Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
J Immunol ; 191(2): 594-607, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23752610

ABSTRACT

Ptpn22 encodes PEST domain-enriched tyrosine phosphatase (Pep), which negatively regulates TCR proximal signaling and is strongly associated with a variety of autoimmune diseases in humans. The net effect of Pep on the balance of immunity and tolerance is uncertain because of the simultaneous inhibition of TCR-mediated signaling of effector and regulatory T cells (T(regs)). In this study, we generated transgenic NOD mice that overexpressed Pep in T cells. The transgenic mice had a significantly lower incidence of spontaneous autoimmune diabetes, which was accompanied by fewer IFN-γ-producing T cells, and an increased ratio of CD4(+)Foxp3(+) T(regs)to CD4(+)IFN-γ(+) or to CD8(+)IFN-γ(+) T cells, respectively, in pancreatic islets. Transgenic T cells showed markedly decreased TCR-mediated effector cell responses such as proliferation and Th1 differentiation. By contrast, the inhibitory effect of transgenic Pep on TCR signaling did not affect the differentiation of T(regs) or their suppressive activity. Adoptive transfer experiments showed that transgenic splenocytes exhibited attenuated diabetogenic ability. To examine further the pathogenic features of transgenic T cells, we generated Ptpn22/BDC2.5 doubly transgenic mice and found reduced proliferation and Th1 differentiation in CD4(+) T lymphocytes with additional Pep in pancreatic lymph nodes but not in inguinal lymph nodes of NOD/SCID recipients. This finding indicates that transgenic Pep attenuates T cell functions in an islet Ag-driven manner. Taken together, our results demonstrate that Pep overexpression in T cells attenuates autoimmune diabetes in NOD mice by preferentially modulating TCR signaling-mediated functions in diabetogenic T cells but not in T(regs).


Subject(s)
Diabetes Mellitus, Type 1/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Differentiation , Cell Proliferation , Cells, Cultured , Diabetes Mellitus, Type 1/epidemiology , Female , Forkhead Transcription Factors/biosynthesis , Genotype , Incidence , Interferon-gamma/biosynthesis , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Signal Transduction , T-Lymphocytes, Regulatory/metabolism
2.
J Immunol ; 183(4): 2277-85, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19635924

ABSTRACT

T cell-mediated immunodestruction of pancreatic beta cells is the key process responsible for both the development of autoimmune diabetes and the induction of rejection during islet transplantation. In this study, we investigate the hypothesis that transgenic expression of an agonistic, membrane-bound single-chain anti-CTLA-4 Fv (anti-CTLA-4 scFv) on pancreatic beta cells can inhibit autoimmune processes by selectively targeting CTLA-4 on pathogenic T cells. Strikingly, transgenic expression of anti-CTLA-4 scFv on pancreatic beta cells significantly protected NOD mice from spontaneous autoimmune diabetes. Interestingly, local expression of this CTLA-4 agonist did not alter the diabetogenic properties of systemic lymphocytes, because splenocytes from transgenic mice or their nontransgenic littermates equally transferred diabetes in NOD/SCID recipients. By analyzing the T cell development in anti-CTLA-4 scFv/Th1/Th2 triple transgenic mice, we found that beta cell-specific expression of CTLA-4 agonist did not affect the development of Th1/Th2 or CD4(+)CD25(+) regulatory T cells. Most strikingly, islets from transgenic mice inhibited T cell response to immobilized anti-CD3 in a T cell-islet coculture system, suggesting a trans-mediated inhibition provided by transgenic islets. Finally, transgenic islets implanted in diabetic recipients survived much longer than did wild-type islets, indicating a therapeutic potential of this genetically modified islet graft in autoimmune diabetes.


Subject(s)
Antigens, CD/immunology , Autoantibodies/biosynthesis , Autoantibodies/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/prevention & control , Immunoglobulin Fragments/genetics , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Animals , Antigens, CD/metabolism , Autoantibodies/metabolism , Autoantibodies/therapeutic use , Binding Sites, Antibody , CTLA-4 Antigen , Cells, Cultured , Coculture Techniques , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Female , Gene Targeting , Immunoglobulin Fragments/biosynthesis , Immunoglobulin Fragments/physiology , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/physiology , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic
3.
Eur J Immunol ; 39(9): 2403-11, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19670381

ABSTRACT

Galectin-9 (gal-9), widely expressed in many tissues, regulates Th1 cells and induces their apoptosis through its receptor, T-cell Ig mucin 3, which is mainly expressed on terminally differentiated Th1 cells. Type 1 diabetes is a Th1-dominant autoimmune disease that specifically destroys insulin-producing beta cells. To suppress the Th1 immune response in the development of autoimmune diabetes, we overexpressed gal-9 in NOD mice by injection of a plasmid encoding gal-9. Mice treated with gal-9 plasmid were significantly protected from diabetes and showed less severe insulitis compared with controls. Flow cytometric analyses in NOD-T1/2 double transgenic mice showed that Th1-cell population in spleen, pancreatic lymph node and pancreas was markedly decreased in gal-9 plasmid-treated mice, indicating a negative regulatory role of gal-9 in the development of pathogenic Th1 cells. Splenocytes from gal-9 plasmid-treated mice were less responsive to mitogenic stimulation than splenocytes from the control group. However, adoptive transfer of splenocytes from gal-9-treated or control mice caused diabetes in NOD/SCID recipients with similar kinetics, suggesting that gal-9 treatment does not induce active tolerance in NOD mice. We conclude that gal-9 may downregulate Th1 immune response in NOD mice and could be used as a therapeutic target in autoimmune diabetes.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Galectins/metabolism , Receptors, Virus/metabolism , Th1 Cells/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Diabetes Mellitus, Type 1/therapy , Down-Regulation/immunology , Galectins/antagonists & inhibitors , Galectins/genetics , Hepatitis A Virus Cellular Receptor 2 , Mice , Mice, Inbred NOD , Mice, Transgenic , Molecular Sequence Data , Pancreas/immunology , Pancreas/metabolism , Polymorphism, Genetic , Receptors, Virus/genetics , Sequence Alignment , Spleen/immunology , Spleen/metabolism , Th1 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL