Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Placenta ; 36(2): 150-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25530499

ABSTRACT

INTRODUCTION: Macrophage migration inhibitory factor (MIF) is expressed by villous and extravillous cytotrophoblast. This study was aimed to investigate functional relevance of MIF for human trophoblast. METHODS: MIF mRNA and protein were documented in cytotrophoblast (CT) and extravillous trophoblast cell line HTR-8/SVneo by RT-PCR, Western blot (WB), and immunocytochemistry. Recombinant human MIF (rhMIF), or its specific inhibitor (S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1) were used in Wound healing migration and Matrigel invasion tests. Potential effectors, integrin subunits and matrix metalloproteinases (MMP) were studied using WB and gelatin zymography, respectively. RESULTS: Blocking endogenous MIF by ISO-1 decreased HTR-8/SVneo cell migration dose dependently, most significantly with 200 µg/ml to 65% of control. Supplementation with rhMIF induced a significant stimulation to 129% of control with 200 ng/ml. In CT cell invasion test, ISO-1 at 200 µg/ml reduced invasion to 59% of control, while rhMIF (200 ng/ml) induced stimulation to 159% of control. In HTR-8/SVneo cells, invasion was significantly inhibited by ISO-1 to 40%, and increased to 150% of control by rhMIF (200 ng/ml). Integrin α1 was reduced by ISO-1 in both cell types, while integrins α5 and ß1 were not changed. Addition of rhMIF increased integrin α1. In the presence of ISO-1, levels of MMP-2 and MMP-9 were reduced in CT and HTR-8/SVneo, while rhMIF stimulated MMP-2 in CT and MMP-9 in HTR-8/SVneo cells. CONCLUSION: Reported findings provide the first insight into the cellular effects of MIF in human trophoblast, which acts to promote cell migration and invasion.


Subject(s)
Cell Movement/drug effects , Embryo Implantation/drug effects , Intramolecular Oxidoreductases/antagonists & inhibitors , Isoxazoles/pharmacology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Trophoblasts/drug effects , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Embryo Implantation/genetics , Female , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Pregnancy , Pregnancy Trimester, First , Recombinant Proteins/pharmacology , Trophoblasts/physiology
2.
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
3.
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
4.
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
5.
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
6.
Food Chem Toxicol ; 50(9): 3181-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22683487

ABSTRACT

Intrinsic characteristics of melanoma cells such as expression of inducible nitric oxide synthase (iNOS), redox status, and activity of signaling pathways involved in proliferation, differentiation and cell death define the response of the cells to the diverse treatments. In this context we compared the effectiveness of herbal antaquinone aloe emodin (AE) against mouse B16 melanoma and human A375, different in initial activity of ERK1/2, constitutive iNOS expression and basal level of reactive oxygen species (ROS). Both cell lines are sensitive to AE treatment. However, while the agent induces differentiation of B16 cells toward melanocytes, in A375 cells promoted massive apoptosis. Differentiation of B16 cells, characterized by enhanced melanin production and tyrosinase activity, was mediated by H(2)O(2) production synchronized with rapid p53 accumulation and enhanced expression of cyclins D1 and D3. Caspase mediated apoptosis triggered in A375 cells was accompanied with Bcl-2 but not iNOS down-regulation. In addition, opposite regulation of Akt-ERK1/2 axis in AE treated B16 and A375 cells correlated with different outcome of the treatment. However, AE in a dose-dependent manner rescued both B16 and A375 cells from doxorubicin- or paclitaxel-induced killing. These data indicate that caution is warranted when AE is administrated to the patients with conventional chemotherapy.


Subject(s)
Anthraquinones/pharmacology , Melanoma, Experimental/pathology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Humans , MAP Kinase Signaling System , Melanoma, Experimental/enzymology , Melanoma, Experimental/metabolism , Mice , Nitric Oxide Synthase Type II/metabolism , Reactive Oxygen Species/metabolism
7.
Immunol Res ; 52(1-2): 120-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22388638

ABSTRACT

Model of systemic Aspergillus fumigatus infection induced by intravenous application of conidia is suitable for studying important aspects of invasive aspergillosis including relationship between infection and mortality, dissemination of infection and immune mechanisms involved in host resistance to this fungus. Use of this model allows the investigation of both innate and adaptive immune response characteristics in resistant/susceptible host, and investigating the contribution of genetic background and cytokine gene deficiency improves the knowledge of the diversity of mechanisms of immune response to Aspergillus infection. Studying of various aspects of systemic aspergillosis contributes to development of antifungal drugs.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Spleen/immunology , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Aspergillosis/mortality , Humans , Macrophage Migration-Inhibitory Factors/immunology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA
8.
Parasite Immunol ; 32(6): 450-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20500676

ABSTRACT

Trichinella spiralis is a helminth that provokes Th2 and anti-inflammatory type responses in an infected host. Our previous studies using Dark Agouti (DA) rats indicated that T. spiralis infection reduced experimental autoimmune encephalomyelitis (EAE) severity in rats. The aim of this study was to analyse the mechanisms underlying EAE suppression driven by T. spiralis infection. Reduced clinical and histological manifestations of the disease were accompanied by increased IL-4 and IL-10 production and decreased IFN-gamma and IL-17 production in draining lymph node cells. This indicates that T. spiralis infection successfully maintains a Th2 cytokine bias regardless of EAE induction. High IL-10 signifies parasite-induced anti-inflammatory and/or regulatory cell responses. Transfer of splenic T cell-enriched population of cells from T. spiralis-infected rats into EAE immunized rats caused amelioration of EAE and in some cases protection from disease development. This population of cells contained higher proportion of CD4(+) CD25(+) Foxp3(+) regulatory cells and produced high level of IL-10 when compared with uninfected rats.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/immunology , Trichinella spiralis/immunology , Trichinellosis/complications , Trichinellosis/immunology , Adoptive Transfer , Animals , CD4 Antigens/analysis , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Forkhead Transcription Factors/analysis , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukin-2 Receptor alpha Subunit/analysis , Interleukin-4/metabolism , Lymph Nodes/immunology , Rats , Severity of Illness Index , T-Lymphocyte Subsets/chemistry , T-Lymphocyte Subsets/immunology , Trichinellosis/parasitology
9.
Clin Exp Rheumatol ; 26(1 Suppl 48): S8-11, 2008.
Article in English | MEDLINE | ID: mdl-18570748

ABSTRACT

Type 1 diabetes mellitus (T1D) and inflammatory bowel diseases (IBD) are multifactorial disorders of autoimmune origin. Several microbial agents have been reported to be associated with the development of type 1 diabetes and inflammatory bowel diseases in animal models by different mechanisms. These models which resemble the phenotype of the human disease they mimic, can be very useful to identify important pathogenetic mechanisms, as well as therapeutical targets to treat the disease. This review is focused on the immune inflammatory pathways which are considered to be associated with the pathogenesis T1D and IBD in transgenic mice.


Subject(s)
Diabetes Mellitus, Type 1 , Infections/complications , Infections/immunology , Inflammatory Bowel Diseases , Animals , Dendritic Cells/immunology , Dendritic Cells/microbiology , Dendritic Cells/virology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/virology , Disease Models, Animal , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/virology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/microbiology , T-Lymphocytes, Regulatory/virology
10.
Food Chem Toxicol ; 46(5): 1825-33, 2008 May.
Article in English | MEDLINE | ID: mdl-18313195

ABSTRACT

Numerous studies have shown immunostimulatory and anti-tumor effects of water and standardized aqueous ethanol extracts derived from the medicinal mushroom, Coriolus versicolor, but the biological activity of methanol extracts has not been examined so far. In the present study we investigated the anti-tumor effect of C. versicolor methanol extract (which contains terpenoids and polyphenols) on B16 mouse melanoma cells both in vitro and in vivo. In vitro treatment of the cells with the methanol extract (25-1600 microg/ml) reduced melanoma cell viability in a dose-dependent manner. Furthermore, in the presence of the methanol extract (200 microg/ml, concentration IC(50)) the proliferation of B16 cells was arrested in the G(0)/G(1) phase of the cell cycle, followed by both apoptotic and secondary necrotic cell death. In vivo methanol extract treatment (i.p. 50 mg/kg, for 14 days) inhibited tumor growth in C57BL/6 mice inoculated with syngeneic B16 tumor cells. Moreover, peritoneal macrophages collected 21 days after tumor implantation from methanol extract-treated animals exerted stronger tumoristatic activity ex vivo than macrophages from control melanoma-bearing mice. Taken together, our results demonstrate that C. versicolor methanol extract exerts pronounced anti-melanoma activity, both directly through antiproliferative and cytotoxic effects on tumor cells and indirectly through promotion of macrophage anti-tumor activity.


Subject(s)
Agaricales/chemistry , Melanoma, Experimental/drug therapy , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Flow Cytometry , L-Lactate Dehydrogenase/metabolism , Macrophages/pathology , Melanoma, Experimental/pathology , Methanol , Mice , Mice, Inbred C57BL , Necrosis , Phenols/pharmacology , Solvents , Terpenes/chemistry , Tetrazolium Salts , Thiazoles , Trypan Blue
11.
Exp Parasitol ; 118(4): 641-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18226814

ABSTRACT

Helminth infection has a potent systemic immunomodulatory effect on the host immune response, which also affects the development of autoimmune diseases. We investigated the dose-dependent influence of Trichinella spiralis infection on experimental autoimmune encephalomyelitis (EAE). Our model of concomitant T. spiralis infection and EAE demonstrates that established infection of Dark Agouti (DA) rats with the parasite causes amelioration of the clinical course of induced EAE in a dose-dependent way. Infection with T. spiralis L1 stage muscle larvae (TSL1) reduced the severity of the autoimmune disease as judged by lower maximal clinical score, cumulative index, duration of illness and degree of mononuclear cell infiltration in T. spiralis infected animals compared to control, EAE-induced group. This study provides a valuable model of worm infection to investigate helminth-induced regulatory mechanisms for optimal benefit to the host.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Dose-Response Relationship, Immunologic , Encephalomyelitis, Autoimmune, Experimental/complications , Female , Rats , Rats, Wistar , Severity of Illness Index , Spinal Cord/pathology , Trichinellosis/complications
12.
Cell Mol Life Sci ; 62(22): 2658-68, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16261264

ABSTRACT

The influence of the proinflammatory cytokine interleukin (IL)-17 on inducible nitric oxide (NO) synthase (iNOS)-mediated NO release was investigated in the mouse insulinoma cell line MIN6 and mouse pancreatic islets. IL-17 markedly augmented iNOS mRNA/protein expression and subsequent NO production induced in MIN6 cells or pancreatic islets by different combinations of interferon-gamma, tumor necrosis factor-alpha, and IL-1beta. The induction of iNOS by IL-17 was preceded by phosphorylation of p38 mitogen-activated protein kinase (MAPK), and inhibition of p38 MAPK activation completely abolished IL-17-stimulated NO release. IL-17 enhanced the NO-dependent toxicity of proinflammatory cytokines toward MIN6 cells, while IL-17-specific neutralizing antibody partially reduced the NO production and rescued insulinoma cells and pancreatic islets from NO-dependent damage induced by activated T cells. Finally, a significant increase in blood IL-17 levels was observed in a multiple low-dose streptozotocin model of diabetes, suggesting that T cell-derived IL-17 might be involved in NO-dependent damage of beta cells in this disease.


Subject(s)
Insulin-Secreting Cells/enzymology , Interleukin-17/physiology , Nitric Oxide Synthase Type II/toxicity , Animals , Cell Line, Tumor , Cell Survival/physiology , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/physiology , Recombinant Proteins/pharmacology , T-Lymphocytes/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Cell Mol Life Sci ; 62(5): 589-98, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15747063

ABSTRACT

The effect of aloe emodin (AE), a herbal anthraquinone derivative, on the rat C6 glioma cell line was investigated. In addition to cell cycle block and caspasedependent apoptosis, AE led to the formation of intracytoplasmic acidic vesicles indicative for autophagic cell death. Moreover, differentiation of surviving cells toward the astrocytic lineage was confirmed by typical morphological changes and increased expression of glial fibrillary acidic protein (GFAP). AE did not affect the activation of mitogen-activated protein kinase p38, Jun-N-terminal kinase, or transcription factor NF-kappaB, but markedly inhibited the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in C6 cells. A selective inhibitor of ERK activation, PD98059, mimicked the effects of AE on glioma cell morphology and GFAP expression, but failed to induce either apoptosis or autophagy. Taken together, these results indicate that the anti-glioma action of AE involves ERK-independent induction of both apoptosis and autophagy, as well as ERK inhibition-mediated differentiation of glioma cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Differentiation/drug effects , Emodin/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Glioma/enzymology , Animals , Anthraquinones , Apoptosis , Autolysis , Cell Differentiation/physiology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/physiology , Rats
14.
Clin Exp Immunol ; 137(1): 109-16, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15196250

ABSTRACT

Although the inhibitory effect of iron on macrophage production of tumoricidal free radical nitric oxide (NO) has been reported, its possible influence on macrophage anti-tumour activity has not been established. In the present study, FeSO4 markedly reduced IFN-gamma + LPS-induced NO synthesis in mouse and rat macrophages. The effect of iron coincided with the loss of macrophage cytotoxic activity against NO-sensitive C6 rat astrocytoma and L929 mouse fibrosarcoma cell lines, as measured by MTT assay for cellular respiration and the crystal violet test for cell viability. Tumour cell survival did not improve further in the presence of FeSO4 if macrophage NO release and cytotoxicity were already blocked by aminoguanidine. In accordance with the results obtained with exogenous iron, cell membrane permeable iron chelator o-phenanthroline enhanced both macrophage NO release and anti-tumour activity. Iron also down-regulated NO production and increased the viability of L929 fibrosarcoma cells stimulated with IFN-gamma + LPS in the absence of macrophages. However, neither NO release nor cell viability was affected by iron addition to cultures of the C6 astrocytoma cell line. Iron was unable to prevent L929 and C6 cell death induced by the NO releasing chemicals SNP and SIN-1, indicating that iron-mediated inhibition of NO synthesis, rather than interference with its cytotoxic action, was responsible for the protection of tumour cells. Collectively, these results indicate that iron might protect tumour cells by reducing both macrophage and tumour cell-derived NO release.


Subject(s)
Iron/immunology , Macrophages/immunology , Neoplasms/immunology , Nitric Oxide/metabolism , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytotoxicity, Immunologic/immunology , Down-Regulation , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Interferon-gamma , Iron/metabolism , Iron Chelating Agents/pharmacology , Lipopolysaccharides , Macrophage Activation/immunology , Mice , Mice, Inbred C57BL , Neoplasms/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Phenanthrolines/pharmacology , Rats , Rats, Inbred Strains
15.
Clin Exp Immunol ; 136(1): 49-55, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15030513

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a well-recognized model for multiple sclerosis (MS) in humans. However, adjuvants used with encephalitogens to induce EAE produce non-specific effects interfering with the mechanisms involved in the autoimmune response to the central nervous system (CNS) tissue. It is therefore important to establish a more suitable model of EAE for analysis of autoimmune phenomena resembling those operative in MS. Here we report that EAE can be induced regularly in Dark Agouti (DA) strain of rats with spinal cord tissue without any adjuvant, as judged by both clinical and histological parameters. The incidence and severity of EAE depended on the origin of the encephalitogen, the rat versus guinea pig spinal cord homogenate being more efficient. Furthermore, EAE could be reinduced in animals which had recovered from disease that had been induced actively with encephalitogen alone, suggesting the role of adjuvant-generated non-specific mechanisms in resistance to reinduction of EAE. Thus, EAE induced in DA rats with encephalitogen alone provides a reproducible model for defining pathogenically relevant events in CNS autoimmunity devoid of the potentially misleading effects of adjuvants.


Subject(s)
Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/etiology , Animals , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Freund's Adjuvant , Lymphocyte Activation/immunology , Male , Multiple Sclerosis/immunology , Rats , Rats, Inbred Strains , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/immunology , Tissue Extracts/immunology
16.
Cell Mol Life Sci ; 60(3): 518-25, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12737311

ABSTRACT

The effect of interleukin (IL)-17 on the activation of inducible nitric oxide (NO) synthase (iNOS) and subsequent production of NO was investigated. IL-17 induced NO production in both mouse and rat endothelial cells in a dose- and time-dependent manner. This was paralleled by the induction of mRNA for iNOS, which was markedly down-regulated by specific antagonists of protein tyrosine kinase, p38 MAP kinase or iNOS transcription factor NF-kappaB. The expression of iNOS transcription factor IRF-1 was also induced by IL-17 and blocked by all three inhibitors, suggesting that the induction of iNOS by IL-17 might be partly exerted through IRF-1 activation. Neutralization with the specific antibody showed that endogenous IL-17 is involved in T cell-mediated NO production in endothelial cells and NO-dependent suppression of T cell growth. These data indicate that IL-17-triggered iNOS activation in endothelial cells might participate in regulation of the T cell-dependent inflammatory response.


Subject(s)
Interleukin-17/metabolism , Myocardium/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/biosynthesis , Animals , DNA-Binding Proteins/metabolism , Endothelium, Vascular/metabolism , Fibroblasts/metabolism , Interferon Regulatory Factor-1 , Mice , Phosphoproteins/metabolism , Rats , T-Lymphocytes/metabolism
17.
Clin Exp Immunol ; 132(2): 239-46, 2003 May.
Article in English | MEDLINE | ID: mdl-12699411

ABSTRACT

Mycophenolate mofetil (MMF) is an immunosuppressive drug that acts as a selective inhibitor of inosine monophosphate dehydrogenase (IMPDH). MMF has recently been shown to inhibit the enzymatic activity of inducible NO synthase (iNOS) and subsequent production of the cytotoxic free radical nitric oxide (NO) in endothelial cells. We here investigated the effect of bioactive MMF compound mycophenolic acid (MPA) on iNOS-mediated NO synthesis in fibroblasts, which are important source of NO in rheumatoid arthritis and during rejection of solid organ transplants. MPA exerted dose-dependent inhibition of NO synthesis, measured as nitrite accumulation, in IFN-gamma + LPS-stimulated L929 mouse fibroblast cell line and rat primary fibroblasts. The effect of MPA was not mediated through interference with IMPDH-dependent synthesis of iNOS co-factor BH4 and subsequent suppression of iNOS enzymatic activity, as direct BH4 precursor sepiapterin failed to block the action of the drug. MPA suppressed the IFN-gamma + LPS-induced expression of fibroblast iNOS protein, as well as mRNA for iNOS and its transcription factor IRF-1, as assessed by cell-based ELISA and semiquantitative RT-PCR, respectively. MPA suppression of fibroblast NO release, iNOS, and IRF-1 activation, was efficiently prevented by exogenous guanosine, indicating that the drug acted through reduction of IMPDH-dependent synthesis of guanosine nucleotides. These results suggest that MPA inhibits NO production in fibroblasts by blocking guanosine nucleotide-dependent expression of iNOS gene, through mechanisms that might involve the interference with the induction of iNOS transcription factor IRF-1.


Subject(s)
Fibroblasts/enzymology , Immunosuppressive Agents/pharmacology , Mycophenolic Acid/pharmacology , Nitric Oxide Synthase/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Fibroblasts/drug effects , Interferon Regulatory Factor-1 , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Mice , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
19.
Cytokine ; 19(4): 181-6, 2002 Aug 21.
Article in English | MEDLINE | ID: mdl-12297111

ABSTRACT

Mycophenolic acid (MPA) and A77 1726, the active components of the immunosuppressants mycophenolate mophetil and leflunomide, respectively, in a dose-dependent manner inhibited interferon (IFN)-gamma/LPS-induced interleukin (IL)-6 release in confluent cultures of mouse L929 fibrosarcoma cells. In addition, both drugs markedly reduced the production of the free radical gas nitric oxide (NO), without affecting the viability of L929 cells. The inhibitors of NO synthase, aminoguanidine and L-NMMA, but not L-NMMA inactive counterpart D-NMMA, mimicked the effects of A77 1726 and MPA on IL-6 generation in L929 fibroblasts. Furthermore, NO-releasing substance SNP completely reverted IL-6 accumulation in L929 cultures treated with A77 1726, while only partial recovery of IL-6 production was observed in the presence of MPA. MPA, but not A77 1726, significantly suppressed NO-independent IL-6 release triggered by cAMP-elevating agent rolipram. Thus, while A77 1726 effect on IL-6 production was mediated through concomitant reduction of NO synthesis, MPA action was mainly independent of the interference with NO generation. Finally, both agents inhibited IFN-gamma/LPS-triggered IL-6 production in mouse primary fibroblasts, but not in mouse peritoneal macrophages, indicating cell-specificity of this novel anti-inflammatory action of A77 1726 and MPA.


Subject(s)
Aniline Compounds/pharmacology , Fibroblasts/metabolism , Hydroxybutyrates/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-6/biosynthesis , Isoxazoles/pharmacology , Mycophenolic Acid/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Animals , Cell Line , Cell Survival , Cells, Cultured , Coloring Agents/pharmacology , Crotonates , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Guanidines/metabolism , Humans , Interferon-gamma/pharmacology , Interleukin-6/metabolism , Leflunomide , Macrophages/metabolism , Mice , Nitric Oxide Synthase/metabolism , Nitriles , Rolipram/pharmacology , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Toluidines , omega-N-Methylarginine/pharmacology
20.
Clin Exp Immunol ; 129(2): 214-23, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12165076

ABSTRACT

The new immunosuppressive agent mycophenolate mofetil (MMF) has been shown recently to exert a protective effects in certain animal models of autoimmunity, including diabetes in diabetes-prone bio-breeding (BB) rats. In the present study, the immunomodulatory potential of MMF was investigated in autoimmune diabetes induced by multiple low doses of streptozotocin (MLD-STZ) in genetically susceptible DA rats 20 mg STZ/kg body weight (b.w.) for 5 days] and CBA/H mice (40 mg STZ/kg b.w. for 5 days). In both species, short time treatment of animals with MMF (25 mg/kg) during the early development of the disease, as well as continuous MMF treatment, prevented the appearance of hyperglycaemia and inflammatory infiltrates in the pancreatic tissue. Moreover, clinical manifestations of diabetes were suppressed by application of the drug after the onset of clinical symptoms. Treatment with guanosine (1 mg/kg) in parallel with MMF completely reversed MMF activity in vivo, indicating that inhibition of inosine monophosphate dehydrogenase (IMPDH) was responsible for the observed suppressive effects. MMF-mediated protection from diabetes correlated with reduced ex vivo spontaneous spleen mononuclear cell (MNC) proliferation and defective adhesive cell interactions. MMF-treated animals also had lower local production of IFN-gamma, as well as IL-12 and nitric oxide (NO) production by peripheral tissues (spleen and peritoneal cells), compared to that in control diabetic groups, while IL-10 level was elevated. Together, these data demonstrate that MMF interferes with autoimmune process in streptozotocin-induced diabetes at multiple levels, including lymphocyte proliferation and adhesion, as well as pro/anti-inflammatory cytokine balance.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Immunosuppressive Agents/pharmacology , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacology , Animals , Autoimmunity/drug effects , Cell Adhesion/drug effects , Cytokines/metabolism , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Enzyme Inhibitors/pharmacology , Guanosine/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , In Vitro Techniques , Inflammation Mediators/metabolism , Interleukin-12/biosynthesis , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Lymphocytes/immunology , Male , Mice , Mice, Inbred CBA , Nitric Oxide/biosynthesis , Rats , Streptozocin/administration & dosage , Streptozocin/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...