expression of CD19 and consequent enhanced uptake and presentation of beta cell membrane-bound antigens to islet invasive T cells. METHODS: CD19 expression and signalling pathways in B cells from NOD and control mice were compared. Expansion of CD8(+) T cells specific for insulin and islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) were compared in CD19-deficient and wild-type NOD mice and this was correlated with insulitis severity. The therapeutic potential of anti-CD19 treatment during the period of T cell activation was assessed for its ability to block invasive insulitis. RESULTS: CD19 expression and signalling in B cells was increased in NOD mice. CD19 deficiency significantly diminished the expansion of CD8(+) T cells with specificity for the membrane-bound beta cell antigen, IGRP. Conversely the reduction in CD8(+) T cells with specificity for the soluble beta cell antigen, insulin, was relatively small and not significant. CONCLUSIONS/INTERPRETATION: Elevated CD19 on NOD B cells promotes presentation of the membrane-bound antigen, IGRP, mediating the expansion of autoreactive T cells specific for antigens integral to beta cells, which are critical for invasive insulitis and diabetes. Downregulating the CD19 signalling pathway in insulin autoantibody-positive individuals before the development of type 1 diabetes may prevent expansion of islet-invasive T cells and preserve beta cell mass.


Subject(s)
Antigens, CD19/metabolism , Diabetes Mellitus, Type 1/immunology , Inflammation/immunology , Islets of Langerhans/immunology , Lymphocyte Activation/immunology , Prediabetic State/immunology , Signal Transduction , Animals , Autoantibodies/immunology , Blotting, Western , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Disease Progression , Female , Flow Cytometry , Inflammation/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred NOD , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology
10.
J Immunol ; 191(1): 70-82, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23740949

ABSTRACT

Several mechanisms have been proposed to explain how certain MHC class II molecules afford dominant resistance to autoimmune diseases like type 1 diabetes (T1D). However, it remains unclear how protective MHC types can blunt autoreactive T cell responses directed against a diverse repertoire of autoantigenic epitopes presented by disease-promoting MHCs. In this study, we show that expression of I-E on dendritic cells (DCs) of NOD mice promotes the differentiation of MHC promiscuous autoreactive CD4(+) clonotypes into antidiabetogenic autoregulatory T cells. We expressed an I-Eα(kloxP) transgene in NOD mice and used cell type-specific I-E ablation to show that I-E-expressing DCs, but not B cells, promote the generation of autoreactive CD4(+)Foxp3(+) regulatory T cells (Tregs) and their accumulation in the pancreas-draining lymph nodes. There, these Tregs suppress the presentation of ß cell Ags to naive autoreactive CD4(+) and CD8(+) T cells restricted by diabetogenic MHC molecules in an I-E-independent manner. Whereas selective removal of I-E on DCs abrogated autoregulatory Treg formation and T1D protection, selective removal of I-E on B cells was inconsequential. These results explain how certain MHC class II molecules can completely suppress antigenically complex autoimmune responses in an Ag-nonspecific manner.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/prevention & control , Genes, MHC Class II/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Differentiation/genetics , Dendritic Cells/pathology , Diabetes Mellitus, Experimental/pathology , Genes, MHC Class II/genetics , Integrases/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , T-Lymphocytes, Regulatory/pathology
11.
Blood ; 121(22): 4555-66, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23550035

ABSTRACT

Thrombosis promotes leukocyte infiltration into inflamed tissues, leading to organ injury in a broad range of diseases; however, the mechanisms by which thrombi guide leukocytes to sites of vascular injury remain ill-defined. Using mouse models of endothelial injury (traumatic or ischemia reperfusion), we demonstrate a distinct process of leukocyte recruitment, termed "directed intravascular migration," specifically mediated by platelet thrombi. Single adherent platelets and platelet aggregates stimulated leukocyte shape change at sites of endothelial injury; however, only thrombi were capable of inducing directed intravascular leukocyte migration. Leukocyte recruitment and migration induced by platelet thrombi occurred most prominently in veins but could also occur in arteries following ischemia-reperfusion injury. In vitro studies demonstrated a major role for platelet-derived NAP-2 (CXCL-7) and its CXCR1/2 receptor in regulating leukocyte polarization and motility. In vivo studies demonstrated the presence of an NAP-2 chemotactic gradient within the thrombus body. Pharmacologic blockade of CXCR1/2 as well as genetic deletion of NAP-2 markedly reduced leukocyte shape change and intrathrombus migration. These studies define a distinct process of leukocyte migration that is initiated by homotypic adhesive interactions between platelets, leading to the development of an NAP-2 chemotactic gradient within the thrombus body that guides leukocytes to sites of vascular injury.


Subject(s)
Blood Platelets/cytology , Chemokines, CXC/metabolism , Leukocytes/cytology , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Thrombosis/immunology , Animals , Blood Platelets/immunology , Blood Platelets/metabolism , Cell Adhesion/immunology , Cell Movement/immunology , Cell Polarity/immunology , Green Fluorescent Proteins/genetics , Leukocytes/immunology , Mesenteric Arteries/immunology , Mesenteric Arteries/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Needlestick Injuries/immunology , Needlestick Injuries/pathology , Neutrophils/cytology , Neutrophils/immunology , Reperfusion Injury/immunology , Reperfusion Injury/pathology
12.
Proc Natl Acad Sci U S A ; 110(9): 3471-6, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23401506

ABSTRACT

Polymorphisms in MHC class II molecules, in particular around ß-chain position-57 (ß57), afford susceptibility/resistance to multiple autoimmune diseases, including type 1 diabetes, through obscure mechanisms. Here, we show that the antidiabetogenic MHC class II molecule I-A(b) affords diabetes resistance by promoting the differentiation of MHC-promiscuous autoreactive CD4(+) T cells into disease-suppressing natural regulatory T cells, in a ß56-67-regulated manner. We compared the tolerogenic and antidiabetogenic properties of CD11c promoter-driven transgenes encoding I-A(b) or a form of I-A(b) carrying residues 56-67 of I-Aß(g7) (I-A(b-g7)) in wild-type nonobese diabetic (NOD) mice, as well as NOD mice coexpressing a diabetogenic and I-A(g7)-restricted, but MHC-promiscuous T-cell receptor (4.1). Both I-A transgenes protected NOD and 4.1-NOD mice from diabetes. However, whereas I-A(b) induced 4.1-CD4(+) thymocyte deletion and 4.1-CD4(+)Foxp3(+) regulatory T-cell development, I-A(b-g7) promoted 4.1-CD4(+)Foxp3(+) Treg development without inducing clonal deletion. Furthermore, non-T-cell receptor transgenic NOD.CD11cP-I-A(b) and NOD.CD11cP-IA(b-g7) mice both exported regulatory T cells with superior antidiabetogenic properties than wild-type NOD mice. We propose that I-A(b), and possibly other protective MHC class II molecules, afford disease resistance by engaging a naturally occurring constellation of MHC-promiscuous autoreactive T-cell clonotypes, promoting their deviation into autoregulatory T cells.


Subject(s)
Cell Differentiation/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Forkhead Transcription Factors/metabolism , Histocompatibility Antigens Class II/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , CD11c Antigen/genetics , Clonal Anergy/immunology , Dendritic Cells/immunology , Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/prevention & control , Down-Regulation , Immune Tolerance/immunology , Keratin-14/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Promoter Regions, Genetic/genetics , Thymocytes/immunology , Transgenes/genetics
13.
Eur J Immunol ; 42(7): 1717-22, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22585671

ABSTRACT

Infiltration of pancreatic islets by immune cells, termed insulitis, increases progressively once it begins and leads to clinical type 1 diabetes. But even after diagnosis some islets remain unaffected and infiltration is patchy rather than uniform. Traffic of autoreactive T cells into the pancreas is likely to contribute to insulitis progression but it could also depend on T-cell proliferation within islets. This study utilizes transgenic NOD mice to assess the relative contributions of these two mechanisms. Progression of insulitis in NOD8.3 TCR transgenic mice was mildly reduced by inhibition of T-cell migration with the drug FTY720. In FTY720-treated mice, reduced beta cell MHC class I expression prevented progression of insulitis both within affected islets and to previously unaffected islets. CTL proliferation was significantly reduced in islets with reduced or absent beta cell expression of MHC class I protein. This indicates that intra-islet proliferation, apparently dependent on beta cell antigen presentation, in addition to recruitment, is a significant factor in progression of insulitis.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Proliferation , Diabetes Mellitus, Type 1/pathology , Disease Progression , Fingolimod Hydrochloride , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Immunohistochemistry , Immunosuppressive Agents/pharmacology , Islets of Langerhans/cytology , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Mice, Transgenic , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , T-Lymphocytes, Cytotoxic/cytology
14.
Curr Drug Targets ; 13(4): 526-40, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22250649

ABSTRACT

The globally rising incidence of Type 1 diabetes (T1D) is no longer restricted to individuals with higher risk genotypes, but is now significantly increasing in a population with lower risk genotypes, likely as the result of environmental factors. In this review, we discuss the potential of advanced glycation end products (AGEs) as environmental contributors to the development of T1D. AGEs are nonenzymatically formed protein modifications found in the body, as well as, consumed in our daily diets. To date, many studies have provided evidence of AGE involvement in ß cell dysfunction, whether by AGE modification itself or via interaction with AGE receptors. The receptor for AGE (RAGE) and AGE-receptor-1 (AGE-R1) are of particular interest, given that studies have demonstrated the deleterious effects of RAGE modulation and the protection afforded by AGE-R1 in the context of diabetes. More interestingly, we have recently found that two RAGE polymorphism are predictive of T1D in humans while the third is protective. Moreover, soluble RAGE (sRAGE) levels (a circulating competitive inhibitor of RAGE) were greatly reduced at seroconversion to autoantibodies in both children on high risk of T1D background and in an animal model of autoiummune diabetes. Taken together with the fact that AGEs have also shown to be involved in immunomodulation, it is tempting to postulate that dietary AGEs, RAGE and even AGE-R1 could be working synergistically or independently to breach the tightly regulated immune system, providing a missing link in the development of T1D.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Environmental Exposure/adverse effects , Glycation End Products, Advanced/metabolism , Animals , Autoantibodies/immunology , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/immunology , Genotype , Humans , Insulin-Secreting Cells/metabolism , Polymorphism, Genetic , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Risk Factors
15.
Immunol Cell Biol ; 90(2): 243-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21383770

ABSTRACT

CD8(+) T cells kill pancreatic ß-cells in a cell-cell contact-dependent mechanism in the non-obese diabetic mouse. CD4(+) T lymphocytes are also able to kill pancreatic ß-cells, but they do not directly contact ß-cells and may use another cell type as the actual cytotoxic cell. Natural killer (NK) cells could have this role but it is uncertain whether they are cytotoxic towards ß-cells. Therefore, the requirement for NK cells in ß-cell destruction in the CD4-dependent T-cell antigen receptor transgenic NOD4.1 mice was examined. NK cells failed to kill ß-cells in vitro, even in the absence of major histocompatibility complex class I. We observed only 9.7±1.1% of islet infiltrating NK cells from NOD4.1 mice expressing the degranulation marker CD107a. Diabetogenic CD4(+) T cells transferred disease to NODscid.IL2Rγ(-/-) mice lacking NK cells, indicating that NK cells do not contribute to ß-cell death in vitro or in vivo. However, depletion of NK cells reduced diabetes incidence in NOD4.1 mice, suggesting that NK cells may help to maintain the right environment for cytotoxicity of effector cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Killer Cells, Natural/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Diabetes Mellitus, Type 1/genetics , HLA-A Antigens/immunology , Insulin-Secreting Cells/cytology , Lysosomal-Associated Membrane Protein 1/metabolism , Major Histocompatibility Complex , Mice , Mice, Inbred NOD , Mice, Transgenic , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Receptors, Interleukin-2/genetics
16.
Diabetes ; 60(10): 2523-32, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21911745

ABSTRACT

OBJECTIVE: Excess accumulation of advanced glycation end products (AGEs) contributes to aging and chronic diseases. We aimed to obtain evidence that exposure to AGEs plays a role in the development of type 1 diabetes (T1D). RESEARCH DESIGN AND METHODS: The effect of AGEs was examined on insulin secretion by MIN6N8 cells and mouse islets and in vivo in three separate rodent models: AGE-injected or high AGE-fed Sprague-Dawley rats and nonobese diabetic (NODLt) mice. Rodents were also treated with the AGE-lowering agent alagebrium. RESULTS: ß-Cells exposed to AGEs displayed acute glucose-stimulated insulin secretory defects, mitochondrial abnormalities including excess superoxide generation, a decline in ATP content, loss of MnSOD activity, reduced calcium flux, and increased glucose uptake, all of which were improved with alagebrium treatment or with MnSOD adenoviral overexpression. Isolated mouse islets exposed to AGEs had decreased glucose-stimulated insulin secretion, increased mitochondrial superoxide production, and depletion of ATP content, which were improved with alagebrium or with MnTBAP, an SOD mimetic. In rats, transient or chronic exposure to AGEs caused progressive insulin secretory defects, superoxide generation, and ß-cell death, ameliorated with alagebrium. NODLt mice had increased circulating AGEs in association with an increase in islet mitochondrial superoxide generation, which was prevented by alagebrium, which also reduced the incidence of autoimmune diabetes. Finally, at-risk children who progressed to T1D had higher AGE concentrations than matched nonprogressors. CONCLUSIONS: These findings demonstrate that AGEs directly cause insulin secretory defects, most likely by impairing mitochondrial function, which may contribute to the development of T1D.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Glycation End Products, Advanced/pharmacology , Insulin-Secreting Cells/metabolism , Adolescent , Animals , Cell Line , Child , Diabetes Mellitus, Type 1/blood , Female , Glycation End Products, Advanced/blood , Glycation End Products, Advanced/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Rats , Rats, Sprague-Dawley
17.
Am J Pathol ; 178(6): 2716-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21641394

ABSTRACT

Cytotoxic T lymphocytes (CTLs) that cause type 1 diabetes are activated in draining lymph nodes and become concentrated as fully active CTLs in inflamed pancreatic islets. It is unclear whether CTL function is driven by signals received in the lymph node or also in the inflamed tissue. We studied whether the development of cytotoxicity requires further activation in islets. Autoreactive CTLs found in the islets of diabetes-prone NOD mice had acquired much higher expression of the cytotoxic effector markers granzyme B, interferon γ, and CD107a than had those in the pancreatic lymph node (PLN). Increased expression seemed to result from stimulation in the islet itself. T cells held up from migrating from the PLN by administration of the sphingosine-1-phosphate agonist FTY720 did not increase expression of cytotoxic molecules in the PLN. Stimulation did not require antigen presentation or cytokine secretion by the target ß cells because it was not affected by the absence of class I major histocompatibility complex expression or by the overexpression of suppressor of cytokine signaling-1. Activation of CD40-expressing cells stimulated increased CTL function and ß-cell destruction, suggesting that signals derived from CD40-expressing cells promote the acquisition of cytotoxicity in the islet environment. These data provide in vivo evidence that stimulation of cytotoxic effector molecule expression occurs in inflamed islets and is independent of ß cells.


Subject(s)
Cross-Priming/immunology , Cytotoxicity, Immunologic , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymph Nodes/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen Presentation/immunology , Biomarkers/metabolism , Cell Proliferation , Cytokines/biosynthesis , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/immunology , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Lymph Nodes/pathology , Mice , Mice, Inbred NOD , Rats , T-Lymphocytes, Cytotoxic/pathology
18.
J Immunol ; 181(5): 3400-12, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18714012

ABSTRACT

Type 1 NKT cells play a critical role in controlling the strength and character of adaptive and innate immune responses. We have previously reported deficiencies in the numbers and function of NKT cells in the NOD mouse strain, which is a well-validated model of type 1 diabetes and systemic lupus erythematosus. Genetic control of thymic NKT cell numbers was mapped to two linkage regions: Nkt1 on distal chromosome 1 and Nkt2 on chromosome 2. Herein, we report the production and characterization of a NOD.Nkrp1(b).Nkt2b(b) congenic mouse strain, which has increased thymic and peripheral NKT cells, a decreased incidence of type 1 diabetes, and enhanced cytokine responses in vivo and increased proliferative responses in vitro following challenge with alpha-galactosylceramide. The 19 highly differentially expressed candidate genes within the congenic region identified by microarray expression analyses included Pxmp4. This gene encodes a peroxisome-associated integral membrane protein whose only known binding partner is Pex19, an intracellular chaperone and component of the peroxisomal membrane insertion machinery encoded by a candidate for the NKT cell control gene Nkt1. These findings raise the possibility that peroxisomes play a role in modulating glycolipid availability for CD1d presentation, thereby influencing NKT cell function.


Subject(s)
Gene Expression Profiling , Killer Cells, Natural/immunology , Membrane Proteins/genetics , Peroxisomes/immunology , Animals , Antigens, CD1 , Antigens, CD1d , Cytokines/biosynthesis , Diabetes Mellitus, Type 1 , Humans , Killer Cells, Natural/cytology , Lymphocyte Count , Mice , Mice, Congenic , Mice, Inbred NOD
19.
Mol Immunol ; 45(7): 1847-62, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18083234

ABSTRACT

AIRE (autoimmune regulator) promotes the establishment of self-tolerance by regulating gene expression in the thymus. Mutations in AIRE lead to an autoimmune disease, APECED. Here we have identified PIAS proteins as novel AIRE interaction partners. Although PIAS proteins function as E3 SUMO ligases, AIRE is not sumoylated. We expressed AIRE, wt PIAS1, and PIAS1 mutants with deleted SP-RING domain or SUMO interaction motif (SIM) in different cell lines and demonstrate that AIRE and PIAS1 localize to adjacent nuclear bodies (NBs). The expression of AIRE enhances the formation of PIAS1 NBs. The ability of PIAS1 to localize into NBs and interconnect with AIRE is neither dependent on the SP-RING domain nor the SIM. Further, we show that PIAS1 is able to attract AIRE into SUMO1-containing complexes and that the process is dependent on the SIM of PIAS1. PIAS1 and AIRE concurrently activate the human insulin promoter, a known target gene of AIRE, and the SP-RING is required for this activation. Moreover, AIRE represses and PIAS1 activates the CSTB promoter, used as a model for a housekeeping promoter, and both the SP-RING and SIM are needed for its activation by PIAS1. Collectively, our data suggest that AIRE and PIAS1 interact functionally to regulate the activities of the target genes of AIRE.


Subject(s)
Gene Expression Regulation , Protein Inhibitors of Activated STAT/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Amino Acid Motifs , Animals , Cell Line , Cystatin B , Cystatins/genetics , Humans , Insulin/genetics , Mice , Mutant Proteins/metabolism , Nuclear Matrix/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Inhibitors of Activated STAT/chemistry , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Transport , Sequence Deletion , Small Ubiquitin-Related Modifier Proteins/metabolism , Subcellular Fractions/metabolism , Transcriptional Activation/genetics , Transfection , AIRE Protein
20.
Proc Natl Acad Sci U S A ; 104(4): 1295-300, 2007 Jan 23.
Article in English | MEDLINE | ID: mdl-17229843

ABSTRACT

Type 1 diabetes (T1D) is caused by the destruction of insulin-producing islet beta cells. CD8 T cells are prevalent in the islets of T1D patients and are the major effectors of beta cell destruction in nonobese diabetic (NOD) mice. In addition to their critical involvement in the late stages of diabetes, CD8 T cells are implicated in the initiation of disease. NOD mice, in which the beta2-microglobulin gene has been inactivated by gene targeting (NOD.beta2M-/-), have a deficiency in CD8 T cells and do not develop insulitis, which suggests that CD8 T cells are required for the initiation of T1D. However, neither in humans nor in NOD mice have the immunological requirements for diabetogenic CD8 T cells been precisely defined. In particular, it is not known in which cell type MHC class I expression is required for recruitment and activation of CD8 T cells. Here we have generated transgenic NOD mice, which lack MHC class I on mature professional antigen-presenting cells (pAPCs). These "class I APC-bald" mice developed periislet insulitis but not invasive intraislet insulitis, and they never became diabetic. Recruitment to the islet milieu does not therefore require cognate interaction between CD8 T cells and MHC class I on mature pAPCs. Conversely, such an interaction is critically essential to allow the crucial shift from periislet insulitis to invasive insulitis. Importantly, our findings demonstrate unequivocally that CD8 T cells cannot be primed to become diabetogenic by islet beta cells alone.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Animals , Base Sequence , DNA Primers , Female , Flow Cytometry , Immunophenotyping , Lymphocyte Activation , Mice , Mice, Inbred NOD , Obesity/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...