Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
2.
Immunology ; 142(3): 321-36, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24428389

ABSTRACT

Natural killer T cells (NKT) can regulate innate and adaptive immune responses. Type I and type II NKT cell subsets recognize different lipid antigens presented by CD1d, an MHC class-I-like molecule. Most type I NKT cells express a semi-invariant T-cell receptor (TCR), but a major subset of type II NKT cells reactive to a self antigen sulphatide use an oligoclonal TCR. Whereas TCR-α dominates CD1d-lipid recognition by type I NKT cells, TCR-α and TCR-ß contribute equally to CD1d-lipid recognition by type II NKT cells. These variable modes of NKT cell recognition of lipid-CD1d complexes activate a host of cytokine-dependent responses that can either exacerbate or protect from disease. Recent studies of chronic inflammatory and autoimmune diseases have led to a hypothesis that: (i) although type I NKT cells can promote pathogenic and regulatory responses, they are more frequently pathogenic, and (ii) type II NKT cells are predominantly inhibitory and protective from such responses and diseases. This review focuses on a further test of this hypothesis by the use of recently developed techniques, intravital imaging and mass cytometry, to analyse the molecular and cellular dynamics of type I and type II NKT cell antigen-presenting cell motility, interaction, activation and immunoregulation that promote immune responses leading to health versus disease outcomes.


Subject(s)
Killer Cells, Natural/immunology , T-Lymphocyte Subsets/immunology , Animals , Disease , Health , Humans , Killer Cells, Natural/metabolism , T-Lymphocyte Subsets/metabolism
3.
PLoS One ; 7(5): e37771, 2012.
Article in English | MEDLINE | ID: mdl-22649557

ABSTRACT

Sulfatide-reactive type II NKT cells have been shown to regulate autoimmunity and anti-tumor immunity. Although, two major isoforms of sulfatide, C16:0 and C24:0, are enriched in the pancreas, their relative role in autoimmune diabetes is not known. Here, we report that sulfatide/CD1d-tetramer(+) cells accumulate in the draining pancreatic lymph nodes, and that treatment of NOD mice with sulfatide or C24:0 was more efficient than C16:0 in stimulating the NKT cell-mediated transfer of a delay in onset from T1D into NOD.Scid recipients. Using NOD.CD1d(-/-) mice, we show that this delay of T1D is CD1d-dependent. Interestingly, the latter delay or protection from T1D is associated with the enhanced secretion of IL-10 rather than IFN-g by C24:0-treated CD4(+) T cells and the deviation of the islet-reactive diabetogenic T cell response. Both C16:0 and C24:0 sulfatide isoforms are unable to activate and expand type I iNKT cells. Collectively, these data suggest that C24:0 stimulated type II NKT cells may regulate protection from T1D by activating DCs to secrete IL-10 and suppress the activation and expansion of type I iNKT cells and diabetogenic T cells. Our results raise the possibility that C24:0 may be used therapeutically to delay the onset and protect from T1D in humans.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Mice, Inbred NOD/metabolism , Natural Killer T-Cells/metabolism , Sulfoglycosphingolipids/pharmacology , Animals , Antigens, CD1d/genetics , CD4-Positive T-Lymphocytes/metabolism , Flow Cytometry , Interleukin-10/metabolism , Interleukin-2/metabolism , Interleukin-4/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Knockout , Pancreas/cytology , Structure-Activity Relationship
4.
Immunology ; 132(3): 361-75, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21070234

ABSTRACT

Invariant natural killer T (iNKT) cells are a small subset of lymphocytes that recognize glycolipid antigens in the context of CD1d and consequently produce large quantities of pro-inflammatory and/or anti-inflammatory cytokines. Several transmembrane glycoproteins have been implicated in the co-stimulation of iNKT cell responses. However, whether glycosylphosphatidylinositol (GPI)-anchored proteins can function in this capacity is not known. Here, we demonstrate that antibody-mediated cross-linking of the prototype mouse GPI-anchored protein Thy-1 (CD90) on the surface of a double-negative (CD4⁻CD8⁻) iNKT cell line leads to cytokine production at both the mRNA and protein levels. In addition, Thy-1 triggering enhanced cytokine secretion by iNKT cells that were concomitantly stimulated with α-galactosylceramide (αGC), consistent with a co-stimulatory role for Thy-1 in iNKT cell activation. This was also evident when a CD4+ mouse iNKT cell line or primary hepatic NKT cells were stimulated with αGC and/or anti-Thy-1 antibody. Cross-linking Ly-6A/E, another GPI-anchored protein, could also boost cytokine secretion by αGC-stimulated iNKT cells, suggesting that the observed effects reflect a general property of GPI-anchored proteins. To extend these results from mouse to human cells, we focused on CD55, a GPI-anchored protein that, unlike Thy-1, is expressed on human iNKT cells. Cross-linking CD55 augmented αGC-induced iNKT cell responses as judged by more vigorous proliferation and higher CD69 expression. Collectively, these findings demonstrate for the first time that GPI-anchored proteins are able to co-stimulate CD1d-restricted, glycolipid-reactive iNKT cells in both mice and humans.


Subject(s)
Glycosylphosphatidylinositols/immunology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Animals , Cell Separation , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Male , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
5.
Diabetes ; 59(11): 2862-71, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20693344

ABSTRACT

OBJECTIVE: The progressive infiltration of pancreatic islets by lymphocytes is mandatory for development of autoimmune type 1 diabetes. This inflammatory process is mediated by several mediators that are potential therapeutic targets to arrest development of type 1 diabetes. In this study, we investigate the role of one of these mediators, interleukin-16 (IL-16), in the pathogenesis of type 1 diabetes in NOD mice. RESEARCH DESIGN AND METHODS: At different stages of progression of type 1 diabetes, we characterized IL-16 in islets using GEArray technology and immunoblot analysis and also quantitated IL-16 activity in cell migration assays. IL-16 expression was localized in islets by immunofluorescence and confocal imaging. In vivo neutralization studies were performed to assess the role of IL-16 in the pathogenesis of type 1 diabetes. RESULTS: The increased expression of IL-16 in islets correlated with the development of invasive insulitis. IL-16 immunoreactivity was found in islet infiltrating T-cells, B-cells, NK-cells, and dendritic cells, and within an insulitic lesion, IL-16 was derived from infiltrating cells. CD4(+) and CD8(+) T-cells as well as B220(+) B-cells were identified as sources of secreted IL-16. Blockade of IL-16 in vivo protected against type 1 diabetes by interfering with recruitment of CD4(+) T-cells to the pancreas, and this protection required the activity of the chemokine CCL4. CONCLUSIONS: IL-16 production by leukocytes in islets augments the severity of insulitis during the onset of type 1 diabetes. IL-16 and CCL4 appear to function as counterregulatory proteins during disease development. Neutralization of IL-16 may represent a novel therapy for the prevention of type 1 diabetes.


Subject(s)
Carbon Tetrachloride/pharmacology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Interleukin-16/physiology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Movement , Diabetes Mellitus, Type 1/physiopathology , Hyperinsulinism/physiopathology , Immunoblotting , Interleukin-16/genetics , Islets of Langerhans/immunology , Islets of Langerhans/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Pancreas/physiology , Pancreas/physiopathology , RNA, Messenger/genetics , T-Lymphocytes/immunology
6.
J Immunol ; 179(11): 7225-32, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18025164

ABSTRACT

Although B cells play a pathogenic role in the initiation of type 1 diabetes (T1D) in NOD mice, it is not known whether activated B cells can maintain tolerance and transfer protection from T1D. In this study, we demonstrate that i.v. transfusion of BCR-stimulated NOD spleen B cells into NOD mice starting at 5-6 wk of age both delays onset and reduces the incidence of T1D, whereas treatment initiated at 9 wk of age only delays onset of T1D. This BCR-activated B cell-induced protection from T1D requires IL-10 production by B cells, as transfusion of activated B cells from NOD.IL-10(-/-) mice does not confer protection from T1D. Consistent with this result, severe insulitis was observed in the islets of NOD recipients of transfused NOD.IL-10(-/-) BCR-stimulated B cells but not in the islets of NOD recipients of transfused BCR-stimulated NOD B cells. The therapeutic effect of transfused activated NOD B cells correlates closely with the observed decreased islet inflammation, reduced IFN-gamma production and increased production of IL-4 and IL-10 by splenocytes and CD4(+) T cells from NOD recipients of BCR-stimulated NOD B cells relative to splenocytes and CD4(+) T cells from PBS-treated control NOD mice. Our data demonstrate that transfused BCR-stimulated B cells can maintain long-term tolerance and protect NOD mice from T1D by an IL-10-dependent mechanism, and raise the possibility that i.v. transfusion of autologous IL-10-producing BCR-activated B cells may be used therapeutically to protect human subjects at risk for T1D.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/transplantation , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Interleukin-10/physiology , Receptors, Antigen, B-Cell/metabolism , Animals , B-Lymphocytes/cytology , Female , Injections, Intravenous , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Mice , Mice, Inbred NOD , Mice, SCID , Transplantation Immunology/immunology
8.
Diabetes ; 56(3): 809-17, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17327452

ABSTRACT

We previously reported that interleukin (IL)-4 treatment of nonobese diabetic (NOD) mice elevates intrapancreatic CCL4 expression and protects from type 1 diabetes. Here, we show that antibody neutralization of CCL4 abrogates the ability of T-cells from IL-4-treated NOD mice to transfer protection against type 1 diabetes. Intradermal delivery of CCL4 via a plasmid vector stabilized by incorporation of the Epstein-Barr virus EBNA1/oriP episomal maintenance replicon (pHERO8100-CCL4) to NOD mice beginning at later stages of disease progression protects against type 1 diabetes. This protection was associated with a Th2-like response in the spleen and pancreas; decreased recruitment of activated CD8(+) T-cells to islets, accompanied by diminished CCR5 expression on CD8(+) T-cells; and regulatory T-cell activity in the draining pancreatic lymph nodes. Thus, inflammatory responses that target islet beta-cells are suppressed by CCL4, which implicates the use of CCL4 therapeutically to prevent type 1 diabetes.


Subject(s)
Chemokines, CC/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/prevention & control , Insulin-Secreting Cells/pathology , Aging , Animals , Chemokine CCL4 , Chemokines, CC/genetics , Diabetes Mellitus, Type 1/metabolism , Genetic Therapy , Inflammation/prevention & control , Interleukin-4/immunology , Interleukin-4/pharmacology , Islets of Langerhans Transplantation , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation
9.
Ann N Y Acad Sci ; 1079: 147-52, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17130546

ABSTRACT

We recently mapped Idd4 to a 5.2 cM interval on chromosome 11 with two subloci, Idd4.1 and Idd4.2, in nonobese diabetic (NOD) mice. Based on the localization of platelet-activating factor acetylhydrolase Ib1 (PAF-AHIb1) and the decreased activity of PAF-AH in type 1 diabetes (T1D) patients, we hypothesized that PAF-AHIb1 in Idd4.1 is a candidate gene. The PAF-AHIb1 gene in NOD mice was cloned and sequenced, and its expression and function were studied. No polymorphisms were detected in PAF-AHIb1 cDNA between NOD and B6 mice. The expression of PAF-AH Ib1 at the mRNA and protein levels was found to be similar in different tissues between NOD and B6 mice. PAF-AH activity does not differ in the pancreatic islets or spleen between NOD and B6 mice. Our findings suggest that PAF-AH Ib1 may not be a diabetes-susceptibility gene in the Idd4.1 sublocus.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Chromosome Mapping , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Genetic Predisposition to Disease , Animals , Diabetes Mellitus, Type 1/immunology , Genetic Markers , Mice , Mice, Inbred NOD , Mice, Mutant Strains , Mice, SCID
10.
J Immunol ; 177(6): 3695-704, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16951329

ABSTRACT

Invariant NK T (iNKT) cells regulate immune responses, express NK cell markers and an invariant TCR, and recognize lipid Ags in a CD1d-restricted manner. Previously, we reported that activation of iNKT cells by alpha-galactosylceramide (alpha-GalCer) protects against type 1 diabetes (T1D) in NOD mice via an IL-4-dependent mechanism. To further investigate how iNKT cells protect from T1D, we analyzed whether iNKT cells require the presence of another subset(s) of regulatory T cells (Treg), such as CD4+ CD25+ Treg, for this protection. We found that CD4+ CD25+ T cells from NOD.CD1d(-/-) mice deficient in iNKT cell function similarly in vitro to CD4+ CD25+ T cells from wild-type NOD mice and suppress the proliferation of NOD T responder cells upon alpha-GalCer stimulation. Cotransfer of NOD diabetogenic T cells with CD4+ CD25+ Tregs from NOD mice pretreated with alpha-GalCer demonstrated that activated iNKT cells do not influence the ability of T(regs) to inhibit the transfer of T1D. In contrast, protection from T1D mediated by transfer of activated iNKT cells requires the activity of CD4+ CD25+ T cells, because splenocytes pretreated with alpha-GalCer and then inactivated by anti-CD25 of CD25+ cells did not protect from T1D. Similarly, mice inactivated of CD4+ CD25+ T cells before alpha-GalCer treatment were also not protected from T1D. Our data suggest that CD4+ CD25+ T cells retain their function during iNKT cell activation, and that the activity of CD4+ CD25+ Tregs is required for iNKT cells to transfer protection from T1D.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Killer Cells, Natural/immunology , Receptors, Interleukin-2/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, SCID , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
12.
Nat Med ; 12(6): 624-6, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16732280

ABSTRACT

The mechanism underlying the autoimmune polyglandular syndrome type-1 (APS1) has been attributed to defective T-cell negative selection resulting from reduced expression and presentation of autoantigens in thymic medullary epithelial cells (MECs). It has also been postulated that Aire is involved in development of regulatory T cells, although supporting evidence is lacking. Here we show that expression of Aire in MECs is required for development of iNKT cells, suggesting a role for iNKT cells in APS1.


Subject(s)
Killer Cells, Natural/physiology , Polyendocrinopathies, Autoimmune/immunology , T-Lymphocyte Subsets/physiology , Transcription Factors/immunology , Animals , Antigens, CD/immunology , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Liver/cytology , Mice , Mice, Knockout , Spleen/cytology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Transcription Factors/genetics , AIRE Protein
13.
Immunol Res ; 31(3): 177-88, 2005.
Article in English | MEDLINE | ID: mdl-15888910

ABSTRACT

Invariant CD1d-restricted natural killer T (iNKT) cells function during innate and adaptive immune responses. A functional and numerical deficiency of iNKT cells is well documented in both nonobese diabetic (NOD) mice and humans with autoimmune type 1 diabetes (T1D). Restoring the numerical and/or functional deficiency of iNKT cells in NOD mice by either treatment with alpha-galactosylceramide, transgenic induction of Valpha14-Jalpha18 expression, or transgenic expression of CD1d in NOD islets under the control of the human insulin promoter confers protection from T1D in these mice. Recently, considerable progress has been made in understanding the developmental and functional activities of iNKT cells. In this review, we discuss the role of iNKT cell deficiency and defective development in the onset of T1D in NOD mice and the different protective mechanisms known to restore these defects.


Subject(s)
Antigens, CD1/metabolism , Diabetes Mellitus, Type 1/immunology , Killer Cells, Natural/immunology , Th2 Cells/immunology , Animals , Antigens, CD1d , Cytokines/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Subsets/immunology , Mice , Mice, Inbred NOD , Promoter Regions, Genetic
14.
J Immunol ; 174(11): 6764-71, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15905517

ABSTRACT

A numerical and functional deficiency in invariant NKT (iNKT) cells detectable by 3 wk of age in the thymus and spleen mediates the pathogenesis of type 1 diabetes in NOD mice, but the stage of T cell development at which this deficiency first occurs is unknown. We report in this study that this deficiency develops after the CD4(+)CD8(+) double-positive stage of thymic T cell development and is due to a lineage-specific depletion of CD4(-)CD8(-) double-negative alphabeta T cells and iNKT cells from the thymus between embryonic day 18 and day 1 after birth. Thus, an inheritable defect in a lineage fate decision that elicits a deficiency in fetal thymic iNKT cell development may predispose to susceptibility to type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Fetus/immunology , Genetic Predisposition to Disease , Killer Cells, Natural/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Diabetes Mellitus, Type 1/genetics , Female , Fetus/pathology , Intracellular Signaling Peptides and Proteins , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymphopenia/genetics , Lymphopenia/immunology , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Organ Culture Techniques , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Thymus Gland/metabolism , Thymus Gland/pathology
15.
J Immunol ; 174(2): 680-7, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15634886

ABSTRACT

Little is known about the pathogenic role of B cell dysfunction in T cell-mediated autoimmune disease. We previously reported that B cell hyper-responsiveness, resistance to apoptosis, and accumulation in islets occur during the onset of insulitis, but not in type 1 diabetes (T1D), in NOD mice. In this study we extended these studies to further determine how islet-infiltrated B cells contribute to this inflammatory insulitis. We demonstrate the presence of an increased percentage of B7-1(+) and a decreased percentage of B7-2(+) B cells in the spleen of autoimmune disease-prone NOD and nonobese diabetes-resistant mice compared with the spleen of nonautoimmune disease-prone C57BL/6 and BALB/c mice. An age-dependent differential expression of B7-1 and B7-2 was associated with the development of insulitis and CD4(+)CD25(+) T cell deficiency in autoimmune disease-prone mice. Whereas BCR and LPS stimulation increased B7-2 expression on B cells from autoimmune disease-prone and nonautoimmune disease-prone mice, LPS-induced B7-1 expression was higher on NOD than C57BL/6 B cells. Interestingly, increased expression of B7-1 and B7-2 was found on islet-infiltrated B cells, and this increase was associated with enhanced T cell costimulation. Islet-infiltrated B cells were shown to be a source of TNF-alpha production in islets. B7 blockade of BCR-stimulated NOD B cells by anti-B7-1 and anti-B7-2 mAbs during coadoptive transfer with diabetogenic T cells into NOD.scid mice protected these recipients from T1D. These results suggest that increased B7-1 and B7-2 expression on islet-infiltrated NOD B cells is associated with increased T cell costimulation and the development of inflammatory insulitis in NOD mice.


Subject(s)
Antigens, CD/biosynthesis , B-Lymphocyte Subsets/immunology , B7-1 Antigen/biosynthesis , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/pathology , Lymphocyte Activation/immunology , Membrane Glycoproteins/biosynthesis , T-Lymphocyte Subsets/immunology , Up-Regulation/immunology , Adoptive Transfer , Animals , Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Blocking/pharmacology , Antigens, CD/immunology , Antigens, CD/physiology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/pathology , B-Lymphocyte Subsets/transplantation , B7-1 Antigen/immunology , B7-1 Antigen/physiology , B7-2 Antigen , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Movement/immunology , Cell Proliferation , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Female , Genetic Predisposition to Disease , Immunoglobulin Fab Fragments/pharmacology , Islets of Langerhans/immunology , Lymphopenia/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Receptors, Interleukin-2/biosynthesis , Spleen/immunology , Spleen/pathology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Tumor Necrosis Factor-alpha/biosynthesis
16.
J Immunol ; 173(7): 4407-16, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15383571

ABSTRACT

Activation-induced cell death (AICD) plays a key role in the homeostasis of the immune system. Autoreactive T cells are eliminated through AICD both from the thymus and periphery. In this study, we show that NOD peripheral T cells, especially CD8(+) T cells, display a decreased susceptibility to anti-CD3-induced AICD in vivo compared with T cells from diabetes-resistant B6, nonobese diabetes-resistant, and NOD.B6Idd4 mice. The susceptibility of NOD CD8(+) T cells to AICD varies in an age- and dose-dependent manner upon stimulation in vivo with either a mitogenic or nonmitogenic anti-CD3. NOD T cells preactivated by anti-CD3 in vivo are less susceptible than B6 T cells to TCR-induced AICD. Treatment of NOD mice with a mitogenic anti-CD3 depletes CD4(+)CD25(-)CD62L(+) but not CD4(+)CD25(+)CD62L(+) T cells, thereby resulting in an increase of the latter subset in the spleen. Treatment with a nonmitogenic anti-CD3 mAb delays the onset of T1D in 8.3 TCR transgenic NOD mice. These results demonstrate that the capacity of anti-CD3 to protect NOD mice from T1D correlates with its ability to perturb T cell homeostasis by inducing CD8(+) T cell AICD and increasing the number of CD4(+)CD25(+)CD62L(+) T cells in the periphery.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/immunology , CD3 Complex/immunology , Diabetes Mellitus, Type 1/immunology , Homeostasis/immunology , Prediabetic State/immunology , T-Lymphocyte Subsets/immunology , Aging/genetics , Aging/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Apoptosis/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Death/genetics , Cell Death/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/prevention & control , Female , Genetic Predisposition to Disease , Homeostasis/genetics , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/metabolism , Interleukin-10/antagonists & inhibitors , Interleukin-10/metabolism , Interleukin-4/antagonists & inhibitors , Interleukin-4/metabolism , L-Selectin/biosynthesis , Lymphocyte Activation/genetics , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Prediabetic State/genetics , Prediabetic State/pathology , Prediabetic State/prevention & control , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Interleukin-2/biosynthesis , Spleen/cytology , Spleen/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
17.
Diabetes ; 53(8): 2003-11, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277379

ABSTRACT

B-cells proliferate after B-cell receptor (BCR) stimulation and are deleted by activation-induced cell death (AICD) during negative selection. We report that B-cells from type 1 diabetes-susceptible NOD and type 1 diabetes-resistant but insulitis-prone congenic NOD.B6Idd4B and NOR mice, relative to B-cells from nonautoimmune disease-prone C57BL/6 and BALB/c mice, display a hyperproliferative response to BCR stimulation and lower activation threshold in the absence or presence of interleukin 4 (IL-4). This hyperproliferation is associated with an increased proportion of NOD and NOR B-cells that enter into the S phase of the cell cycle and undergo cell division. The relative resistance to BCR-induced AICD of B-cells from NOD, NOR, and NOD.B6Idd4B mice, all of which develop insulitis, correlates with the presence of a higher percentage of hyperactivated B-cells in the spleen and islets of these mice than in nonautoimmune disease-prone C57BL/6 and BALB/c mice. The NOD islet-infiltrated activated B-cells are more responsive to further stimulation by IL-4 than activated spleen B-cells. Our results suggest that resistance to AICD and accumulation of hyperactivated B-cells in islets is associated with the onset of an inflammatory insulitis, but not type 1 diabetes.


Subject(s)
B-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Apoptosis , B-Lymphocytes/cytology , Cell Cycle/immunology , Cell Division , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Spleen/immunology
18.
Diabetes ; 53(5): 1303-10, 2004 May.
Article in English | MEDLINE | ID: mdl-15111500

ABSTRACT

In nonobese diabetic (NOD) mice, a deficiency in the number and function of invariant natural killer T-cells (iNKT cells) contributes to the onset of type 1 diabetes. The activation of CD1d-restricted iNKT cells by alpha-galactosylceramide (alpha-GalCer) corrects these deficiencies and protects against spontaneous and recurrent type 1 diabetes. Although interleukin (IL)-4 and IL-10 have been implicated in alpha-GalCer-induced protection from type 1 diabetes, a precise role for these cytokines in iNKT cell regulation of susceptibility to type 1 diabetes has not been identified. Here we use NOD.IL-4(-/-) and NOD.IL-10(-/-) knockout mice to further evaluate the roles of IL-4 and IL-10 in alpha-GalCer-induced protection from type 1 diabetes. We found that IL-4 but not IL-10 expression mediates protection against spontaneous type 1 diabetes, recurrent type 1 diabetes, and prolonged syngeneic islet graft function. Increased transforming growth factor-beta gene expression in pancreatic lymph nodes may be involved in alpha-GalCer-mediated protection in NOD.IL-10(-/-) knockout mice. Unlike the requirement of IL-7 and IL-15 to maintain iNKT cell homeostasis, IL-4 and IL-10 are not required for alpha-GalCer-induced iNKT cell expansion and/or survival. Our data identify an important role for IL-4 in the protection against type 1 diabetes by activated iNKT cells, and these findings have important implications for cytokine-based therapy of type 1 diabetes and islet transplantation.


Subject(s)
Antigens, CD1/analysis , Diabetes Mellitus, Type 1/prevention & control , Interleukin-10/metabolism , Interleukin-4/metabolism , Killer Cells, Natural/immunology , Lymphocyte Activation , Animals , Antigens, CD1d , Cyclophosphamide , Cytokines/genetics , Diabetes Mellitus, Type 1/chemically induced , Galactosylceramides/pharmacology , Gene Expression , Gene Expression Profiling , Graft Survival/drug effects , Islets of Langerhans Transplantation , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Oligonucleotide Array Sequence Analysis , Protein Isoforms/pharmacology , Secondary Prevention
19.
Endocrinology ; 145(2): 627-38, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14617576

ABSTRACT

IGF-I regulates islet beta-cell growth, survival, and metabolism and protects against type 1 diabetes (T1D). However, the therapeutic efficacy of free IGF-I may be limited by its biological half-life in vivo. We investigated whether prolongation of its half-life as an IGF-I/IGF binding protein (IGFBP)-3 complex affords increased protection against T1D and whether this occurs by influencing T cell function and/or islet beta-cell growth and survival. Administration of IGF-I either alone or as an IGF-I/IGFBP-3 complex reduced the severity of insulitis and delayed the onset of T1D in nonobese diabetic mice, but IGF-I/IGFBP-3 was significantly more effective. Protection from T1D elicited by IGF-I/IGFBP-3 was mediated by up-regulated CCL4 and down-regulated CCL3 gene expression in pancreatic draining lymph nodes, activation of the phosphatidylinositol 3-kinase and Akt/protein kinase B signaling pathway of beta-cells, reduced beta-cell apoptosis, and stimulation of beta-cell replication. Reduced beta-cell apoptosis resulted from elevated Bcl-2 and Bcl-X(L) activity and diminished caspase-9 activity, indicating a novel role for a mitochondrial-dependent pathway of beta-cell death. Thus, IGF-I/IGFBP-3 affords more efficient protection from insulitis, beta-cell destruction, and T1D than IGF-I, and this complex may represent an efficacious therapeutic treatment for the prevention of T1D.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Insulin-Like Growth Factor Binding Protein 3/administration & dosage , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/metabolism , Animals , Apoptosis/drug effects , Caspase 9 , Caspases/metabolism , Cell Division , Chemokines/genetics , Diabetes Mellitus, Type 1/pathology , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Inflammation/prevention & control , Islets of Langerhans/enzymology , Islets of Langerhans/pathology , Lymph Nodes/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Pancreas , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , T-Lymphocytes
20.
Novartis Found Symp ; 252: 146-60; discussion 160-4, 203-10, 2003.
Article in English | MEDLINE | ID: mdl-14609217

ABSTRACT

Deficiencies in NKT cell number and function mediate the development of Type 1 diabetes (TID). NKT cell activation with the CD1d ligand alpha-galactosylceramide (alpha-GalCer) corrects these deficiencies and prevents the onset and recurrence of T1D in NOD mice. To investigate how alpha-GalCer accomplishes this, we conducted three sets of studies. First, gene microarray analyses showed that alpha-GalCer treatment decreases interleukin (IL)16 and increases IL10 and MIP1beta gene expression in the spleen. Anti-IL16 antibody treatment protects NOD mice against insulitis and T1D, and neutralization of MIP1beta abrogates IL4 induced protection from T1D. Second, alpha-GalCer treatment of NOD.ILA(-/-) mice demonstrated that IL4 expression is required for prevention of T1D but not for NKT cell development. Third, we found that diabetes resistance in three novel congenic NOD.B6Idd4 mouse strains is associated with an increased number of NKT cells in pancreatic lymph nodes (PLNs). This increase was not evident in the spleen or PLNs of NOD.MIP1a(-/-) mice after alpha-GalCer treatment. Our data suggest that MIP1beta is a candidate gene in Idd4 that regulates NKT cell function and diabetes susceptibility. By controlling the expression and activity of IL16 and MIP1beta alpha-GalCer treatment may modulate the number, localization and function of NKT cells and regulate susceptibility to T1D.


Subject(s)
Antigens, CD1/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Killer Cells, Natural/immunology , Adoptive Transfer , Animals , Antigens, CD1d , Chemokines/genetics , Diabetes Mellitus, Type 1/genetics , Gene Expression Regulation/immunology , Humans , Mice , Mice, Inbred NOD , Receptors, Chemokine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...