Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Metab ; 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38959897

ABSTRACT

A mechanistic connection between aging and development is largely unexplored. Through profiling age-related chromatin and transcriptional changes across 22 murine cell types, analyzed alongside previous mouse and human organismal maturation datasets, we uncovered a transcription factor binding site (TFBS) signature common to both processes. Early-life candidate cis-regulatory elements (cCREs), progressively losing accessibility during maturation and aging, are enriched for cell-type identity TFBSs. Conversely, cCREs gaining accessibility throughout life have a lower abundance of cell identity TFBSs but elevated activator protein 1 (AP-1) levels. We implicate TF redistribution toward these AP-1 TFBS-rich cCREs, in synergy with mild downregulation of cell identity TFs, as driving early-life cCRE accessibility loss and altering developmental and metabolic gene expression. Such remodeling can be triggered by elevating AP-1 or depleting repressive H3K27me3. We propose that AP-1-linked chromatin opening drives organismal maturation by disrupting cell identity TFBS-rich cCREs, thereby reprogramming transcriptome and cell function, a mechanism hijacked in aging through ongoing chromatin opening.

2.
Front Immunol ; 15: 1370907, 2024.
Article in English | MEDLINE | ID: mdl-38533515

ABSTRACT

Introduction: Chronic activation of self-reactive T cells with beta cell antigens results in the upregulation of immune checkpoint molecules that keep self-reactive T cells under control and delay beta cell destruction in autoimmune diabetes. Inhibiting PD1/PD-L1 signaling results in autoimmune diabetes in mice and humans with pre-existing autoimmunity against beta cells. However, it is not known if other immune checkpoint molecules, such as TIGIT, can also negatively regulate self-reactive T cells. TIGIT negatively regulates the CD226 costimulatory pathway, T-cell receptor (TCR) signaling, and hence T-cell function. Methods: The phenotype and function of TIGIT expressing islet infiltrating T cells was studied in non-obese diabetic (NOD) mice using flow cytometry and single cell RNA sequencing. To determine if TIGIT restrains self-reactive T cells, we used a TIGIT blocking antibody alone or in combination with anti-PDL1 antibody. Results: We show that TIGIT is highly expressed on activated islet infiltrating T cells in NOD mice. We identified a subset of stem-like memory CD8+ T cells expressing multiple immune checkpoints including TIGIT, PD1 and the transcription factor EOMES, which is linked to dysfunctional CD8+ T cells. A known ligand for TIGIT, CD155 was expressed on beta cells and islet infiltrating dendritic cells. However, despite TIGIT and its ligand being expressed, islet infiltrating PD1+TIGIT+CD8+ T cells were functional. Inhibiting TIGIT in NOD mice did not result in exacerbated autoimmune diabetes while inhibiting PD1-PDL1 resulted in rapid autoimmune diabetes, indicating that TIGIT does not restrain islet infiltrating T cells in autoimmune diabetes to the same degree as PD1. Partial inhibition of PD1-PDL1 in combination with TIGIT inhibition resulted in rapid diabetes in NOD mice. Discussion: These results suggest that TIGIT and PD1 act in synergy as immune checkpoints when PD1 signaling is partially impaired. Beta cell specific stem-like memory T cells retain their functionality despite expressing multiple immune checkpoints and TIGIT is below PD1 in the hierarchy of immune checkpoints in autoimmune diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Animals , Humans , Mice , Immune Checkpoint Proteins , Ligands , Mice, Inbred NOD , Receptors, Immunologic/metabolism
3.
J Autoimmun ; 140: 103090, 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37572540

ABSTRACT

CXCL10 is an IFNγ-inducible chemokine implicated in the pathogenesis of type 1 diabetes. T-cells attracted to pancreatic islets produce IFNγ, but it is unclear what attracts the first IFNγ -producing T-cells in islets. Gut dysbiosis following administration of pathobionts induced CXCL10 expression in pancreatic islets of healthy non-diabetes-prone (C57BL/6) mice and depended on TLR4-signaling, and in non-obese diabetic (NOD) mice, gut dysbiosis induced also CXCR3 chemokine receptor in IGRP-reactive islet-specific T-cells in pancreatic lymph node. In amounts typical to low-grade endotoxemia, bacterial lipopolysaccharide induced CXCL10 production in isolated islets of wild type and RAG1 or IFNG-receptor-deficient but not type-I-IFN-receptor-deficient NOD mice, dissociating lipopolysaccharide-induced CXCL10 production from T-cells and IFNγ. Although mostly myeloid-cell dependent, also ß-cells showed activation of innate immune signaling pathways and Cxcl10 expression in response to lipopolysaccharide indicating their independent sensitivity to dysbiosis. Thus, CXCL10 induction in response to low levels of lipopolysaccharide may allow islet-specific T-cells imprinted in pancreatic lymph node to enter in healthy islets independently of IFN-g, and thus link gut dysbiosis to early islet-autoimmunity via dysbiosis-associated low-grade endotoxemia.

4.
Clin Transl Immunology ; 11(11): e1425, 2022.
Article in English | MEDLINE | ID: mdl-36325490

ABSTRACT

Objectives: Immune checkpoint inhibitors have achieved clinical success in cancer treatment, but this treatment causes immune-related adverse events, including type 1 diabetes (T1D). Our aim was to test whether a JAK1/JAK2 inhibitor, effective at treating spontaneous autoimmune diabetes in nonobese diabetic (NOD) mice, can prevent diabetes secondary to PD-L1 blockade. Methods: Anti-PD-L1 antibody was injected into NOD mice to induce diabetes, and JAK1/JAK2 inhibitor LN3103801 was administered by oral gavage to prevent diabetes. Flow cytometry was used to study T cells and beta cells. Mesothelioma cells were inoculated into BALB/c mice to induce a transplantable tumour model. Results: Anti-PD-L1-induced diabetes was associated with increased immune cell infiltration in the islets and upregulated MHC class I on islet cells. Anti-PD-L1 administration significantly increased islet T cell proliferation and islet-specific CD8+ T cell numbers in peripheral lymphoid organs. JAK1/JAK2 inhibitor treatment blocked IFNγ-mediated MHC class I upregulation on beta cells and T cell proliferation mediated by cytokines that use the common γ chain receptor. As a result, anti-PD-L1-induced diabetes was prevented by JAK1/JAK2 inhibitor administered before or after checkpoint inhibitor therapy. Diabetes was also reversed when the JAK1/JAK2 inhibitor was administered after the onset of anti-PD-L1-induced hyperglycaemia. Furthermore, JAK1/JAK2 inhibitor intervention after checkpoint inhibitors did not reverse or abrogate the antitumour effects in a transplantable tumour model. Conclusion: A JAK1/JAK2 inhibitor can prevent and reverse anti-PD-L1-induced diabetes by blocking IFNγ and γc cytokine activities. Our study provides preclinical validation of JAK1/JAK2 inhibitor use in checkpoint inhibitor-induced diabetes.

5.
Diabetologia ; 64(4): 878-889, 2021 04.
Article in English | MEDLINE | ID: mdl-33483762

ABSTRACT

AIMS/HYPOTHESIS: Stimulator of IFN genes (STING) is a central hub for cytosolic nucleic acid sensing and its activation results in upregulation of type I IFN production in innate immune cells. A type I IFN gene signature seen before the onset of type 1 diabetes has been suggested as a driver of disease initiation both in humans and in the NOD mouse model. A possible source of type I IFN is through activation of the STING pathway. Recent studies suggest that STING also has antiproliferative and proapoptotic functions in T cells that are independent of IFN. To investigate whether STING is involved in autoimmune diabetes, we examined the impact of genetic deletion of STING in NOD mice. METHODS: CRISPR/Cas9 gene editing was used to generate STING-deficient NOD mice. Quantitative real-time PCR was used to assess the level of type I IFN-regulated genes in islets from wild-type and STING-deficient NOD mice. The number of islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)206-214-specific CD8+ T cells was determined by magnetic bead-based MHC tetramer enrichment and flow cytometry. The incidence of spontaneous diabetes and diabetes after adoptive transfer of T cells was determined. RESULTS: STING deficiency partially attenuated the type I IFN gene signature in islets but did not suppress insulitis. STING-deficient NOD mice accumulated an increased number of IGRP206-214-specific CD8+ T cells (2878 ± 642 cells in NOD.STING-/- mice and 728.8 ± 196 cells in wild-type NOD mice) in peripheral lymphoid tissue, associated with a higher incidence of spontaneous diabetes (95.5% in NOD.STING-/- mice and 86.2% in wild-type NOD mice). Splenocytes from STING-deficient mice rapidly induced diabetes after adoptive transfer into irradiated NOD recipients (median survival 75 days for NOD recipients of NOD.STING-/- mouse splenocytes and 121 days for NOD recipients of NOD mouse splenocytes). CONCLUSIONS/INTERPRETATION: Data suggest that sensing of endogenous nucleic acids through the STING pathway may be partially responsible for the type I IFN gene signature but not autoimmunity in NOD mice. Our results show that the STING pathway may play an unexpected intrinsic role in suppressing the number of diabetogenic T cells.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Diabetes Mellitus, Type 1/metabolism , Islets of Langerhans/metabolism , Lymphocyte Activation , Membrane Proteins/metabolism , Adoptive Transfer , Animals , Autoimmunity , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Female , Gene Expression Regulation , Islets of Langerhans/immunology , Male , Membrane Proteins/genetics , Mice, Inbred NOD , Mice, Knockout , Signal Transduction
6.
Front Immunol ; 11: 588543, 2020.
Article in English | MEDLINE | ID: mdl-33343569

ABSTRACT

Cytokines that signal through the JAK-STAT pathway, such as interferon-γ (IFN-γ) and common γ chain cytokines, contribute to the destruction of insulin-secreting ß cells by CD8+ T cells in type 1 diabetes (T1D). We previously showed that JAK1/JAK2 inhibitors reversed autoimmune insulitis in non-obese diabetic (NOD) mice and also blocked IFN-γ mediated MHC class I upregulation on ß cells. Blocking interferons on their own does not prevent diabetes in knockout NOD mice, so we tested whether JAK inhibitor action on signaling downstream of common γ chain cytokines, including IL-2, IL-7 IL-15, and IL-21, may also affect the progression of diabetes in NOD mice. Common γ chain cytokines activate JAK1 and JAK3 to regulate T cell proliferation. We used a JAK1-selective inhibitor, ABT 317, to better understand the specific role of JAK1 signaling in autoimmune diabetes. ABT 317 reduced IL-21, IL-2, IL-15 and IL-7 signaling in T cells and IFN-γ signaling in ß cells, but ABT 317 did not affect GM-CSF signaling in granulocytes. When given in vivo to NOD mice, ABT 317 reduced CD8+ T cell proliferation as well as the number of KLRG+ effector and CD44hiCD62Llo effector memory CD8+ T cells in spleen. ABT 317 also prevented MHC class I upregulation on ß cells. Newly diagnosed diabetes was reversed in 94% NOD mice treated twice daily with ABT 317 while still on treatment at 40 days and 44% remained normoglycemic after a further 60 days from discontinuing the drug. Our results indicate that ABT 317 blocks common γ chain cytokines in lymphocytes and interferons in lymphocytes and ß cells and are thus more effective against diabetes pathogenesis than IFN-γ receptor deficiency alone. Our studies suggest use of this class of drug for the treatment of type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Interferon-gamma/immunology , Interleukin Receptor Common gamma Subunit/immunology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Animals , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/immunology , Janus Kinase Inhibitors/pharmacokinetics , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Signal Transduction/drug effects , Spleen/immunology
7.
Sci Rep ; 10(1): 13077, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753746

ABSTRACT

Type 17 immune responses, typified by the production of the cytokines IL-17A and IL-17F, have been implicated in the development of type 1 diabetes in animal models and human patients, however the underlying pathogenic mechanisms have not been clearly elucidated. While previous studies show that IL-17A enhances inflammatory gene expression and cell death in mouse ß-cells and human islets, the function of IL-17F in pancreatic ß-cells is completely untested to date. Here we show that IL-17F exhibits potent pathogenic effects in mouse ß-cell lines and islets. IL-17F signals via the IL-17RA and -RC subunits in ß-cells and in combination with other inflammatory cytokines induces expression of chemokine transcripts, suppresses the expression of ß-cell identity genes and impairs glucose stimulated insulin secretion. Further IL-17F induces cell death in primary mouse islets. This occurs via Jnk, p38 and NF-κB dependent induction of Nos2 and is completely ablated in the presence of an inducible nitric oxide synthase (iNOS) inhibitor. Together these data indicate that IL-17F possesses similar pathogenic activities to IL-17A in mouse ß-cell lines and islets and is likely to be a type 17 associated pathogenic factor in type 1 diabetes.


Subject(s)
Interleukin-17/pharmacology , Islets of Langerhans/cytology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Death/drug effects , Cell Line , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Islets of Langerhans/pathology , Mice , Nitric Oxide Synthase Type II/metabolism
8.
Sci Rep ; 9(1): 15302, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31653894

ABSTRACT

In type 1 diabetes, maturation of activated autoreactive CD8+ T cells to fully armed effector cytotoxic T lymphocytes (CTL) occurs within the islet. At present the signals required for the maturation process are poorly defined. Cytokines could potentially provide the necessary "third signal" required to generate fully mature CTL capable of killing insulin-producing ß-cells. To determine whether autoreactive CTL within islets respond to cytokines we generated non-obese diabetic (NOD) mice with a reporter for cytokine signalling. These mice express a reporter gene, hCD4, under the control of the endogenous regulatory elements for suppressor of cytokine signalling (SOCS)1, which is itself regulated by pro-inflammatory cytokines. In NOD mice, the hCD4 reporter was expressed in infiltrated islets and the expression level was positively correlated with the frequency of infiltrating CD45+ cells. SOCS1 reporter expression was induced in transferred ß-cell-specific CD8+ 8.3T cells upon migration from pancreatic draining lymph nodes into islets. To determine which cytokines induced SOCS1 promoter activity in islets, we examined hCD4 reporter expression and CTL maturation in the absence of the cytokine receptors IFNAR1 or IL-21R. We show that IFNAR1 deficiency does not confer protection from diabetes in 8.3 TCR transgenic mice, nor is IFNAR1 signalling required for SOCS1 reporter upregulation or CTL maturation in islets. In contrast, IL-21R-deficient 8.3 mice have reduced diabetes incidence and reduced SOCS1 reporter activity in islet CTLs. However IL-21R deficiency did not affect islet CD8+ T cell proliferation or expression of granzyme B or IFNγ. Together these data indicate that autoreactive CD8+ T cells respond to IL-21 and not type I IFNs in the islets of NOD mice, but neither IFNAR1 nor IL-21R are required for islet intrinsic CTL maturation.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Interleukins/pharmacology , Islets of Langerhans/drug effects , Suppressor of Cytokine Signaling 1 Protein/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Humans , Islets of Langerhans/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Suppressor of Cytokine Signaling 1 Protein/genetics , T-Lymphocytes, Cytotoxic/immunology
9.
Diabetes ; 64(9): 3229-38, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25948683

ABSTRACT

Because regulatory T-cell (Treg) development can be induced by the same agonist self-antigens that induce negative selection, perturbation of apoptosis will affect both negative selection and Treg development. But how the processes of thymocyte deletion versus Treg differentiation bifurcate and their relative importance for tolerance have not been studied in spontaneous organ-specific autoimmune disease. We addressed these questions by removing a critical mediator of thymocyte deletion, BIM, in the NOD mouse model of autoimmune diabetes. Despite substantial defects in the deletion of autoreactive thymocytes, BIM-deficient NOD (NODBim(-/-)) mice developed less insulitis and were protected from diabetes. BIM deficiency did not impair effector T-cell function; however, NODBim(-/-) mice had increased numbers of Tregs, including those specific for proinsulin, in the thymus and peripheral lymphoid tissues. Increased levels of Nur77, CD5, GITR, and phosphorylated IκB-α in thymocytes from NODBim(-/-) mice suggest that autoreactive cells receiving strong T-cell receptor signals that would normally delete them escape apoptosis and are diverted into the Treg pathway. Paradoxically, in the NOD model, reduced thymic deletion ameliorates autoimmune diabetes by increasing Tregs. Thus, modulating apoptosis may be one of the ways to increase antigen-specific Tregs and prevent autoimmune disease.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Clonal Deletion/genetics , Diabetes Mellitus, Type 1/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , T-Lymphocytes, Regulatory/immunology , Thymocytes/immunology , Animals , Apoptosis Regulatory Proteins/immunology , Bcl-2-Like Protein 11 , CD5 Antigens/metabolism , Clonal Deletion/immunology , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Glucocorticoid-Induced TNFR-Related Protein/metabolism , I-kappa B Proteins/metabolism , Membrane Proteins/immunology , Mice , Mice, Inbred NOD , NF-KappaB Inhibitor alpha , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phosphorylation , Proto-Oncogene Proteins/immunology , Thymocytes/metabolism
10.
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
11.
J Biol Chem ; 286(19): 16669-80, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21454529

ABSTRACT

Genomic uracil is a DNA lesion but also an essential key intermediate in adaptive immunity. In B cells, activation-induced cytidine deaminase deaminates cytosine to uracil (U:G mispairs) in Ig genes to initiate antibody maturation. Uracil-DNA glycosylases (UDGs) such as uracil N-glycosylase (UNG), single strand-selective monofunctional uracil-DNA glycosylase 1 (SMUG1), and thymine-DNA glycosylase remove uracil from DNA. Gene-targeted mouse models are extensively used to investigate the role of these enzymes in DNA repair and Ig diversification. However, possible species differences in uracil processing in humans and mice are yet not established. To address this, we analyzed UDG activities and quantities in human and mouse cell lines and in splenic B cells from Ung(+/+) and Ung(-/-) backcrossed mice. Interestingly, human cells displayed ∼15-fold higher total uracil excision capacity due to higher levels of UNG. In contrast, SMUG1 activity was ∼8-fold higher in mouse cells, constituting ∼50% of the total U:G excision activity compared with less than 1% in human cells. In activated B cells, both UNG and SMUG1 activities were at levels comparable with those measured for mouse cell lines. Moreover, SMUG1 activity per cell was not down-regulated after activation. We therefore suggest that SMUG1 may work as a weak backup activity for UNG2 during class switch recombination in Ung(-/-) mice. Our results reveal significant species differences in genomic uracil processing. These findings should be taken into account when mouse models are used in studies of uracil DNA repair and adaptive immunity.


Subject(s)
Uracil-DNA Glycosidase/chemistry , Animals , DNA Repair , Humans , Immunoglobulin Class Switching , Immunoglobulins/chemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Recombination, Genetic , Species Specificity , Thymine DNA Glycosylase/chemistry
12.
J Cell Biol ; 186(5): 645-54, 2009 Sep 07.
Article in English | MEDLINE | ID: mdl-19736315

ABSTRACT

Numerous proteins, many essential for the DNA replication machinery, interact with proliferating cell nuclear antigen (PCNA) through the PCNA-interacting peptide (PIP) sequence called the PIP box. We have previously shown that the oxidative demethylase human AlkB homologue 2 (hABH2) colocalizes with PCNA in replication foci. In this study, we show that hABH2 interacts with a posttranslationally modified PCNA via a novel PCNA-interacting motif, which we term AlkB homologue 2 PCNA-interacting motif (APIM). We identify APIM in >200 other proteins involved in DNA maintenance, transcription, and cell cycle regulation, and verify a functional APIM in five of these. Expression of an APIM peptide increases the cellular sensitivity to several cytostatic agents not accounted for by perturbing only the hABH2-PCNA interaction. Thus, APIM is likely to mediate PCNA binding in many proteins involved in DNA repair and cell cycle control during genotoxic stress.


Subject(s)
Amino Acid Motifs/genetics , DNA Repair Enzymes/metabolism , Dioxygenases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , AlkB Homolog 2, Alpha-Ketoglutarate-Dependent Dioxygenase , Animals , DNA Repair , DNA Repair Enzymes/genetics , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Dioxygenases/genetics , Fluorescence Resonance Energy Transfer , HeLa Cells , Humans , Molecular Sequence Data , Point Mutation , Proliferating Cell Nuclear Antigen/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism
13.
Diabetes ; 55(8): 2212-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16873683

ABSTRACT

Perforin-deficient NOD mice are protected from diabetes, suggesting that cytotoxic granule contents of CD8(+) T-cells have a significant role in killing beta-cells. Despite this, cytotoxic granule effects on human or mouse pancreatic islets have not been reported. We tested the susceptibility of human and mouse islet cells to purified recombinant perforin and granzyme B and measured apoptotic death using a number of assays. Perforin and granzyme B impaired insulin secretion from islet cells, and this was accompanied by cytochrome c release, caspase activation, and DNA fragmentation. Granzyme B-mediated apoptotic changes only occurred in the presence of perforin. When compared with hemopoietic cells, traditionally used as targets to measure cytotoxic T-cell function in vitro, islet cells were relatively resistant to perforin and granzyme B. Inhibition of caspases prevented DNA fragmentation but not cytochrome c release, indicating that mitochondrial disruption due to granzyme B is independent of caspase activation. Consistent with this, islet cells from mice deficient in the BH3-only protein Bid were resistant to cytochrome c release and were protected from apoptosis after exposure to perforin/granzyme B. Our data suggest that Bid cleavage by granzyme B precedes mitochondrial disruption and apoptosis in pancreatic islets.


Subject(s)
Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/physiology , Islets of Langerhans/cytology , Serine Endopeptidases/physiology , Animals , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/deficiency , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cytochromes c/metabolism , DNA Fragmentation , Enzyme Activation/drug effects , Granzymes , Herpesvirus 4, Human , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/physiology , Male , Mastocytoma , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mitochondria/drug effects , Perforin , Pore Forming Cytotoxic Proteins , Recombinant Proteins/pharmacology , Serine Endopeptidases/metabolism , Serine Endopeptidases/pharmacology
14.
J Immunol ; 175(1): 293-301, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15972661

ABSTRACT

In type 1 diabetes, many effector mechanisms damage the beta cell, a key one being perforin/granzyme B production by CD8(+) T cells. The death receptor pathway has also been implicated in beta cell death, and we have therefore generated NOD mice that express a dominant-negative form of the Fas-associated death domain protein (FADD) adaptor to block death receptor signaling in beta cells. Islets developed normally in these animals, indicating that FADD is not necessary for beta cell development as it is for vasculogenesis. beta cells from the transgenic mice were resistant to killing via the Fas pathway in vitro. In vivo, a reduced incidence of diabetes was found in mice with higher levels of dominant-negative FADD expression. This molecule also blocked signals from the IL-1R in culture, protecting isolated islets from the toxic effects of cytokines and also marginally reducing the levels of Fas up-regulation. These data support a role for death receptors in beta cell destruction in NOD mice, but blocking the perforin/granzyme pathway would also be necessary for dominant-negative FADD to have a beneficial clinical effect.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Diabetes Mellitus, Type 1/prevention & control , Islets of Langerhans/metabolism , Membrane Glycoproteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/drug effects , Base Sequence , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Fas Ligand Protein , Fas-Associated Death Domain Protein , Female , Gene Expression , Humans , In Vitro Techniques , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Recombinant Proteins/pharmacology
15.
Int Immunol ; 15(6): 679-90, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12750352

ABSTRACT

Fas and TNFRI, two members of the tumor necrosis factor receptor family with an intracellular death domain, each play critical roles in apoptotic death of lymphocytes and certain other cell types. We determined the overlapping functions of Fas and TNFRI by breeding non-obese diabetic (NOD) mutant mice that lacked both receptors. NODlpr mice developed extensive lymphadenopathy, splenomegaly, CD4(-)CD8(-) B220(+) alpha beta TCR(+) T cells and autoimmune kidney disease. This pathology was not modified by concomitant deficiency in TNFRI as was reported for lpr mice on a B6 background. NODlpr mice lacking CD8(+) T cells, because of a null mutation in beta(2)-microglobulin (beta(2)m), also developed a similar disease profile to NODlpr animals, but the CD4(-)CD8(-) B220(+) alpha beta TCR(+) T cells now derived from a CD4(+) T cell lineage. These results demonstrate that, as in the autoimmune-prone MRL stain, the NOD genetic background promotes lupus nephritis-like pathology and extensive lymphadenopathy when lpr is present. Loss of TNFRI does not exacerbate the pathology caused by deficiency in Fas and loss of beta(2)m does not reduce it.


Subject(s)
Autoimmune Diseases/genetics , Kidney Diseases/genetics , Lymphatic Diseases/genetics , Receptors, Tumor Necrosis Factor/deficiency , beta 2-Microglobulin/deficiency , Animals , Autoimmune Diseases/pathology , Fas Ligand Protein , Female , Flow Cytometry , Kidney Diseases/immunology , Kidney Diseases/pathology , Lymphatic Diseases/immunology , Lymphatic Diseases/pathology , Lymphocyte Activation/genetics , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred NOD , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...