Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 9(9): e107935, 2014.
Article in English | MEDLINE | ID: mdl-25268801

ABSTRACT

Combining immune intervention with therapies that directly influence the functional state of the ß-cells is an interesting strategy in type 1 diabetes cure. Dipeptidyl peptidase-4 (DPP-4) inhibitors elevate circulating levels of active incretins, which have been reported to enhance insulin secretion and synthesis, can support ß-cell survival and possibly stimulate ß-cell proliferation and neogenesis. In the current study, we demonstrate that the DPP-4 inhibitor MK626, which has appropriate pharmacokinetics in mice, preceded by a short-course of low-dose anti-CD3 generated durable diabetes remission in new-onset diabetic non-obese diabetic (NOD) mice. Induction of remission involved recovery of ß-cell secretory function with resolution of destructive insulitis and preservation of ß-cell volume/mass, along with repair of the islet angioarchitecture via SDF-1- and VEGF-dependent actions. Combination therapy temporarily reduced the CD4-to-CD8 distribution in spleen although not in pancreatic draining lymph nodes (PLN) and increased the proportion of effector/memory T cells as did anti-CD3 alone. In contrast, only combination therapy amplified Foxp3+ regulatory T cells in PLN and locally in pancreas. These findings open new opportunities for the treatment of new-onset type 1 diabetes by introducing DPP-4 inhibitors in human CD3-directed clinical trials.


Subject(s)
Antibodies, Monoclonal/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Spleen/drug effects , Animals , CD3 Complex/genetics , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cricetinae , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Drug Synergism , Drug Therapy, Combination , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred NOD , Pancreas/drug effects , Pancreas/immunology , Pancreas/pathology , Remission Induction , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
2.
J Immunol ; 192(9): 4210-20, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24663679

ABSTRACT

The biologically active form of vitamin D, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], is able to promote the generation of tolerogenic mature dendritic cells (mDCs) with an impaired ability to activate autoreactive T cells. These cells could represent a reliable tool for the promotion or restoration of Ag-specific tolerance through vaccination strategies, for example in type 1 diabetes patients. However, successful transfer of 1,25(OH)2D3-treated mDCs (1,25D3-mDCs) depends on the capacity of 1,25(OH)2D3 to imprint a similar tolerogenic profile in cells derived from diabetes-prone donors as from diabetes-resistant donors. In this study, we examined the impact of 1,25(OH)2D3 on the function and phenotype of mDCs originating from healthy (C57BL/6) and diabetes-prone (NOD) mice. We show that 1,25(OH)2D3 is able to imprint a phenotypic tolerogenic profile on DCs derived from both mouse strains. Both NOD- and C57BL/6-derived 1,25D3-mDCs decreased the proliferation and activation of autoreactive T cells in vitro, despite strain differences in the regulation of cytokine/chemokine expression. In addition, 1,25D3-mDCs from diabetes-prone mice expanded CD25(+)Foxp3(+) regulatory T cells and induced intracellular IL-10 production by T cells in vitro. Furthermore, 1,25D3-mDCs exhibited an intact functional migratory capacity in vivo that favors homing to the liver and pancreas of adult NOD mice. More importantly, when cotransferred with activated CD4(+) T cells into NOD.SCID recipients, 1,25D3-mDCs potently dampened the proliferation of autoreactive donor T cells in the pancreatic draining lymph nodes. Altogether, these results argue for the potential of 1,25D3-mDCs to restore Ag-specific immune tolerance and arrest autoimmune disease progression in vivo.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/drug effects , Vitamin D/analogs & derivatives , Animals , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Immune Tolerance/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Phenotype , Real-Time Polymerase Chain Reaction , Vitamin D/pharmacology
3.
Diabetes ; 63(6): 2026-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24550187

ABSTRACT

High doses of the active form of vitamin D3, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], prevent diabetes in the NOD mouse but also elicit unwanted calcemic side effects. Because immune cells themselves can convert vitamin D3 into 1,25(OH)2D3 locally, we hypothesized that dietary vitamin D3 can also prevent disease. Thus, we evaluated whether dietary administration of high doses of regular vitamin D3 (800 IU/day) during different periods of life (pregnancy and lactation, early life [3-14 weeks of age], or lifelong [3-35 weeks of age]) safely prevents diabetes in NOD mice. We found that only lifelong treatment raised serum 25-hydroxyvitamin D3 from 173 nmol/L in controls to 290 nmol/L, without inducing signs of calcemic or bone toxicity, and significantly reduced diabetes development in both male and female NOD mice. This diabetes protection by vitamin D3 correlated with preserved pancreatic insulin content and improved insulitis scores. Moreover, vitamin D3 treatment decreased interferon-γ-positive CD8(+) T cells and increased CD4(+)(CD25(+))FoxP3(+) T cells in pancreatic draining lymph nodes. In conclusion, this study shows for the first time that high doses of regular dietary vitamin D3 can safely prevent diabetes in NOD mice when administered lifelong, although caution is warranted with regards to administering equivalently high doses in humans.


Subject(s)
Cholecalciferol/administration & dosage , Cholecalciferol/pharmacology , Diabetes Mellitus, Type 1/prevention & control , Vitamins/administration & dosage , Vitamins/pharmacology , Animals , Diabetes Mellitus, Type 1/immunology , Dietary Supplements , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Immunotherapy, Adoptive , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/drug effects , Time Factors
4.
Diabetes ; 56(8): 2169-73, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17473223

ABSTRACT

OBJECTIVE: Exposure of beta-cells to inflammatory cytokines leads to apoptotic cell death through the activation of gene networks under the control of specific transcription factors, such as interferon-gamma-induced signal transducer and activator of transcription (STAT)-1. We previously demonstrated that beta-cells lacking STAT-1 are resistant to cytokine-induced cell death in vitro. The aim of this study was to investigate the effect of STAT-1 elimination on immune-mediated beta-cell destruction in vivo. RESEARCH DESIGN AND METHODS: Multiple low-dose streptozotocin (STZ) was given to C57BL/6 mice after syngeneic STAT-1(-/-) or wild-type islet transplantation. STAT-1(-/-) and wild-type islets were also transplanted in alloxan-diabetic BALB/c and spontaneously diabetic nonobese diabetic (NOD) mice. Additionally, mice were treated with interleukin (IL)-1 blockade (IL-1 receptor antagonist [IL-1ra]) and low-dose T-cell suppression (cyclosporine A [CsA]). RESULTS: When exposed to multiple low-dose STZ in an immune-competent host, STAT-1(-/-) islets were more resistant to destruction than wild-type islets (28 vs. 100% diabetes incidence, P < or = 0.05). STAT-1 deletion also protected allogeneic islet grafts against primary nonfunction in autoimmune NOD mice (0 vs. 17% using wild-type islets). However, no difference in survival time was observed. Additionally, treating recipients with IL-1ra and CsA prolonged graft survival in chemically diabetic BALB/c mice, whereas no difference was seen between STAT-1(-/-) and C57BL/6 grafts. CONCLUSIONS: These data indicate that STAT-1 is a key player in immune-mediated early beta-cell dysfunction and death. When considering the many effector mechanisms contributing to beta-cell death following islet transplantation, multiple combined interventions will be needed for prolongation of beta-cell survival in the autoimmune context of type 1 diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Graft Rejection/immunology , Islets of Langerhans Transplantation/immunology , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/metabolism , Alloxan/pharmacology , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/immunology , Dose-Response Relationship, Drug , Gene Expression Regulation , Graft Rejection/prevention & control , Interleukin-1beta/metabolism , Mice , Mice, Knockout , STAT1 Transcription Factor/genetics , Signal Transduction/drug effects , Streptozocin/pharmacology , Survival Rate , Time Factors
5.
Diabetes ; 54(8): 2396-403, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16046307

ABSTRACT

beta-cells under immune attack are destroyed by the aberrant activation of key intracellular signaling cascades. The aim of the present study was to evaluate the contribution of the signal transducer and activator of transcription (STAT)-1 pathway for beta-cell apoptosis by studying the sensitivity of beta-cells from STAT-1 knockout (-/-) mice to immune-mediated cell death in vitro and in vivo. Whole islets from STAT-1-/- mice were completely resistant to interferon (IFN)-gamma (studied in combination with interleukin [IL]-1beta)-mediated cell death (92 +/- 4% viable cells in STAT-1-/- mice vs. 56 +/- 3% viable cells in wild-type controls, P < or = 0.001) and had preserved insulin release after exposure to IL-1beta and IFN-gamma. Moreover, analysis of cell death in cytokine-exposed purified beta-cells confirmed that protection was due to absence of STAT-1 in the beta-cells themselves. Deficiency of STAT-1 in islets completely prevented cytokine-induced upregulation of IL-15, interferon inducible protein 10, and inducible nitric oxide synthase transcription but did not interfere with monocyte chemoattractant protein 1 and macrophage inflammatory protein 3alpha expression. In vivo, STAT-1-/- mice were partially resistant to development of diabetes after multiple low-dose streptozotocin injections as reflected by mean blood glucose at 12 days after first injection (159 +/- 28 vs. 283 +/- 81 mg/dl in wild-type controls, P < or = 0.05) and diabetes incidence at the end of the follow-up period (39 vs. 73% in wild-type controls, P < or = 0.05). In conclusion, the present results indicate that STAT-1 is a crucial transcription factor in the process of IFN-gamma-mediated beta-cell death and the subsequent development of immune-mediated diabetes.


Subject(s)
Apoptosis , DNA-Binding Proteins/physiology , Interferon-gamma/physiology , Islets of Langerhans/immunology , Signal Transduction , Trans-Activators/physiology , Animals , Cell Survival , Cytokines/pharmacology , DNA-Binding Proteins/deficiency , Diabetes Mellitus, Experimental/immunology , Female , Gene Expression/drug effects , Insulin/metabolism , Insulin Secretion , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans/cytology , Male , Mice , Mice, Knockout , STAT1 Transcription Factor , Trans-Activators/deficiency
6.
Endocrinology ; 146(4): 1956-64, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15637289

ABSTRACT

1,25-Dihydroxyvitamin D(3) (1,25-(OH)(2)D(3)) is an immune modulator that prevents experimental autoimmune diseases. Receptors for 1,25-(OH)(2)D(3) are present in pancreatic beta-cells, the target of an autoimmune assault in nonobese diabetic (NOD) mice. The aim of this study was to investigate the in vivo and in vitro effects of 1,25-(OH)(2)D(3) on beta-cell gene expression and death and correlate these findings to in vivo diabetes development in NOD mice. When female NOD mice were treated with 1,25-(OH)(2)D(3) (5 microg/kg per 2 d), there was a decrease in islet cytokine and chemokine expression, which was accompanied by less insulitis. Complementing these findings, we observed that exposure to 1,25-(OH)(2)D(3) in three cell systems INS-1(E) cell line, fluorescence-activated cell sorting purified rat beta-cells, and NOD-severe combined immunodeficient islets) suppressed IP-10 and IL-15 expression in the beta-cell itself but did not prevent cytokine-induced beta-cell death. This 1,25-(OH)(2)D(3)-induced inhibition of chemokine expression in beta-cells was associated with a decreased diabetes incidence in some treatment windows targeting early insulitis. Thus, although a short and early intervention with 1,25-(OH)(2)D(3) (3-14 wk of age) reduced diabetes incidence (35 vs. 58%, P < or = 0.05), a late intervention (from 14 wk of age, when insulitis is present) failed to prevent disease. Of note, only early and long-term treatment (3-28 wk of age) prevented disease to a major extent (more than 30% decrease in diabetes incidence). We conclude that 1,25-(OH)(2)D(3) monotherapy is most effective in preventing diabetes in NOD mice when applied early. This beneficial effect of 1,25-(OH)(2)D(3) is associated with decreased chemokine and cytokine expression by the pancreatic islets.


Subject(s)
Calcitriol/pharmacology , Chemokines/genetics , Cytokines/genetics , Diabetes Mellitus, Type 1/prevention & control , Gene Expression Regulation/drug effects , Islets of Langerhans/metabolism , Animals , Calcitriol/therapeutic use , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD
SELECTION OF CITATIONS
SEARCH DETAIL
...