Medicine (St Vincent's) - Theses

Permanent URI for this collection

Search Results

Now showing 1 - 2 of 2
  • Item
    Thumbnail Image
    Induction of antigen-specific tolerance and development of autoreactive T cells in an experimental model of autoimmune diabetes
    Jhala, Gaurang ( 2016)
    Immune responses to proinsulin initiate anti-islet autoimmunity in non-obese diabetic (NOD) mice and possibly in humans. This results in autoimmune destruction of insulin secreting beta cells leading to type 1 diabetes (T1D). Therapies that bolster immune tolerance to islet antigens are highly desirable, however such approaches have failed to prevent clinical T1D. The major aim of this thesis was to determine a stage of life when antigen-specific tolerance is most effective in preventing anti-islet immune responses. Chapter 2 describes generation and validation of transgenic NOD mice engineered to express islet antigens proinsulin (TIP mice) and IGRP (TII mice) in the antigen presenting cells (APCs) in a tetracycline dependent manner. MHC class II IEα promoter in combination with tet-OFF transactivator induced robust, doxycycline dependent and APC specific expression of proinsulin and IGRP in TIP and TII mice respectively. TIP mice expressing proinsulin did not develop insulitis and were protected from cyclophosphamide-induced diabetes, suggesting that proinsulin expression in TIP mice was sufficient to induce functional antigen-specific tolerance. In chapter 3, we examined the impact of antigen-specific tolerance on the development of autoreactive T cells and spontaneous diabetes by expressing islet antigens proinsulin and IGRP in the APCs during defined periods of life in TIP and TII mice. Our results indicate that tolerance to proinsulin in early life until the weaning period is sufficient to prevent diabetes development in TIP mice. The protection from diabetes was not due to dominant tolerance, but mainly due to a significant reduction in the insulin reactive T cells. Although insulin reactive T cells were not completely absent, the residual autoreactive T cells lacked pathogenic potential. By tracking IGRP reactive T cells in TII mice we demonstrate that IGRP T cells also emerge during early life. These data suggest that early life is a vulnerable period for escape of islet reactive T cells, and that boosting immune tolerance to islet antigens during this time imparts durable protection from islet autoimmunity. Immune tolerance to proinsulin-2 imparts robust protection from autoimmune diabetes in the NOD mice. Whether dampening immune responses to proinsulin-1 would influence diabetes development in NOD mice remains to be investigated. Chapter 4 describes the generation of transgenic NOD mice that express proinsulin-1 in the APCs (TIP-1 mice) in a tetracycline dependent manner. TIP-1 mice displayed a significantly reduced incidence of spontaneous diabetes, which was associated with reduced severity of insulitis and insulin autoantibody development. Antigen experienced proinsulin specific T cells were significantly reduced in number in TIP-1 mice indicating immune tolerance. Although immune response to downstream antigen IGRP was reduced in TIP-1 mice, tolerance to proinsulin-1 was unable to prevent diabetes in NOD 8.3 mice with a pre-existing repertoire of IGRP reactive T cells. Thus, despite being highly conserved to proinsulin-2, tolerance to proinsulin-1 only partially prevents islet-autoimmunity in NOD mice, which suggests an ongoing residual immune response to proinsulin-2 epitopes in TIP-1 mice.
  • Item
    Thumbnail Image
    Dissecting the interferon response required for triggering autoimmune diabetes
    Quah, Hong Sheng ( 2016)
    Type 1 diabetes (T1D) is characterized by the autoimmune destruction of β cells in the islets of Langerhans by immune cells. The interferon (IFN) family of cytokines has been implicated in diabetes pathogenesis, possibly via the activation of innate immune pathways. However, how IFNs contribute to the pathogenesis of autoimmune diabetes remains unclear. The overall aim of this thesis is to dissect the role of IFNs in the triggering of autoimmune diabetes using the non-obese diabetic (NOD) mouse model. Chapter 2 describes the contribution of type I IFN to the development of autoimmune diabetes in NOD mice. The islets of NOD mice displayed a unique mRNA expression pattern of IFN-induced genes that peaked at 3-4 weeks of age. Genetic ablation of type I IFN receptor in NOD mice (NOD.Ifnar1-/-) significantly reduced the expression of these genes. However, lack of IFNAR1 did not affect the development of diabetes. The overlapping function of type III IFN could be a reason for the lack of effect of IFNAR1-deficiency. Another reason could be that the natural level of type I IFN produced in wild-type NOD mice is insufficient to have an effect on diabetes development. In Chapter 3, the possible role of type III IFN in autoimmune diabetes was examined. β cells isolated from humans and mice expressed the type III IFN receptor and β cells were able to respond to type III IFN stimulation. Type III IFN was detected in the islets in NOD mice. These results indicate that type III IFN may contribute to diabetes development in NOD mice. Future experiments using NOD mice deficient in the type III IFN receptor will determine whether type III IFN contributes to the development of autoimmune diabetes. Engagement of pattern recognition receptors (PRRs) with exogenous and endogenous danger signals can trigger type I IFN production. Endogenous danger signals, such as aberrant cytosolic DNA, are normally eliminated to prevent unnecessary induction of PRRs. Nucleases, such as the 3’ exonuclease TREX1, are important in degrading aberrant DNA, and have been implicated in protection from autoimmunity. TREX1 is normally found in the SET complex, and one protease that can activate this complex is granzyme A. In Chapter 4, I show that granzyme A deficiency resulted in increased diabetes in NOD mice. Single-stranded DNA accumulated in the cytoplasm of dendritic cells and NK cells in the islets and spleens, and this was observed more frequently in NOD.Gzma-/- mice. Consistent with poor clearance of DNA and increased PRR activation, expression of IFN-induced genes was higher in the islets of NOD.Gzma-/- mice than NOD mice at 4 weeks of age. When NOD.Gzma-/- mice were crossed with NOD.Ifnar1-/- mice, diabetes returned to the rate observed in NOD mice. Overall, the data indicate that the natural level of type I IFN produced in wild-type NOD mice is not sufficient to cause diabetes, but excessive type I IFN can increase diabetes development in NOD.Gzma-/- mice. The results provide mechanistic insight into the triggering of autoimmune diabetes, suggesting that aberrant accumulation of cytoplasmic DNA and type I IFN production are important. In this study, these were caused by loss of granzyme A, but it is also possible that virus infection could similarly activate type I IFN and possibly also type III IFN to trigger T1D.