Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
J Autoimmun ; 90: 39-48, 2018 06.
Article in English | MEDLINE | ID: mdl-29439835

ABSTRACT

Dendritic cell (DC)-mediated T cell tolerance deficiencies contribute to the pathogenesis of autoimmune diseases such as type 1 diabetes. Delivering self-antigen to dendritic-cell inhibitory receptor-2 (DCIR2)+ DCs can delay but not completely block diabetes development in NOD mice. These DCIR2-targeting antibodies induce tolerance via deletion and anergy, but do not increase islet-specific Tregs. Because low-dose IL-2 (LD-IL-2) administration can preferentially expand Tregs, we tested whether delivering islet-antigen to tolerogenic DCIR2+ DCs along with LD-IL-2 would boost islet-specific Tregs and further block autoimmunity. But, surprisingly, adding LD-IL-2 did not increase efficacy of DC-targeted antigen to inhibit diabetes. Here we show the effects of LD-IL-2, with or without antigen delivery to DCIR2+ DCs, on both polyclonal and autoreactive Treg and conventional T cells (Tconv). As expected, LD-IL-2 increased total Tregs, but autoreactive Tregs required both antigen and IL-2 stimulation for optimal expansion. Also, islet-specific Tregs had lower CD25 expression and IL-2 sensitivity, while islet-specific Tconv had higher CD25 expression, compared to polyclonal populations. LD-IL-2 increased activation and expansion of Tconv, and was more pronounced for autoreactive cells after treatment with IL-2 + islet-antigen. Therefore, LD-IL-2 therapy, especially when combined with antigen stimulation, may not optimally activate and expand antigen-specific Tregs in chronic autoimmune settings.


Subject(s)
Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation , Autoantigens/immunology , Autoimmunity , Cells, Cultured , Female , Humans , Immune Tolerance , Mice , Mice, Inbred NOD , Mice, Transgenic , Receptors, Cell Surface/metabolism
2.
J Immunol ; 196(5): 2031-40, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26826238

ABSTRACT

Innate immune signals help break self-tolerance to initiate autoimmune diseases such as type 1 diabetes, but innate contributions to subsequent regulation of disease progression are less clear. Most studies have measured in vitro innate responses of GM-CSF dendritic cells (DCs) that are functionally distinct from conventional DCs (cDCs) and do not reflect in vivo DC subsets. To determine whether autoimmune NOD mice have alterations in type 1 IFN innate responsiveness, we compared cDCs from prediabetic NOD and control C57BL/6 (B6) mice stimulated in vivo with the TLR9 ligand CpG, a strong type 1 IFN inducer. In response to CpG, NOD mice produce more type 1 IFN and express higher levels of CD40, and NOD monocyte DCs make more TNF. However, the overall CpG-induced transcriptional response is muted in NOD cDCs. Of relevance the costimulatory proteins CD80/CD86, signals needed for regulatory T cell homeostasis, are upregulated less on NOD cDCs. Interestingly, NOD Rag1(-/-) mice also display a defect in CpG-induced CD86 upregulation compared with B6 Rag1(-/-), indicating this particular innate alteration precedes adaptive autoimmunity. The impaired response in NOD DCs is likely downstream of the IFN-α/ß receptor because DCs from NOD and B6 mice show similar CpG-induced CD86 levels when anti-IFN-α/ß receptor Ab is added. IFN-α-induced nuclear localization of activated STAT1 is markedly reduced in NOD CD11c(+) cells, consistent with lower type 1 IFN responsiveness. In conclusion, NOD DCs display altered innate responses characterized by enhanced type 1 IFN and activation of monocyte-derived DCs but diminished cDC type 1 IFN response.


Subject(s)
Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Immunity, Innate/immunology , Interferon Type I/immunology , STAT1 Transcription Factor/immunology , Self Tolerance/immunology , Active Transport, Cell Nucleus , Animals , Blotting, Western , Cell Lineage , Cell Nucleus/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Microscopy, Confocal , Monocytes/cytology , Monocytes/immunology , Oligodeoxyribonucleotides/immunology , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , STAT1 Transcription Factor/metabolism , Toll-Like Receptor 9/agonists
3.
F1000Res ; 7: 318, 2018.
Article in English | MEDLINE | ID: mdl-29707204

ABSTRACT

Background: We previously identified the transcriptional regulator Zbtb32 as a factor that can promote T cell tolerance in the Non-Obese Diabetic (NOD) mouse, a model of Type 1 diabetes. Antigen targeted to DCIR2 + dendritic cells (DCs) in vivo inhibited both diabetes and effector T cell expansion in NOD mice. Furthermore, Zbtb32 was preferentially induced in autoreactive CD4 T cells stimulated by these tolerogenic DCIR2 + DCs, and overexpression of Zbtb32 in islet-specific T cells inhibited the diabetes development by limiting T cell proliferation and cytokine production. Methods: To further understand the role of Zbtb32 in T cell tolerance induction, we have now used CRISPR to target the Zbtb32 gene for deletion directly in NOD mice and characterized the mutant mice. We hypothesized that the systemic loss of Zbtb32 in NOD mice would lead to increased T cell activation and increased diabetes pathogenesis. Results: Although NOD.Zbtb32 -/- male NOD mice showed a trend towards increased diabetes incidence compared to littermate controls, the difference was not significant. Furthermore, no significant alteration in lymphocyte number or function was observed. Importantly, in vitro stimulation of lymphocytes from NOD.Zbtb32 -/- mice did not produce the expected hypersensitive phenotype observed in other genetic strains, potentially due to compensation by homologous genes. Conclusions: The loss of Zbtb32 in the NOD background does not result in the expected T cell activation phenotype.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Diabetes Mellitus, Experimental/epidemiology , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Repressor Proteins/physiology , Animals , CRISPR-Cas Systems , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Female , Incidence , Male , Mice , Mice, Inbred NOD , Repressor Proteins/antagonists & inhibitors
4.
JCI Insight ; 3(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29415894

ABSTRACT

Type I IFN (IFN-I) dysregulation contributes to type 1 diabetes (T1D) development, and although increased IFN-I signals are pathogenic at the initiation of autoimmune diabetes, IFN-I dysregulation at later pathogenic stages more relevant for therapeutic intervention is not well understood. We discovered that 5 key antigen-presenting cell subsets from adult prediabetic NOD mice have reduced responsiveness to IFN-I that is dominated by a decrease in the tonic-sensitive subset of IFN-I response genes. Blockade of IFNAR1 in prediabetic NOD mice accelerated diabetes and increased Th1 responses. Therefore, IFN-I responses shift from pathogenic to protective as autoimmunity progresses, consistent with chronic IFN-I exposure. In contrast, IL-1-associated inflammatory pathways were elevated in prediabetic mice. These changes correlated with human T1D onset-associated gene expression. Prostaglandin E2 (PGE2) and prostaglandin receptor 4 (PTGER4), a receptor for PGE2 that mediates both inflammatory and regulatory eicosanoid signaling, were higher in NOD mice and drive innate immune dysregulation. Treating prediabetic NOD mice with a PTGER4 antagonist restored IFNAR signaling, decreased IL-1 signaling, and decreased infiltration of leukocytes into the islets. Therefore, innate cytokine alterations contribute to both T1D-associated inflammation and autoimmune pathogenesis. Modulating innate immune balance via signals such as PTGER4 may contribute to treatments for autoimmunity.


Subject(s)
Autoimmunity/drug effects , Diabetes Mellitus, Type 1/immunology , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Sulfonylurea Compounds/administration & dosage , Th1 Cells/immunology , Administration, Oral , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Diabetes Mellitus, Type 1/drug therapy , Dinoprostone/immunology , Dinoprostone/metabolism , Disease Models, Animal , Female , Humans , Interleukin-1/immunology , Interleukin-1/metabolism , Mice , Mice, Inbred NOD , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Receptors, Prostaglandin E, EP4 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Th1 Cells/drug effects , Th1 Cells/metabolism
5.
J Leukoc Biol ; 100(1): 65-80, 2016 07.
Article in English | MEDLINE | ID: mdl-26792821

ABSTRACT

Type 1 diabetes is an autoimmune disease that results from the defective induction or maintenance of T cell tolerance against islet ß cell self-antigens. Under steady-state conditions, dendritic cells with tolerogenic properties are critical for peripheral immune tolerance. Tolerogenic dendritic cells can induce T cell anergy and deletion and, in some contexts, induce or expand regulatory T cells. Dendritic cells contribute to both immunomodulatory effects and triggering of pathogenesis in type 1 diabetes. This immune equilibrium is affected by both genetic and environmental factors that contribute to the development of type 1 diabetes. Genome-wide association studies and disease association studies have identified >50 polymorphic loci that lend susceptibility or resistance to insulin-dependent diabetes mellitus. In parallel, diabetes susceptibility regions known as insulin-dependent diabetes loci have been identified in the nonobese diabetic mouse, a model for human type 1 diabetes, providing a better understanding of potential immunomodulatory factors in type 1 diabetes risk. Most genetic candidates have annotated immune cell functions, but the focus has been on changes to T and B cells. However, it is likely that some of the genomic susceptibility in type 1 diabetes directly interrupts the tolerogenic potential of dendritic cells in the pathogenic context of ongoing autoimmunity. Here, we will review how gene polymorphisms associated with autoimmune diabetes may influence dendritic cell development and maturation processes that could lead to alterations in the tolerogenic function of dendritic cells. These insights into potential tolerogenic and pathogenic roles for dendritic cells have practical implications for the clinical manipulation of dendritic cells toward tolerance to prevent and treat type 1 diabetes.


Subject(s)
Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Immune Tolerance/immunology , Animals , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Humans
6.
Diabetes ; 64(10): 3521-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26070317

ABSTRACT

During autoimmunity, the normal ability of dendritic cells (DCs) to induce T-cell tolerance is disrupted; therefore, autoimmune disease therapies based on cell types and molecular pathways that elicit tolerance in the steady state may not be effective. To determine which DC subsets induce tolerance in the context of chronic autoimmunity, we used chimeric antibodies specific for DC inhibitory receptor 2 (DCIR2) or DEC-205 to target self-antigen to CD11b(+) (cDC2) DCs and CD8(+) (cDC1) DCs, respectively, in autoimmune-prone nonobese diabetic (NOD) mice. Antigen presentation by DCIR2(+) DCs but not DEC-205(+) DCs elicited tolerogenic CD4(+) T-cell responses in NOD mice. ß-Cell antigen delivered to DCIR2(+) DCs delayed diabetes induction and induced increased T-cell apoptosis without interferon-γ (IFN-γ) or sustained expansion of autoreactive CD4(+) T cells. These divergent responses were preceded by differential gene expression in T cells early after in vivo stimulation. Zbtb32 was higher in T cells stimulated with DCIR2(+) DCs, and overexpression of Zbtb32 in T cells inhibited diabetes development, T-cell expansion, and IFN-γ production. Therefore, we have identified DCIR2(+) DCs as capable of inducing antigen-specific tolerance in the face of ongoing autoimmunity and have also identified Zbtb32 as a suppressive transcription factor that controls T cell-mediated autoimmunity.


Subject(s)
Antibodies , Antigens, CD/metabolism , Autoimmunity/physiology , CD4-Positive T-Lymphocytes/physiology , Dendritic Cells/physiology , Diabetes Mellitus/immunology , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Repressor Proteins/metabolism , Animals , Antigens, CD/genetics , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/genetics , CD40 Antigens/metabolism , Diabetes Mellitus/prevention & control , Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , Lectins, C-Type/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Minor Histocompatibility Antigens , Receptors, Cell Surface/genetics , Repressor Proteins/genetics , Specific Pathogen-Free Organisms
SELECTION OF CITATIONS
SEARCH DETAIL