Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Rev Immunol ; 3(3): 211-22, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12658269

ABSTRACT

Invariant CD1D-restricted natural killer T (iNKT) cells function during innate and adaptive immunity and regulate numerous immune responses, such as autoimmune disease, tumour surveillance, infectious disease and abortions. However, the molecular basis of their functions and the nature of disease-associated defects of iNKT cells are unclear and have been the subject of recent controversy. Here, we review recent findings that underscore the potential importance of interactions between iNKT cells and dendritic cells (DCs) that indicate that iNKT cells regulate DC activity to shape both pro-inflammatory and tolerogenic immune responses. The ability to modulate iNKT-cell activity in vivo using the ligand alpha-galactosylceramide and to treat patients with autoimmune disease or cancer is evaluated also.


Subject(s)
Autoimmune Diseases/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Animals , Antigens, CD1 , Antigens, CD1d , Cell Communication , Dendritic Cells/immunology , Galactosylceramides/pharmacology , Humans , Immunologic Surveillance , Killer Cells, Natural/drug effects , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology
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.
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
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 ; 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
7.
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
8.
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
9.
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
10.
Diabetes ; 51(1): 215-23, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11756344

ABSTRACT

Twenty diabetes susceptibility loci on 12 mouse chromosomes have been identified to control the development of type 1 diabetes at the level of either initiation of insulitis or progression from insulitis to overt diabetes or both. Previously, we demonstrated that the genetic control of T-cell proliferative unresponsiveness in nonobese diabetic (NOD) mice is linked to Idd4 on mouse chromosome 11. Here, we show by congenic mapping of three newly generated NOD.B6Idd4 diabetes-resistant mouse strains that Idd4 is limited to a 5.2-cM interval of chromosome 11. This B6-derived region expressed in NOD.B6Idd4A mice maps between the D11Nds1 (43.8 cM) and D11Mit38/D11Mit325 (49.0 cM) markers and dramatically reduces the development of both insulitis and type 1 diabetes. NOD.B6Idd4B and NOD.B6Idd4C mice, which carry a smaller B6-derived segment of chromosome 11 that spans <5.2 cM distal to D11Nds1, exhibit protection against type 1 diabetes with the restoration of T-cell proliferation. Our findings suggest that diabetes resistance conferred by Idd4 may be mediated by the Idd4.1 and Idd4.2 subloci. Idd4.1 is localized in the D11Nds1 interval that influences both diabetes and insulitis. Idd4.2 is localized within the D11Mit38/325 interval that mainly influences diabetes incidence and restores T-cell proliferative responsiveness. Three potential candidate genes, platelet activating factor acetylhydrolase Ib1, nitric oxide synthase-2, and CC chemokine genes, are localized in the 5.2-cM interval.


Subject(s)
Chromosome Mapping , Diabetes Mellitus, Type 1/genetics , Mice, Inbred NOD/genetics , Aging , Animals , Blood Glucose/analysis , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/physiopathology , Female , Genetic Markers , Genetic Predisposition to Disease/genetics , Incidence , Islets of Langerhans/pathology , Mice , Microsatellite Repeats/genetics
11.
Diabetes ; 52(8): 1967-75, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12882912

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is expressed in different tissues and cells, including pancreas and lymphocytes, and can induce apoptosis in various tumor cells but not in most normal cells. The specific roles of TRAIL in health and disease remain unclear. Here we show by cDNA array analyses that TRAIL gene expression is upregulated in pancreatic islets during the development of autoimmune type 1 diabetes in nonobese diabetic (NOD) mice and in Min6 islet beta-cells activated by TNF-alpha + interferon-gamma. However, stimulation of freshly isolated pancreatic islets or Min6 cells with TRAIL did not induce their apoptosis. TRAIL blockade exacerbates the onset of type 1 diabetes in NOD.Scid recipients of transferred diabetogenic T-cells and in cyclophosphamide-treated NOD mice. TRAIL inhibits the proliferation of NOD diabetogenic T-cells by suppressing interleukin (IL)-2 production and cell cycle progression, and this inhibition can be rescued in the presence of exogenous IL-2. cDNA array and Western blot analyses indicate that TRAIL upregulates the expression of the cdk inhibitor p27(kip1). Our data suggest that TRAIL is an important immune regulator of the development of type 1 diabetes.


Subject(s)
Apoptosis/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/physiopathology , Membrane Glycoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division/immunology , Cell Line , Cyclin-Dependent Kinase Inhibitor p27 , Diabetes Mellitus, Type 1/pathology , Female , Islets of Langerhans/pathology , Islets of Langerhans/physiology , Ligands , Membrane Glycoproteins/antagonists & inhibitors , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , T-Lymphocytes/cytology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
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
13.
J Mol Med (Berl) ; 80(5): 290-300, 2002 May.
Article in English | MEDLINE | ID: mdl-12021841

ABSTRACT

Natural killer T (NKT) cells express phenotypic characteristics shared by conventional natural killer cells and T cells, and reside in several primary and secondary lymphoid as well as nonlymphoid organs. Although these cells possess important effector functions in immunity against cancer and microbial pathogens, their immunoregulatory function has received much recent attention. There is convincing evidence to suggest a regulatory role for these cells in the control of susceptibility to autoimmune disease. NKT cells are reduced in number and function in autoimmune disease prone mice and humans. Studies conducted in mice have shown that transfer of NKT cells to disease-susceptible recipients prevents the development of autoimmune disease. The recent discovery that alpha-galactosylceramide, a glycolipid, can specifically target NKT cells expressing the invariant T cell receptor (TCR) to proliferate and produce an array of regulatory cytokines and chemokines has generated considerable interest to utilize these cells as targets of new therapeutic interventions for the immunoregulation of autoimmune disease


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Killer Cells, Natural/immunology , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Homeostasis/immunology , Humans
14.
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
15.
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
16.
Ann N Y Acad Sci ; 958: 77-88, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12021086

ABSTRACT

Autoimmune diseases, especially type 1 diabetes (T1D), may be caused by dysregulation of the immune system, which leads to hyporesponsiveness of regulatory T helper 2 (Th2) cells and promotion of autoimmune Th1 cells. Natural killer T (NKT) cells, which comprise a minor subpopulation of T cells, play a critical role in immunoregulation as a result of a rapid burst of IL-4 and IFN-gamma secretion. These cells are functionally and numerically deficient in individuals at risk of T1D, as well as in nonobese diabetic (NOD) mice. It is conceivable that protection from T1D may be achieved by correction of this deficiency. Alpha-galactosylceramide (alpha-GalCer) specifically binds to NKT cells in a CD1-dependent manner and stimulates these cells to proliferate and to produce various cytokines, including IFN-gamma, IL-4, and IL-10. In this review, we present evidence that a multiple-dose alpha-GalCer treatment regimen, which is known to promote a dominant Th2 environment, can prevent the onset of spontaneous and cyclophosphamide (CY)-accelerated T1D. This protection is associated with elevated IL-4 and IL-10 in the spleen and pancreas of protected female NOD mice. Concomitantly, IFN-gamma levels are reduced in both tissues. More importantly, the protective effect of gamma-GalCer in CY-accelerated T1D is abrogated by the in vivo blockade of IL-10 activity. We also show that alpha-GalCer treatment significantly prolongs syngeneic islet graft survival in recipient diabetic NOD mice. These findings raise the possibility that alpha-GalCer treatment may be used therapeutically to prevent the onset and recurrence of human T1D.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Animals , Autoimmunity/immunology , Cell Differentiation/drug effects , Cell Division , Diabetes Mellitus, Type 1/drug therapy , Galactosylceramides/therapeutic use , Humans , Killer Cells, Natural/cytology , Recurrence , T-Lymphocytes/cytology , Transplantation Immunology/immunology
18.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL