Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Cell Immunol ; 332: 129-133, 2018 10.
Article in English | MEDLINE | ID: mdl-30093071

ABSTRACT

GARP is a transmembrane protein that presents latent TGF-ß1 on the surface of regulatory T cells (Tregs). Neutralizing anti-GARP monoclonal antibodies that prevent the release of active TGF-ß1, inhibit the immunosuppressive activity of human Tregs in vivo. In this study, we investigated the contribution of GARP on mouse Tregs to immunosuppression in experimental tumors. Unexpectedly, Foxp3 conditional garp knockout (KO) mice challenged orthotopically with GL261 tumor cells or subcutaneously with MC38 colon carcinoma cells did not show prolonged survival or delayed tumor growth. Also, the suppressive function of KO Tregs was similar to that of wild type Tregs in the T cell transfer model in allogeneic, immunodeficient mice. In conclusion, garp deletion in mouse Tregs is not sufficient to impair their immunosuppressive activity in vivo.


Subject(s)
Membrane Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Forkhead Transcription Factors/immunology , Immunosuppressive Agents/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , Sequence Deletion/immunology , Transforming Growth Factor beta1/immunology
2.
Genes Immun ; 18(3): 176-183, 2017 09.
Article in English | MEDLINE | ID: mdl-28794505

ABSTRACT

Tolerogenic dendritic cells (tolDCs) are assessed as immunomodulatory adjuvants to regulate autoimmunity. The underlying gene expression endorsing their regulatory features remains ill-defined. Using deep mRNA sequencing, we compared transcriptomes of 1,25-dihydroxyvitaminD3/dexametasone-modulated tolDCs with that of non-modulated mature inflammatory DCs (mDCs). Differentially expressed genes controlled cellular interactions, metabolic pathways and endorse tolDCs with the capacity to regulate cell activation through nutrient and signal deprivation, collectively gearing tolDCs into tolerogenic immune regulators. Gene expression differences correlated with protein expression, designating low CD86 and high CD52 on the cell surface as superior discriminators between tolDCs and mDCs. Of 37 candidate genes conferring risk to developing type 1 diabetes (T1D), 11 genes differentially expressed in tolDCs and mDCs regulated immune response and antigen-presenting activity. Differential-expressed transcripts of candidate risk loci for T1D suggest a role of these 'risk genes' in immune regulation, which targeting may modulate the genetic contribution to autoimmunity.


Subject(s)
Autoimmunity/genetics , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/genetics , Immune Tolerance/genetics , Transcriptome , Antigen Presentation/genetics , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , CD52 Antigen/genetics , CD52 Antigen/metabolism , Calcitriol/pharmacology , Cell Line , Cells, Cultured , Dendritic Cells/drug effects , Dexamethasone/pharmacology , Diabetes Mellitus, Type 1/immunology , Humans
3.
J Steroid Biochem Mol Biol ; 164: 337-343, 2016 11.
Article in English | MEDLINE | ID: mdl-26232637

ABSTRACT

The differentiation of embryonic mesenchymal cells into chondrocytes and the subsequent formation of a cartilaginous scaffold that enables the formation of long bones are hallmarks of endochondral ossification. During this process, chondrocytes undergo a remarkable sequence of events involving proliferation, differentiation, hypertrophy and eventually apoptosis. Forkhead Box O (FoxO) transcription factors (TFs) are well-known regulators of such cellular processes. Although FoxO3a was previously shown to be regulated by 1,25-dihydroxyvitamin D3 in osteoblasts, a possible role for this family of TFs in chondrocytes during endochondral ossification remains largely unstudied. By crossing Collagen2-Cre mice with FoxO1lox/lox;FoxO3alox/lox;FoxO4lox/lox mice, we generated mice in which the three main FoxO isoforms were deleted in growth plate chondrocytes (chondrocyte triple knock-out; CTKO). Intriguingly, CTKO neonates showed a distinct elongation of the hypertrophic zone of the growth plate. CTKO mice had increased overall body and tail length at eight weeks of age and suffered from severe skeletal deformities at older ages. CTKO chondrocytes displayed decreased expression of genes involved in redox homeostasis. These observations illustrate the importance of FoxO signaling in chondrocytes during endochondral ossification.


Subject(s)
Bone and Bones/metabolism , Chondrocytes/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O3/genetics , Forkhead Transcription Factors/genetics , Osteogenesis/genetics , Animals , Bone and Bones/cytology , Cell Cycle Proteins , Cell Differentiation , Cell Proliferation , Chondrocytes/cytology , Collagen Type II/genetics , Collagen Type II/metabolism , Crosses, Genetic , Female , Forkhead Box Protein O1/deficiency , Forkhead Box Protein O3/deficiency , Forkhead Transcription Factors/deficiency , Gene Expression Profiling , Gene Expression Regulation, Developmental , Integrases/genetics , Integrases/metabolism , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Oxidoreductases/genetics , Oxidoreductases/metabolism , Primary Cell Culture , Signal Transduction
4.
Benef Microbes ; 6(4): 591-601, 2015.
Article in English | MEDLINE | ID: mdl-25576592

ABSTRACT

Type 1 diabetes (T1D) is a chronic autoimmune disease characterised by excessive immune reactions against auto-antigens of pancreatic ß-cells. Restoring auto-antigen tolerance remains the superior therapeutic strategy. Oral auto-antigen administration uses the tolerogenic nature of the gut-associated immune system to induce antigen-specific tolerance. However, due to gastric degradation, proper mucosal product delivery often imposes a challenge. Recombinant Lactococcus lactis have proven to be effective and safe carriers for gastrointestinal delivery of therapeutic products: L. lactis secreting diabetes-associated auto-antigens in combination with interleukin (IL)-10 have demonstrated therapeutic efficacy in a well-defined mouse model for T1D. Here, we describe the construction of recombinant L. lactis secreting the 65 kDa isoform of glutamic acid decarboxylase (GAD65) and tyrosine phosphatase-like protein ICA512 (IA-2), two major T1D-related auto-antigens. Attempts to secrete full size human GAD65 and IA-2 protein by L. lactis were unsuccessful. Trimming of GAD65 and IA-2 was investigated to optimise antigen secretion while maintaining sufficient bacterial growth. GAD65370-575 and IA-2635-979 showed to be efficiently secreted by recombinant L. lactis. Antigen secretion was verified by immunoblotting. Plasmid-derived GAD65 and IA-2 expression was combined in single strains with human IL-10 expression, a desired combination to allow tolerance induction. This study reports the generation of recombinant L. lactis secreting two major diabetes-related auto-antigens: human GAD65 and IA-2, by themselves or combined with the anti-inflammatory cytokine human IL-10. Prohibitive sequence obstacles hampering antigen secretion were resolved by trimming the full size proteins.


Subject(s)
Gene Expression , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 8/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 8/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Diabetes Mellitus, Type 1/therapy , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Plasmids , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion
5.
Diabetes Obes Metab ; 15 Suppl 3: 98-104, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24003926

ABSTRACT

Pancreatic insulin-producing ß-cells have traditionally been viewed as a quiescent cell population. However, several recent lines of evidence indicated that like most tissues the ß-cell mass is dynamically regulated with ongoing ß-cell regeneration throughout life to replenish lost or damaged ß-cells. In type 1 diabetes (T1D), this fine-tuned balance between ß-cell death and ß-cell renewal in the endocrine pancreas is lost and the deficit in ß-cell mass is largely caused by autoimmune-mediated apoptosis. Currently, the concept that a cure for T1D will require both re-establishment of immunological tolerance along with replacement or regeneration of a functional ß-cell mass in T1D patients is generally accepted. In this study our current understanding of the events directing ß-cell replication, ß-cell reprogramming from different cell types and ß-cell regeneration is reviewed, in view of the results of various immunomodulatory strategies aiming at blocking autoimmune responses against pancreatic ß-cells and at improving ß-cell mass and function in subjects with T1D.


Subject(s)
Cell Differentiation , Diabetes Mellitus, Type 1/physiopathology , Insulin-Secreting Cells/physiology , Regeneration/physiology , Animals , Cell Proliferation , Cell Transdifferentiation/physiology , Diabetes Mellitus, Type 1/pathology , Humans , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Islets of Langerhans/physiology
6.
J Steroid Biochem Mol Biol ; 136: 68-79, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23137852

ABSTRACT

INTRODUCTION: Vitamin D deficiency has been linked to type 1 and 2 diabetes, whereas supplementation may prevent both diseases. However, the extent of the effects of vitamin D or its metabolites directly on pancreatic islets is still largely unknown. The aim of the present study was to investigate how active vitamin D, 1,25(OH)2D3, affects beta cells directly by establishing its effects on global gene expression in healthy murine islets. MATERIALS AND METHODS: Pancreatic islets were isolated from 2 to 3 week old C57BL/6 mice and cultured in vitro with 1,25(OH)2D3 or vehicle for 6 and 24h. Total RNA was extracted from the islets and the effects on global gene expression were analyzed using Affymetrix microarrays. RESULTS AND DISCUSSION: Exposure to 1,25(OH)2D3 compared to vehicle resulted in 306 and 151 differentially expressed genes after 6 and 24h, respectively (n=4, >1.3-fold, p<0.02). Of these 220 were up-regulated, whereas 86 displayed a decreased expression after 6h. Furthermore, expression levels were increased for 124 and decreased for 27 genes following 24h of exposure. Formation of intercellular junctions, cytoskeletal organization, and intracellular trafficking as well as lipid metabolism and ion transport were among the most affected gene classes. Effects on several genes already identified as being part of vitamin D signaling in other cell types were observed along with genes known to affect insulin release, although with our assay we were not able to detect any effects of 1,25(OH)2D3 on glucose-stimulated insulin release from healthy pancreatic islets. CONCLUSION: The effects of 1,25(OH)2D3 on the expression of cytoskeletal and intracellular trafficking genes along with genes involved in ion transport may influence insulin exocytosis. However, an effect of 1,25(OH)2D3 on insulin release could not be detected for healthy islets in contrast to islets subjected to pathological conditions such as cytokine exposure and vitamin D deficiency as suggested by other studies. Thus, in addition to previously identified tolerogenic effects on the immune system, 1,25(OH)2D3 may affect basic functions of pancreatic beta cells, with the potential to render them more resistant to the detrimental conditions encountered during type 1 and 2 diabetes. This article is part of a Special Issue entitled 'Vitamin D Workshop'.


Subject(s)
Calcitriol/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cytoskeletal Proteins/genetics , Gene Expression Regulation/drug effects , Genes, cdc/drug effects , Insulin/metabolism , Insulin Secretion , Intercellular Junctions/drug effects , Intercellular Junctions/genetics , Islets of Langerhans/cytology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL
7.
Diabetologia ; 55(10): 2723-2732, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22752077

ABSTRACT

AIMS/HYPOTHESIS: Anti-CD3 monoclonal antibodies remain the most promising immune therapy for reversing recent-onset type 1 diabetes. However, current clinical trials have revealed their major drawback, namely the narrow therapeutic window in which low doses are ineffective and higher doses that preserve functional beta cell mass cause side effects. Strategies that sidestep these limitations while preserving or improving anti-CD3's therapeutic efficiency are essential. We hypothesised that combining a potent vitamin D(3) analogue (TX527), ciclosporin A (CsA) and anti-CD3 would act to lower the dose while maintaining or even boosting therapeutic efficacy to counteract autoimmune destruction of transplanted islets. METHODS: This study involved the use of syngeneic islet transplantation, immunofluorescence microscopy, immune phenotyping by flow cytometry, RT-PCR analysis, and in vitro and in vivo suppression assays. RESULTS: Combination therapy with TX527, CsA and anti-CD3 was well tolerated on the basis of weight, bone and calcium variables. Remarkably, combining all three agents at sub-therapeutic doses greatly reduced recurrent autoimmune responses to a grafted islet mass (mean ± SEM: 79.5 ± 18.6 days; p < 0.01), by far exceeding the therapeutic efficacy of monotherapy (24.8 ± 7.3 days for anti-CD3) and dual therapy (25.5 ± 12.4 days for anti-CD3+CsA). Combination therapy surpassed anti-CD3 monotherapy in reducing islet infiltration by effector/memory phenotype CD8(+) T cells, as well as by reducing proinflammatory cytokine responses and increasing the frequency of T regulatory cells that were functional in vitro and in vivo, and acted in a cytotoxic T lymphocyte antigen 4-dependent manner. CONCLUSIONS/INTERPRETATION: Combining the immunomodulatory actions of anti-CD3 mAb with CsA and the vitamin D(3) analogue, TX527, delivers therapeutic efficacy in an islet-transplanted NOD mouse model of diabetes.


Subject(s)
Alkynes/therapeutic use , Antibodies, Monoclonal/therapeutic use , CD3 Complex/immunology , Cholecalciferol/therapeutic use , Cyclosporine/therapeutic use , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation , Animals , Antibodies, Monoclonal/immunology , Cell Proliferation , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Female , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/pathology , Male , Mice , Mice, Inbred NOD , Secondary Prevention , T-Lymphocytes/pathology , Vitamin D/analogs & derivatives
8.
Diabetologia ; 55(4): 1167-78, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22237685

ABSTRACT

AIMS/HYPOTHESIS: Endoplasmic reticulum (ER) stress has been implicated in the development of type 2 diabetes, via effects on obesity, insulin resistance and pancreatic beta cell health. C/EBP homologous protein (CHOP) is induced by ER stress and has a central role in apoptotic execution pathways triggered by ER stress. The aim of this study was to characterise the role of CHOP in obesity and insulin resistance. METHODS: Metabolic studies were performed in Chop ( -/- ) and wild-type C57Bl/6 mice, and included euglycaemic-hyperinsulinaemic clamps and indirect calorimetry. The inflammatory state of liver and adipose tissue was determined by quantitative RT-PCR, immunohistology and macrophage cultures. Viability and absence of ER stress in islets of Langerhans was determined by electron microscopy, islet culture and quantitative RT-PCR. RESULTS: Systemic deletion of Chop induced abdominal obesity and hepatic steatosis. Despite marked obesity, Chop ( -/- ) mice had preserved normal glucose tolerance and insulin sensitivity. This discrepancy was accompanied by lower levels of pro-inflammatory cytokines and less infiltration of immune cells into fat and liver. CONCLUSIONS/INTERPRETATION: These observations suggest that insulin resistance is not induced by fat accumulation per se, but rather by the inflammation induced by ectopic fat. CHOP may play a key role in the crosstalk between excessive fat deposition and induction of inflammation-mediated insulin resistance.


Subject(s)
Fatty Liver/metabolism , Inflammation/metabolism , Insulin Resistance/physiology , Obesity/metabolism , Transcription Factor CHOP/metabolism , Adipose Tissue/metabolism , Animals , Fatty Liver/genetics , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Inflammation/genetics , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Liver/metabolism , Mice , Mice, Knockout , Obesity/genetics , Transcription Factor CHOP/genetics
9.
Oncogene ; 31(13): 1723-32, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-21841823

ABSTRACT

Destruction of insulin-producing pancreatic ß-cells by local autoimmune inflammation is a hallmark of type 1 diabetes. Histochemical analysis of pancreases from non-obese diabetic mice indicated activation of the transcription factor JunB/AP-1 (activator protein-1) after autoimmune infiltration of the islets. In vitro studies demonstrated that the cytokines tumor necrosis factor (TNF)-α and interferon (IFN)-γ induce JunB expression as a protective mechanism against apoptosis in both human and rodent ß-cells. The gene network affected was studied by microarray analysis showing that JunB regulates nearly 20% of the cytokine-modified ß-cell genes, including the transcription factor ATF3. Direct transcriptional induction of ATF3 by JunB is a key event for ß-cell survival after TNF-α+IFN-γ treatment. Moreover, pharmacological upregulation of JunB/ATF3 via increased cAMP protected rodent primary ß-cells and human islet cells against pro-inflammatory mediators. These results were confirmed in genetically modified islets derived from Ubi-JunB transgenic mice. Our findings identify ATF3 as a novel downstream target of JunB in the survival mechanism of ß-cells under inflammatory stress.


Subject(s)
Activating Transcription Factor 3/metabolism , Diabetes Mellitus, Type 1/metabolism , Inflammation/metabolism , Insulin-Secreting Cells/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Animals , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Proto-Oncogene Proteins c-jun/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
10.
Diabetologia ; 52(11): 2374-2384, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19756487

ABSTRACT

AIMS/HYPOTHESIS: IFN-gamma, together with other inflammatory cytokines such as IL-1beta and TNF-alpha, contributes to beta cell death in type 1 diabetes. We analysed the role of the transcription factor interferon regulatory factor (IRF)-1, a downstream target of IFN-gamma/signal transducer and activator of transcription (STAT)-1, in immune-mediated beta cell destruction. METHODS: Islets from mice lacking Irf-1 (Irf-1 (-/-)) and control C57BL/6 mice were transplanted in overtly diabetic NOD mice. Viability and functionality of islets were evaluated in vitro. Chemokine expression by Irf-1 (-/-) islets and INS-1E cells transfected with Irf-1 short interfering RNA (siRNA) was measured by real-time PCR as well as in functional assays in vitro. RESULTS: IRF-1 deletion in islets was associated with higher prevalence of primary non-function (63% vs 25%, p

Subject(s)
Insulin-Secreting Cells/physiology , Interferon Regulatory Factor-1/metabolism , Transcription Factors/metabolism , Animals , Cell Death , Cell Survival , DNA Primers , Glucose/pharmacology , Graft Survival/immunology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/immunology , Interferon Regulatory Factor-1/deficiency , Interferon Regulatory Factor-1/genetics , Islets of Langerhans Transplantation/methods , Mice , Mice, Inbred C57BL , Mice, Inbred NOD/surgery , Mice, Knockout , Polymerase Chain Reaction/methods , RNA, Small Interfering/genetics , Transfection , Transplantation, Homologous/immunology , Transplantation, Homologous/physiology
11.
Cell Transplant ; 16(5): 527-37, 2007.
Article in English | MEDLINE | ID: mdl-17708342

ABSTRACT

Islet transplantation is a promising treatment in type 1 diabetes, but the need for chronic immunosuppression is a major hurdle to broad applicability. Ex vivo introduction of agents by lentiviral vectors-improving beta-cell resistance against immune attack-is an attractive path to pursue. The aim of this study was to investigate whether dissociation of islets to single cells prior to viral infection and reaggregation before transplantation would improve viral transduction efficacy without cytotoxicity. This procedure improved transduction efficacy with a LV-pWPT-CMV-EGFP construct from 11.2 +/- 4.1% at MOI 50 in whole islets to 80.0 +/- 2.8% at MOI 5. Viability (as measured by Hoechst/PI) and functionality (as measured by glucose challenge) remained high. After transplantation, the transfected pseudoislet aggregates remained EGFP positive for more than 90 days and the expression of EGFP colocalized primarily with the insulin-positive beta-cells. No increased vulnerability to immune attack was observed in vitro or in vivo. These data demonstrate that dispersion of islets prior to lentiviral transfection and reaggregation prior to transplantation is a highly efficient way to introduce genes of interest into islets for transplantation purposes in vitro and in vivo, but the amount of beta-cells needed for normalization of glycemia was more than eightfold higher when using dispersed cell aggregates versus unmanipulated islets. The high price to pay to reach stable and strong transgene expression in islet cells is certainly an important cell loss.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Lentivirus/genetics , Transduction, Genetic , Animals , Cell Aggregation/drug effects , Cell Death/drug effects , Cell Line , Cell Separation , Cell Survival/drug effects , Cytokines/pharmacology , Flow Cytometry , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Rats , Rats, Wistar , Time Factors , Transgenes
12.
Curr Med Chem ; 14(17): 1893-910, 2007.
Article in English | MEDLINE | ID: mdl-17627525

ABSTRACT

1alpha,25-Dihydroxyvitamin D(3) [1,25-(OH) (2)D(3)] can exert its biological actions through binding with the nuclear vitamin D receptor (VDR), a ligand-activated transcription factor. Next to control of bone and mineral homeostasis, these actions include an immunomodulatory effect and a potent growth-inhibitory, antiproliferative or prodifferentiating action on a wide variety of cell types. The molecular mechanisms underlying this antiproliferative action form an intriguing research topic and they remain, although thoroughly studied, not completely understood. Important cell cycle regulators are involved such as cyclins, cyclin dependent kinases and their corresponding inhibitors as well as E2F transcription factors and accompanying pocket proteins. Whether 1,25-(OH)(2)D(3) influences the expression of all these proteins directly through the nuclear VDR or rather in an indirect manner is not always clear. The antiproliferative action makes 1,25-(OH) (2)D(3) a possible therapeutic tool to treat hyperproliferative disorders, among which different types of cancer. Clinical application, however, is severely hampered by calcemic effects such as hypercalcemia, hypercalciuria and increased bone resorption. Rational design of chemically modified 1,25-(OH) (2)D(3)-analogs tries to overcome this problem. As such, several thousands of analogs have been synthesized and evaluated, some of which display the desired dissociation between beneficial antiproliferative and unwanted calcemic effects. A number of those analogs are 'superagonistic' and have a several-fold stronger antiproliferative action than the parent compound. This review focuses on recent findings about the complex mechanisms behind the antiproliferative and prodifferentiating effect of 1,25-(OH) (2)D(3). Furthermore, the mode of action and possible clinical application of chemically modified 1,25-(OH) (2)D(3)-analogs will be discussed.


Subject(s)
Growth Inhibitors , Vitamin D/analogs & derivatives , Vitamin D/pharmacology , Animals , Calcitriol/pharmacology , Cell Proliferation/drug effects , Humans , Receptors, Calcitriol/drug effects , Steroids/chemical synthesis , Steroids/pharmacology , Structure-Activity Relationship
13.
Av. diabetol ; 22(3): 187-193, jul.-sept. 2006. ilus
Article in En | IBECS | ID: ibc-050112

ABSTRACT

The existence of impaired glucose tolerance in vitamin D-deficient individuals and the discovery of receptors for 1,25-dihydroxy vitamin D3 -the activated form of vitamin D- in isletbeta-cells and immune cells, the main cells involved in the pathogenesis of both types of diabetes, have aroused scientific and clinical interest in the potential role of vitamin D in the pathogenesis of the diseases, but even more so with respect to its therapeutic potential in the prevention of both forms of diabetes. Vitamin Ddeficiency is detrimental to insulin synthesis and secretion in animal models, as well as in humans, and predisposes them to type 2 diabetes. Interventions with pharmacological doses of 1,25-dihydroxyvitamin D3 or newer structural analogues can delay onset of type 1 diabetes in non-obese diabetic mice, mainly through immunomodulation, but, to date, no human data are available. Epidemiological studies suggest links between onset of type 1 diabetes and vitamin D deficiency in early life and with certain polymorphisms of the vitamin D receptor. At present, the most important conclusion from the studies on vitamin D and diabetes is that avoiding vitamin D deficiency is a priority not only for calcium and bone issues, but also for diabetes prevention


No disponible


Subject(s)
Animals , Humans , Vitamin D/metabolism , Vitamin D Deficiency/complications , Diabetes Mellitus/physiopathology , Calcium/metabolism , Immune System/physiopathology , Islets of Langerhans/metabolism
14.
Diabetologia ; 48(7): 1247-57, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15971062

ABSTRACT

Vitamin D deficiency predisposes individuals to type 1 and type 2 diabetes, and receptors for its activated form-1alpha,25-dihydroxyvitamin D3-have been identified in both beta cells and immune cells. Vitamin D deficiency has been shown to impair insulin synthesis and secretion in humans and in animal models of diabetes, suggesting a role in the development of type 2 diabetes. Furthermore, epidemiological studies suggest a link between vitamin D deficiency in early life and the later onset of type 1 diabetes. In some populations, type 1 diabetes is associated with certain polymorphisms within the vitamin D receptor gene. In studies in nonobese diabetic mice, pharmacological doses of 1alpha,25-dihydroxyvitamin D3, or its structural analogues, have been shown to delay the onset of diabetes, mainly through immune modulation. Vitamin D deficiency may, therefore, be involved in the pathogenesis of both forms of diabetes, and a better understanding of the mechanisms involved could lead to the development of preventive strategies.


Subject(s)
Calcitriol/therapeutic use , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Receptors, Calcitriol/physiology , Vitamin D Deficiency/complications , Animals , Calcitriol/metabolism , Diabetes Mellitus, Type 1/prevention & control , Humans , Islets of Langerhans/physiology , Mice , Mice, Inbred NOD , Models, Biological
15.
Transplant Proc ; 37(1): 516-7, 2005.
Article in English | MEDLINE | ID: mdl-15808695

ABSTRACT

UNLABELLED: To further study the interactions between innate and adaptive immunity in xenotransplantation, we explored the relative contribution of T-cell subsets in vascularized (heart) and cellular (islets) xenografts in a model with established xeno-non-reactivity of the innate system. MATERIALS: Specific innate xenotolerance was induced in xenoheart (hamster) recipients (nude rats) by a tolerizing regimen (TR), consisting of donor antigen infusion, temporary natural killer (NK)-cell depletion and a 4-week administration of leflunomide. Hamster pancreatic islets were transplanted either 1 week after heart transplantation or alone and syngeneic T-cell adoptive transfer was performed 10 days later. Purified CD3(+), CD4(+), and CD8(+) T cells were given 2 weeks after withdrawal of all drugs. At the day of rejection, xenografts were removed for histology. Serum was taken and IgM and IgG xenoantibody titers were measured by flow cytometry. RESULTS: Both heart and islet grafts were rejected after CD4(+) reconstitution. After CD8(+) T-cell adoptive transfer, cellular grafts were not rejected but vascularized grafts were rejected, although only after several months. Rejection in CD4(+) reconstituted nude rats was accompanied by the generation of predominantly IgG xenoantibodies. CONCLUSION: CD4(+) T lymphocytes are able to rapidly initiate the rejection of islet xenografts in the presence of a xenotolerant innate immune system either by breaking the "innate tolerance" (e.g., by activating macrophages and NK-cells) or through a mechanism without any involvement of the innate tolerance (e.g., T-dependent IgG antibody production). In contrast, CD8(+) T cells provoke a late rejection of only xenoheart grafts.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Heart Transplantation/immunology , Islets of Langerhans Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Cricetinae , Killer Cells, Natural/immunology , Lymphocyte Depletion , Rats
16.
J Autoimmun ; 23(1): 9-15, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15236748

ABSTRACT

During the development of type 1 diabetes, pancreatic beta-cells are subject to an immune attack, leading to their apoptotic or necrotic cell death. Apoptotic beta-cells are also present during periods of tissue remodeling, such as in early life. Macrophages should clear apoptotic cells silently without production of pro-inflammatory cytokines. The aim of the present study was to investigate the cytokine pattern of NOD macrophages exposed to apoptotic or necrotic cells in vitro. In contrast to the limited response of macrophages from C57BL/6 or NOR mice, NOD macrophages reacted aberrantly to both necrotic and apoptotic cells, with secretion of inappropriately high amounts of IL1beta and TNFalpha. Further exploration of the macrophage behavior showed an excessive response of NOD macrophages when exposed to LPS (high iNOS and IL12p40 levels), accompanied by hyper-activation of NF-kappaB(p65). In contrast, NOD macrophages failed to up-regulate IL1beta and IL12p40 in response to IFNgamma. This failure correlated with low protein levels and a low phosphorylation state of STAT1alpha. We conclude that NOD macrophages have severely aberrant cytokine expression patterns that could contribute to the initiation or continuation of an immune attack towards the pancreatic beta-cells and thus onset and progression of type 1 diabetes.


Subject(s)
Apoptosis/physiology , Cytokines/metabolism , Macrophages/metabolism , Necrosis/metabolism , Animals , Cytokines/genetics , Female , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred NOD , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription Factors/physiology
17.
Diabetologia ; 47(3): 451-462, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14758446

ABSTRACT

AIMS/HYPOTHESIS: 1,25-dihydroxyvitamin D(3), the active form of vitamin D, prevents Type 1 diabetes in non-obese diabetic (NOD) mice. Epidemiological data show a threefold increase in human Type 1 diabetes when vitamin D deficiency was present in the first months of life. To evaluate whether a similar dietary deficiency affects diabetes incidence in NOD mice, we generated NOD mice with vitamin D deficiency in early life. METHODS: Breeding pairs of NOD mice, as well as their offspring (test mice), were kept in surroundings devoid of ultraviolet light and were fed a vitamin D-depleted diet for 100 days. Mice were followed for 250 days. RESULTS: At 250 days, 35% (12/35) male and 66% (22/33) female vitamin D-deficient mice were diabetic compared to 15% (6/40, p=0.05) and 45% (13/29, p<0.01) of the control mice. At 100 days no difference in insulitis was seen, but more vitamin D-deficient mice were glucose intolerant. Higher IL1 expression was detected in islets of vitamin D-deficient mice and their peritoneal macrophages had an aberrant cytokine profile (low IL1 and IL6, high IL15). Thymus and lymph nodes of vitamin D-deficient mice contained less CD4(+)CD62L(+) cells. CONCLUSION/INTERPRETATION: Vitamin D status increases the expression of Type 1 diabetes in NOD mice. Our data in NOD mice, as well as human epidemiological data, point to the importance of preventing vitamin D deficiency in early childhood. Controlling this dietary factor could be an easy and safe way to reduce the incidence of Type 1 diabetes in subjects who are genetically at risk.


Subject(s)
Diabetes Mellitus, Type 1/physiopathology , Vitamin D Deficiency/complications , Animals , Body Weight , Calcitriol/blood , Calcium/blood , Calcium/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Female , Male , Mice , Mice, Inbred NOD , Reverse Transcriptase Polymerase Chain Reaction , Vitamin D Deficiency/immunology , Vitamin D Deficiency/pathology
18.
Diabetologia ; 46(8): 1115-23, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12879250

ABSTRACT

AIMS/HYPOTHESIS: High levels of inflammation locally in the graft during the initial days after transplantation can cause primary non-function (PNF) of grafted xenogeneic islets in NOD mice. The aim of this study was to explore in a model of spontaneous diabetes, the NOD mouse, the potential of anti-inflammatory agents in the prevention of PNF after xenogeneic islet transplantation. METHODS: Spontaneously diabetic NOD mice were transplanted with 300 rat islets. Animals were treated with acetylsalicylic acid (AsA), rofecoxib, TGF-beta or IL-1 receptor antagonist (IL-1ra). Intra-graft expression of inflammation-related molecules was measured by real time PCR 8 h post-transplantation. At the same time point, plasma nitrite levels were measured. RESULTS: Xenogeneic islets transplanted in control spontaneously diabetic mice resulted in PNF in 16 out of 38 mice (42%). Initial graft loss was not altered by administration of rofecoxib (30%) or TGF-beta (25%). AsA reduced the rate of rapid graft loss to 8% ( p<0.05 vs controls) and administration of IL-1ra even totally prevented PNF (0%, p<0.05 vs controls). Furthermore, all therapies prolonged the mean survival time of xenogeneic islet grafts. The inhibition of PNF by AsA was associated with decreased intra-islet levels of inflammation-related molecules (IL-1, TNF-alpha, iNOS, COX-2) and chemokines (MCP-1 and MIP-3alpha). Finally, also a diminished production of systemic nitrite levels was observed in AsA- and IL-1ra-treated islet recipients. CONCLUSIONS/INTERPRETATION: These data show that treatment with AsA or IL-1ra prevents PNF after islet transplantation in spontaneously diabetic NOD mice. Moreover, the involvement of non-specific inflammation is recognized in xenogeneic islet PNF in spontaneously diabetic NOD mice.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Diabetes Mellitus, Type 1/surgery , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Aspirin/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Diabetes Mellitus, Type 1/immunology , Graft Survival/drug effects , Lactones/therapeutic use , Mice , Mice, Inbred NOD , Sulfones
19.
Diabetologia ; 46(2): 255-66, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12627325

ABSTRACT

AIMS/HYPOTHESIS: Cytokines and chemokines are important mediators of immune responses due to their ability to recruit and activate leukocytes. Using microarray analysis we observed that rat beta cells exposed to IL-1beta and IFN-gamma have increased mRNA levels of chemokines and IL-15. The aim of this study was to characterize the expression of IP-10, MIP-3alpha, fractalkine and IL-15 in rat beta cells, human pancreatic islets, and in islets isolated from NOD mice, both during the pre-diabetic period and following islet transplantation. METHODS: FACS-purified rat beta cells and human islets were cultured with IL-1beta, IFN-gamma and/or TNF-alpha. Islets were isolated from NOD or BALB/c mice at different ages. For syngeneic islet transplantation, 2- or 3-week-old NOD islets were grafted under the kidney capsule of spontaneously diabetic NOD recipients. Chemokine and IL-15 mRNA expression and protein release were evaluated, respectively, by RT-PCR and ELISA. RESULTS: Human islets and rat beta cells express IP-10, MIP-3alpha, fractalkine and IL-15 mRNAs upon exposure to cytokines. The expression of IL-15, IP-10 and fractalkine is regulated by IFN-gamma, while the expression of MIP-3alpha is IL-1beta-dependent. Moreover, cytokines induced IL-15, IP-10, Mig, I-TAC and MIP-3alpha protein accumulation in culture medium from human islets. In vivo, there was an age-related increase in IL-15, IP-10 and MIP-3alpha expression in islets isolated from NOD mice. Following syngeneic islet transplantation, increased expression of IL-1beta, IFN-gamma, fractalkine, IP-10, MCP-1 and MIP-3alpha mRNAs were observed in the grafts. CONCLUSION/INTERPRETATION: Cytokine-exposed islets or beta cells express chemokines and IL-15. This could contribute to the recruitment and activation of mononuclear cells and development of insulitis in early Type 1 diabetes and during graft destruction.


Subject(s)
Chemokines/metabolism , Interferon-gamma/pharmacology , Interleukin-15/metabolism , Interleukin-1/pharmacology , Islets of Langerhans/metabolism , Mice, Inbred NOD/metabolism , Adult , Animals , Chemokine CCL20 , Chemokine CX3CL1 , Chemokine CXCL10 , Chemokines, CC/genetics , Chemokines, CX3C/genetics , Chemokines, CXC/genetics , Humans , Interleukin-15/genetics , Macrophage Inflammatory Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Rats , Rats, Wistar
20.
Clin Exp Immunol ; 128(2): 213-20, 2002 May.
Article in English | MEDLINE | ID: mdl-11985511

ABSTRACT

Autoimmune diabetes recurrence is in part responsible for islet graft destruction in type 1 diabetic individuals. The aim of the present study was to design treatment modalities able to prevent autoimmune diabetes recurrence after islet transplantation in spontaneously diabetic NOD mice. In order to avoid confusion between autoimmune diabetes recurrence and allograft rejection, we performed syngeneic islet transplantations in spontaneously diabetic NOD mice. Mice were treated with mouse interferon-beta (IFN-beta, 1 x 105 IU/day), a new 14-epi-1,25-(OH)2D3-analogue (TX 527, 5 microg/kg/day) and cyclosporin A (CsA, 7.5 mg/kg/day) as single substances and in combinations. Treatment was stopped either 20 days (IFN-beta and CsA) or 30 days (TX 527) after transplantation. Autoimmune diabetes recurred in 100% of control mice (MST 11 days). None of the mono-therapies significantly prolonged islet graft survival. Combining CsA with TX 527 maintained graft function in 67% of recipients as long as treatment was given (MST 31 days, P < 0.01 versus controls). Interestingly, 100% of the IFN-beta plus TX 527-treated mice had normal blood glucose levels during treatment, and even had a more pronounced prolongation of graft survival (MST 62 days, P < 0.005 versus controls). Cytokine mRNA analysis of the grafts 6 days after transplantation revealed a significant decrease in IL-2, IFN-gamma and IL-12 messages in both IFN-beta plus TX 527- and CsA plus TX 527-treated mice, while only in the IFN-beta with TX 527 group were higher levels of IL-10 transcripts observed. Therefore, we conclude that a combination of IFN-beta and TX 527 delays autoimmune diabetes recurrence in islet grafts in spontaneously diabetic NOD mice.


Subject(s)
Alkynes , Cholecalciferol/therapeutic use , Diabetes Mellitus, Type 1/therapy , Immunologic Factors/therapeutic use , Interferon-beta/therapeutic use , Islets of Langerhans Transplantation , Animals , Cholecalciferol/administration & dosage , Combined Modality Therapy , Cyclosporine/administration & dosage , Cyclosporine/therapeutic use , Diabetes Mellitus, Type 1/physiopathology , Drug Synergism , Drug Therapy, Combination , Immunologic Factors/administration & dosage , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Interferon-beta/administration & dosage , Mice , Mice, Inbred NOD , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL
...