Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
J Immunol ; 181(4): 2438-45, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18684934

ABSTRACT

Stimulated by an agonistic ligand, alpha-galactosylceramide (alphaGalCer), invariant NKT (iNKT) cells are capable of both eliciting antitumor responses and suppressing autoimmunity, while they become anergic after an initial phase of activation. It is unknown how iNKT cells act as either activators or regulators in different settings of cellular immunity. We examined effects of alphaGalCer administration on autoimmune inflammation and characterized phenotypes and functional status of iNKT cells and dendritic cells in alphaGalCer-treated NOD mice. Although iNKT cells became and remained anergic after the initial exposure to their ligand, anergic iNKT cells induce noninflammatory DCs in response to alphaGalCer restimulation, whereas activated iNKT cells induce immunogenic maturation of DCs in a small time window after the priming. Induction of noninflammatory DCs results in the activation and expansion of islet-specific T cells with diminished proinflammatory cytokine production. The noninflammatory DCs function at inflammation sites in an Ag-specific fashion, and the persistence of noninflammatory DCs critically inhibits autoimmune pathogenesis in NOD mice. Anergic differentiation is a regulatory event that enables iNKT cells to transform from promoters to suppressors, down-regulating the ongoing inflammatory responses, similar to other regulatory T cells, through a ligand-dependent mechanism.


Subject(s)
Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , Cell Differentiation/immunology , Clonal Anergy , Dendritic Cells/cytology , Dendritic Cells/pathology , Killer Cells, Natural/immunology , T-Lymphocyte Subsets/immunology , Animals , Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/prevention & control , Dose-Response Relationship, Immunologic , Galactosylceramides/administration & dosage , Immunophenotyping , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Ligands , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Self Tolerance/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
2.
Diabetes ; 56(6): 1686-93, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17360980

ABSTRACT

OBJECTIVE: Pregnancy induces a state of immunological tolerance that aims at suppressing immune responses against the fetus and has been linked to temporal remission of preexisting autoimmune disorders. To understand the mechanisms of this reversible immune regulation, we investigated the role of a key pregnancy hormone, human chorionic gonadotropin (hCG), in immune tolerance against autoimmune type 1 diabetes in nonobese diabetic (NOD) mice. RESEARCH DESIGN AND METHODS: We injected hCG into cytokine gene-deficient NOD mice and evaluated the effects of hCG administration on T-cells and dendritic cells (DCs). RESULTS: We show that administration of hCG to NOD mice inhibits both the activation of diabetogenic CD4(+) and CD8(+) T-cells, in vitro and in vivo, and the progression of type 1 diabetes by upregulating the expression of indoleamine 2,3-dioxygenase (IDO) in DCs. IDO upregulation is transient and declined shortly after hCG withdrawal. DC depletion restores the diabetetogenic activity of splenic T-cells from hCG-treated mice, and inhibition of IDO activity by 1-methyl-tryptophan abrogates the hCG-induced T-cell suppression and resistance to type 1 diabetes. CONCLUSIONS: We propose that hCG-induced upregulation of IDO in DCs plays a major role in pregnancy-associated resistance to autoimmunity.


Subject(s)
Chorionic Gonadotropin/deficiency , Chorionic Gonadotropin/pharmacology , Dendritic Cells/enzymology , Diabetes Mellitus, Type 1/prevention & control , Gene Expression Regulation, Enzymologic , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Chorionic Gonadotropin/administration & dosage , Female , Injections, Intraperitoneal , Interleukin-10/deficiency , Interleukin-7/deficiency , Lymphocyte Activation/drug effects , Mice , Mice, Inbred NOD , Mice, Knockout
3.
J Immunol ; 178(5): 2755-62, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17312118

ABSTRACT

A burst release of cytokines by Valpha14 invariant NKT (iNKT) cells upon their TCR engagement critically regulates innate and adaptive immune responses. However, it remains unclear in vivo why iNKT cells respond efficiently to microbial or intracellular lipid Ags that are at low levels or that possess suboptimal antigenicity. We found that dendritic cells (DCs) potentiated iNKT cells to respond to a minimal amount of ligand alpha-galactosylceramide (alphaGalCer) through CD1d-dependent autoreactive responses that require endosomal processing and CD1d trafficking. The ability of potentiation of NKT cells was DC specific and did not depend on costimulatory signals and IL-12 production by DCs. However, DCs that failed to synthesize a major endogenous lipid Ag isoglobotrihexosylceramide were unable to potentiate NKT cells for efficient activation. Further analysis showed that differences in the level and pattern of endogenous lipid Ag presentation differentiate DCs and B cells for effective potentiation and subsequent activation of iNKT cells in the presence of an exogenous Ag. Thus, CD1d-dependent potentiation by DCs may be crucial for iNKT cell-mediated immunity against infectious agents.


Subject(s)
Dendritic Cells/immunology , Galactosylceramides/immunology , Globosides/immunology , Killer Cells, Natural/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Trihexosylceramides/immunology , Animals , Antigen Presentation/immunology , Antigens/immunology , Antigens, CD1/immunology , Antigens, CD1d , B-Lymphocytes/immunology , Endosomes/immunology , Immunity, Cellular , Interleukin-12/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Protein Transport/immunology , Self Tolerance/immunology , Signal Transduction/immunology
4.
J Immunol ; 175(8): 5240-7, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16210629

ABSTRACT

Polymorphism of MHC and MHC-linked genes is tightly associated with susceptibility to type 1 diabetes (T1D) in human and animal models. Despite the extensive studies, however, the role of MHC and MHC-linked genes expressed by T cells on T1D susceptibility remains unclear. Because T cells develop from TCR(-) thymic precursor (pre-T) cells that undergo MHC restriction mediated by thymic stroma cells, we reconstituted the T cell compartment of NOD.scid-RIP-B7.1 mice using pre-T cells isolated from NOD, NOR, AKR, and C57BL/6 (B6) mice. T1D developed rapidly in the mice reconstituted with pre-T cells derived from NOD or NOR donors. In contrast, most of the NOD.scid-RIP-B7.1 mice reconstituted with pre-T cells from AKR or B6 donors were free of T1D. Further analysis revealed that genes within MHC locus of AKR or B6 origin reduced incidence of T1D in the reconstituted NOD.scid-RIP-B7.1 mice. The expression of MHC class I genes of k, but not b haplotype, in T cells conferred T1D resistance. Replacement of an interval near the distal end of the D region in T cells of B6 origin with an identical allele of 129.S6 origin resulted in T1D development in the reconstituted mice. These results provide evidence that the expression of MHC class I and MHC-linked genes in T cells of NOD mice indeed contributes to T1D susceptibility, while expression of specific resistance alleles of MHC or MHC-linked genes in T cells alone would effectively reduce or even prevent T1D.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Genetic Linkage , Genetic Predisposition to Disease , Major Histocompatibility Complex , T-Lymphocytes/immunology , Animals , Dose-Response Relationship, Immunologic , Gene Dosage , Hematopoietic Stem Cell Transplantation , Immunity, Innate/genetics , Mice , Mice, Inbred AKR , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Microsatellite Repeats , T-Lymphocytes/transplantation , beta 2-Microglobulin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...