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










Database
Language
Publication year range
1.
Br J Pharmacol ; 171(24): 5898-909, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25158597

ABSTRACT

BACKGROUND AND PURPOSE: Type 1 diabetes is a multifactorial inflammatory disease that develops as a result of deregulated immune responses, causing progressive autoimmune destruction of insulin-producing beta cells of pancreas. 2-((4-acetoxyphenyl)-2-chloro-N-methyl) ethylammonium chloride, compound A (CpdA), is a selective glucocorticoid receptor (GR) agonist that displays strong anti-inflammatory and immunomodulatory activities. We investigated the therapeutic effectiveness of CpdA in a pharmacological model of type 1 diabetes in mice. EXPERIMENTAL APPROACH: The utility of CpdA in diabetes prevention was evaluated in vivo through its prophylactic administration to male C57BL/6 mice that received multiple low doses of streptozotocin for immunoinflammatory diabetes induction. The effect of CpdA on disease development was studied by measuring blood glucose and insulin level, histopathological examination, determination of the nature of infiltrating cells, pro- and anti-inflammatory cytokine production, and signalling pathways. KEY RESULTS: Prophylactic in vivo therapy with CpdA conferred protection against development of immunoinflammatory diabetes in mice by dampening the M1/Th1/Th17 immune response and switching it towards an anti-inflammatory M2/Th2/Treg profile, thus preserving beta cell function. CONCLUSIONS AND IMPLICATIONS: Anti-diabetic properties of CpdA are mediated through modulation of immune cell-mediated pathways, but without triggering adverse events. These findings provide basic information for the therapeutic use of selective GR agonists in the amelioration of islet-directed autoimmunity.


Subject(s)
Acetates/pharmacology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Macrophages/drug effects , Receptors, Glucocorticoid/agonists , Th1 Cells/drug effects , Th17 Cells/drug effects , Tyramine/analogs & derivatives , Animals , Antibiotics, Antineoplastic/administration & dosage , Blood Glucose/drug effects , Blood Glucose/metabolism , Cytokines/drug effects , Cytokines/immunology , Insulin/metabolism , Insulin-Secreting Cells/immunology , Macrophages/immunology , Mice , Streptozocin/administration & dosage , Th1 Cells/immunology , Th17 Cells/immunology , Tyramine/pharmacology
2.
Scand J Immunol ; 80(2): 95-100, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24845722

ABSTRACT

Carbon monoxide (CO) is endogenously produced by haeme oxygenase-1 and has profound effects on intracellular signalling processes, generating anti-inflammatory, antiproliferative and antiapoptotic effects. A boron-containing compound CORM-A1 is capable of releasing CO in such a way to mimic physiological functions of haeme oxygenase-1. Considering the importance of Th1/Th17 versus Th2 balance in the final outcome of immune and inflammatory responses in this study we focused on immune-modulatory effects of CORM-A1 on murine lymph node-derived T cells in vitro and its influence on T-cell proliferation, activation and differentiation. Anti-CD3/CD28 antibody-triggered lymph node cells proliferation remained unaffected after 24-hour CORM-A1 treatment, as well as the expression of the early activation marker CD25. However, CORM-A1 successfully reduced the secretion of the two representative pro-inflammatory cytokines, IFN-γ and IL-17, while the secretion of anti-inflammatory cytokine IL-4 remained unchanged. Furthermore, CORM-A1 efficiently reduced the percentage of CD4(+) IFN-γ(+) and CD4(+) IL-17(+) cells, whereas CD4(+) IL-4(+) cell population increased after treatment. Also, CORM-A1 significantly reduced expression of transcription factor RORγT, necessary for Th17 development, but the expression of Th1-related and Th2-related transcription factors (T-bet and GATA-3, respectively) remained unchanged. In conclusion, our findings indicate that CO has anti-inflammatory role through the regulation of balance between pro-inflammatory Th1/Th17 and anti-inflammatory Th2 cells. Observed immunomodulatory effects of CORM-A1 could be useful for developing novel therapeutic approaches in managing Th1/Th17-mediated immune disorders.


Subject(s)
Apoptosis/immunology , Boranes/pharmacology , Carbonates/pharmacology , Cell Differentiation/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Animals , Antibodies/immunology , Apoptosis/drug effects , CD28 Antigens/immunology , CD3 Complex/immunology , Carbon Monoxide/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , GATA3 Transcription Factor/biosynthesis , Heme Oxygenase-1/metabolism , Inflammation/immunology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-4/metabolism , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , T-Box Domain Proteins/biosynthesis , Th1 Cells/drug effects , Th17 Cells/drug effects , Th2 Cells/drug effects
3.
Clin Exp Immunol ; 169(3): 244-52, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22861364

ABSTRACT

The transferrin (Tf) family of iron binding proteins includes important endogenous modulators of the immune function that may modulate autoimmune diseases. To define more clearly the role of apotransferrin (apoTf) in type 1 diabetes we determined the impact of this protein on type 1 diabetes as investigated in islet cells, animal models and patient sera. First, we demonstrated that recombinant apoTf counteracts the cytokine-induced death of murine pancreatic islet cells. Secondly, human apoTf administration favourably influences the course of type 1 diabetes in animal models, resulting in protection against disease development that was associated with reduction of insulitis and reduced levels of proinflammatory cytokines. Finally, we confirmed that patients with newly diagnosed type 1 diabetes manifest significantly lower apoTf serum levels compared to healthy controls and patients with long-lasting disease. In conclusion, our data suggest the apoTf pivotal role in the perpetuation of type 1 diabetes pathology.


Subject(s)
Apoproteins/immunology , Diabetes Mellitus, Type 1/immunology , Transferrin/immunology , Adult , Animals , Apoproteins/blood , Apoproteins/chemistry , Cell Line, Tumor/drug effects , Cytokines/metabolism , Cytokines/pharmacology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/prevention & control , Disease Progression , Female , Humans , Insulinoma/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pancreatitis/immunology , Pancreatitis/prevention & control , Rats , Rats, Inbred BB , Recombinant Proteins/pharmacology , T-Lymphocyte Subsets/immunology , Transferrin/chemistry , Young Adult
4.
Clin Exp Immunol ; 169(2): 156-63, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22774990

ABSTRACT

During pathogenesis of diabetes, pancreatic islets are exposed to high levels of cytokines and other inflammatory mediators that induce deterioration of insulin-producing beta cells. Macrophage migration inhibitory factor (MIF) plays a key role in the onset and development of several immunoinflammatory diseases and also controls apoptotic cell death. Because the occurrence of apoptosis plays a pathogenetic role in beta cell death during type 1 diabetes development and MIF is expressed in beta cells, we explored the influence of MIF deficiency on cytokine-induced apoptosis in pancreatic islets. The results indicated clearly that elevated MIF secretion preceded C57BL/6 pancreatic islets death induced by interferon (IFN)-γ + tumour necrosis factor (TNF)-α + interleukin (IL)-1ß. Consequently, MIF-deficient [MIF-knock-out (KO)] pancreatic islets or islet cells showed significant resistance to cytokine-induced death than those isolated from C57BL/6 mice. Furthermore, upon exposure to cytokines pancreatic islets from MIF-KO mice maintained normal insulin expression and produced less cyclooxygenase-2 (COX-2) than those from wild-type C57BL6 mice. The final outcome of cytokine-induced islet apoptosis in islets from wild-type mice was the activation of mitochondrial membrane pore-forming protein Bcl-2-associated X protein and effector caspase 3. In contrast, these apoptotic mediators remained at normal levels in islets from MIF-KO mice suggesting that MIF absence prevented initiation of the mitochondrial apoptotic pathway. Additionally, the protection from apoptosis was also mediated by up-regulation of prosurvival kinase extracellular-regulated kinase 1/2 in MIF-KO islets. These data indicate that MIF is involved in the propagation of pancreatic islets apoptosis probably via nuclear factor-κB and mitochondria-related proteins.


Subject(s)
Apoptosis , Cytokines/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Macrophage Migration-Inhibitory Factors/deficiency , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caspase 3/metabolism , Enzyme Activation/drug effects , Inflammation Mediators/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...