Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cell Mol Immunol ; 18(6): 1353-1374, 2021 06.
Article in English | MEDLINE | ID: mdl-33958746

ABSTRACT

Multiple sclerosis (MS) is a leading cause of chronic neurological disability in young to middle-aged adults, affecting ~2.5 million people worldwide. Currently, most therapeutics for MS are systemic immunosuppressive or immunomodulatory drugs, but these drugs are unable to halt or reverse the disease and have the potential to cause serious adverse events. Hence, there is an urgent need for the development of next-generation treatments that, alone or in combination, stop the undesired autoimmune response and contribute to the restoration of homeostasis. This review analyzes current MS treatments as well as different cell-based therapies that have been proposed to restore homeostasis in MS patients (tolerogenic dendritic cells, regulatory T cells, mesenchymal stem cells, and vaccination with T cells). Data collected from preclinical studies performed in the experimental autoimmune encephalomyelitis (EAE) model of MS in animals, in vitro cultures of cells from MS patients and the initial results of phase I/II clinical trials are analyzed to better understand which parameters are relevant for obtaining an efficient cell-based therapy for MS.


Subject(s)
Cell- and Tissue-Based Therapy , Multiple Sclerosis/therapy , Animals , Clinical Trials as Topic , Combined Modality Therapy , Humans , Immune Tolerance , Models, Biological , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology
2.
Eur Rev Med Pharmacol Sci ; 22(8): 2461-2467, 2018 04.
Article in English | MEDLINE | ID: mdl-29762853

ABSTRACT

OBJECTIVE: We investigated the effect of metformin and caffeine on fibrosarcoma in hamsters. MATERIALS AND METHODS: 32 Syrian golden hamsters of both sexes, weighing approximately 100 g, were randomly allocated to 3 experimental and 2 control groups, with a minimum of 6 animals per group. 2 x 106 BHK-21/C13 cells in 1 ml were injected subcutaneously into the animals' back in 4 groups. The first experimental group started peroral treatment with metformin 500 mg/kg daily, the second with caffeine 100 mg/kg daily and the third with a combination of metformin 500 mg/kg and caffeine 100 mg/kg daily, via a gastric probe 3 days before tumor inoculation. After 2 weeks, when the tumors were approximately 2 cm in the control group, all animals were sacrificed. The blood was collected for glucose and other analyses. The tumors were excised and weighed and their diameters were measured. The tumor samples were pathohistologically (HE) and immunohistochemically (Ki-67, CD 31, COX IV, GLUT-1, iNOS) assessed and the main organs toxicologically analyzed, including the control animals that had received metformin and caffeine. Tumor volume was determined using the formula LxS2/2, where L was the longest and S the shortest diameter. Ki-67-positive cells in the tumor samples were quantified. Images were taken and processed by software UTHSCSA Image Tools for Windows Version 3.00. Statistical significances were determined by the Student's t-test. RESULTS: The combination of metformin and caffeine inhibited fibrosarcoma growth in hamsters without toxicity. CONCLUSIONS: Administration of metformin with caffeine might be an effective and safe approach in novel nontoxic adjuvant anticancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Caffeine/administration & dosage , Fibrosarcoma/drug therapy , Fibrosarcoma/pathology , Metformin/administration & dosage , Animals , Cricetinae , Drug Synergism , Female , Male , Mesocricetus , Random Allocation , Treatment Outcome
3.
Eur Rev Med Pharmacol Sci ; 21(23): 5499-5505, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29243803

ABSTRACT

OBJECTIVE: We investigated the effect of metformin on an in vivo solid tumor model of fibrosarcoma in hamsters. MATERIALS AND METHODS: 33 Syrian golden hamsters of both sexes, weighing approximately 100 g, were randomly allocated to 3 experimental and 2 control groups. 2 x 106 BHK-21/C13 cells in 1 ml were injected subcutaneously into the animals' back in 4 groups. The first experimental group (7 animals) started peroral treatment with metformin 500 mg/kg daily via a gastric probe 7 days before tumor inoculation, the second (8 animals) 3 days before inoculation and the third (6 animals) immediately after inoculation. After 2 weeks, when the tumors were approximately 2-3 cm in the control group with tumors (6 hamsters), all animals were sacrificed. The blood was collected for glucose and other analyses. The tumors were excised and weighed and their diameters were measured. The tumor samples were histologically assessed and the main organs toxicologically analyzed, including 6 control animals that had received metformin without tumor inoculation. Tumor volume was determined using the formula Lx S2/2, where L was the longest and S the shortest diameter. Ki-67-positive cells in the tumor samples were quantified; images were taken and processed by software UTHSCSA Image Tools for Windows Version 3.00. Statistical significances of differences in tumor weight, volume, number of Ki-67-positive cells and other parameters were determined by the Student´s t-test. RESULTS: Metformin inhibited fibrosarcoma growth in hamsters without toxicity. The seven-day pretreatment was important for the statistically significant effect. CONCLUSIONS: Administration of metformin as an anti-tumor drug might be an effective and safe therapeutic approach in novel non-toxic therapies for human sarcomas.


Subject(s)
Fibrosarcoma/drug therapy , Metformin/therapeutic use , Animals , Blood Glucose/analysis , Cell Line , Cricetinae , Female , Fibrosarcoma/pathology , Ki-67 Antigen/metabolism , Male , Transplantation, Homologous
4.
Benef Microbes ; 7(3): 363-73, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26839070

ABSTRACT

Gut microbiota and gut-associated lymphoid tissue have been increasingly appreciated as important players in pathogenesis of various autoimmune diseases, including multiple sclerosis. Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis that can be induced with an injection of spinal cord homogenate emulsified in complete Freund's adjuvant in Dark Agouti (DA) rats, but not in Albino Oxford (AO) rats. In this study, mesenteric lymph nodes (MLN), Peyer's patches (PP) and gut microbiota were analysed in these two rat strains. There was higher proportion of CD4(+) T cells and regulatory T cells in non-immunised DA rats in comparison to AO rats. Also, DA rat MLN and PP cells were higher producers of pro-inflammatory cytokines interferon-γ and interleukin-17. Finally, microbial analyses showed that uncultivated species of Turicibacter and Atopostipes genus were exclusively present in AO rats, in faeces and intestinal tissue, respectively. Thus, it is clear that in comparison of an EAE-susceptible with an EAE-resistant strain of rats, various discrepancies at the level of gut associated lymphoid tissue, as well as at the level of gut microbiota can be observed. Future studies should determine if the differences have functional significance for EAE pathogenesis.


Subject(s)
Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/microbiology , Gastrointestinal Microbiome , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Peyer's Patches/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Disease Models, Animal , Feces/microbiology , Firmicutes/classification , Firmicutes/isolation & purification , Intestinal Mucosa/microbiology , Lymph Nodes/immunology , Rats , T-Lymphocytes, Regulatory/immunology
5.
Neuroscience ; 292: 1-12, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25701126

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a model of multiple sclerosis (MS), inflammatory, demyelinating and neurodegenerative disease of the central nervous system (CNS). Clinically manifested EAE can be induced in Dark Agouti (DA) rats, but not in Albino Oxford (AO) rats by immunization with spinal cord homogenate (SCH) and complete Freund's adjuvant (CFA). Matrix metalloproteinases (MMP) play important roles in various steps of MS and EAE pathogenesis. Expression of gelatinases MMP2 and MMP9, their activator MMP14 and their inhibitor tissue inhibitor of MMP (TIMP)1 in the CNS of AO and DA rats immunized with SCH+CFA was determined. Expression of mRNA for MMP2, MMP9 and MMP14 was higher and expression of TIMP1 mRNA was lower in AO rats. However, gelatinase activity in spinal cords was higher in samples obtained from DA rats. Further, while there was no strain difference in MMP2 and MMP9 mRNA expression in lymph nodes of the immunized rats, gelatinase activity was higher in DA rats. This activity was reduced by antiinflammatory cytokines interleukin (IL)-10 and IL-4. Interestingly, gelatinase activity was detected in the nuclei of cells within the CNS, but not of those in lymph nodes. Our results imply that posttranscriptional regulation of MMP2 and MMP9 expression and/or function determines low gelatinase activity within the CNS and in immune cells of EAE-resistant AO rats.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Cell Nucleus/enzymology , Genetic Predisposition to Disease , Interleukin-10/metabolism , Interleukin-4/metabolism , Lymph Nodes/enzymology , RNA, Messenger/metabolism , Rats , Spinal Cord/enzymology
6.
Allergy ; 69(9): 1154-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24924975

ABSTRACT

BACKGROUND: Group 2 innate lymphoid cells (ILC2s) were shown to be involved in the initiation and coordination of Th2-type immune responses in allergic disease animal models. Recently, ILC2s enrichment was noted in chronic rhinosinusitis (CRS) patients; however, the role of ILC2s in coordinating the Th2 response in CRS remains to be elucidated. Here, we characterize the ILC2 compartment in CRS by investigating the correlations between ILC2s, Th2 cells and Th2 cytokines expression in CRS patients. METHODS: We used flow cytometric analysis of sinonasal mucosal tissues of 29 CRS patients and 5 controls to quantify ILC2s and Th2 cells. Messenger RNA expression levels of IL-5, IL-13, IL-25, IL-33, TSLP and GATA3 were determined using qRT-PCR. RESULTS: ILC2s were significantly enriched in nasal polyps (CRSwNP) patients. Multivariate linear regression showed a significant positive association of ILC2 numbers with CRSwNP and allergic CRS and a negative association with the number of previous endoscopic sinus surgeries. Group 2 innate lymphoid cell numbers significantly correlated with Th2 cell frequencies. Messenger RNA expression levels of IL-5 and IL-13 were increased in CRSwNP compared with controls, while mRNA levels of IL-25 and GATA3 were significantly reduced. CONCLUSIONS: Our results characterize the complex interactions between ILC2s and other Th2 response elements in the context of CRS and suggest that ILC2 enrichment occurs in CRSwNP and in allergic CRS patients.


Subject(s)
Hypersensitivity/immunology , Lymphocytes/immunology , Nasal Polyps/immunology , Rhinitis/immunology , Sinusitis/immunology , Adult , Aged , Chronic Disease , Female , Flow Cytometry , Humans , Hypersensitivity/complications , Immunity, Innate , Immunophenotyping , Male , Middle Aged , Nasal Polyps/complications , Rhinitis/complications , Sinusitis/complications , Th2 Cells
7.
Rhinology ; 51(4): 315-22, 2013 12.
Article in English | MEDLINE | ID: mdl-24260763

ABSTRACT

BACKGROUND: The role of inflammasomes in chronic inflammation has been the subject of intense research in recent years. Chronic rhinosinusitis (CRS), a persistent inflammatory disease, continues to be investigated hoping that a clearer pathophysiologic description will guide discovery of future treatment modalities. This study investigates the role of inflammasome complexes in CRS patients with Staphylococcus aureus biofilm infection, a key culprit associated with disease severity and recalcitrance. METHODOLOGY: Sinonasal tissue samples were collected from CRS patients with (P+) and without (P-) polyps and controls. S. aureus biofilm status was obtained using fluorescence in situ hybridization and classified as biofilm positive (B+) or negative (B-). RNA was analysed using a Human Inflammasome PCR array, profiling the expression of 84 genes involved in inflammasome function. RESULTS: Sixteen samples were obtained: 5 B+P+, 5 B-P- and 6 controls. Comparing B+P+ vs. controls showed the greatest number of differentially expressed genes. In particular, Absent in Melanoma 2 (AIM2) was consistently and significantly up-regulated in the B+P+ vs. B-P- and controls. In contrast, when comparing the B-P- vs. controls, no genes showed significant changes. CONCLUSION: Our results indicate the involvement of inflammasome complexes and their signalling pathways in CRS patients with polyps and S. aureus biofilms. In particular, AIM2, activated by intracellular double-stranded DNA, is up-regulated in this group, implying that S. aureus may play a role in intracellular triggering of the inflammasome response. Studies with further patient stratification and assessing corresponding protein expression are needed to further characterize the role of inflammasomes in CRS.


Subject(s)
Biofilms , Inflammasomes/metabolism , Rhinitis/etiology , Sinusitis/etiology , Staphylococcal Infections/etiology , Staphylococcus aureus/physiology , Adult , Aged , Case-Control Studies , Chronic Disease , Female , Humans , Inflammasomes/genetics , Male , Middle Aged , Nasal Polyps/etiology , Nasal Polyps/metabolism , Nasal Polyps/pathology , RNA, Messenger/metabolism , Rhinitis/metabolism , Rhinitis/pathology , Sinusitis/metabolism , Sinusitis/pathology , Staphylococcal Infections/metabolism , Staphylococcal Infections/pathology
8.
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
9.
Genetika ; 48(4): 556-60, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22730776

ABSTRACT

Previous studies revealed significant phenotypic plasticity, genetic variability and population differentiation of flower morphometric traits on dwarf bearded iris Iris pumila. Also, study of I. pumila flowering phenology revealed significant impact of habitat type as well as population differentiation for flowering time. Since the flowering time can influence other flower traits, we performed this analysis of flower morphometric traits in three time points during the flower bud ontogenic development in two habitat types (open vs. shaded). Analysis revealed that for most of the traits greater trait values were recorded for open habitat but only on latter time points. For most of the analyzed traits direction of differences in bud stage was the opposite to the direction of differences in mature flower stage detected in previous studies. However, length of the stem, a trait that showed the greatest variability between habitats and populations and therefore greatest genetic differentiation and phenotypic plasticity, was significantly greater in the samples from the late flowering shaded habitat in all time samples, indicating that in case of this trait different mechanisms were involved. Those findings have implications for design of the future studies on I. pumila.


Subject(s)
Flowers/genetics , Iris Plant/genetics , Quantitative Trait, Heritable , Ecosystem , Flowers/growth & development , Genetic Variation , Iris Plant/growth & development , Phenotype , Population
10.
J Nutr Metab ; 20102010.
Article in English | MEDLINE | ID: mdl-20814437

ABSTRACT

This study aimed to determine the effect of varying dietary intake of the major n-3 PUFA in human diets, alpha-linolenic acid (ALA; 18 : 3n-3), on expression of lipogenic genes in adipose tissue. Rats were fed diets containing from 0.095%en to 6.3%en ALA and a constant n-6 PUFA level for 3 weeks. Samples from distinct adipose depots (omental and retroperitoneal) were collected and mRNA expression of the pro-lipogenic transcription factors Sterol-Retinoid-Element-Binding-Protein1c (SREBP1c) and Peroxisome Proliferator Activated Receptor-gamma (PPARgamma), lipogenic enzymes Sterol-coenzyme Desaturase1 (SCD-1), Fatty Acid Synthase (FAS), lipoprotein lipase (LPL) and glycerol-3-phosphate dehydrogenase (G3PDH) and adipokines leptin and adiponectin determined by qRT-PCR. Increasing dietary ALA content resulted in altered expression of SREBP1c, FAS and G3PDH mRNA in both adipose depots. SREBP1c mRNA expression was related directly to n-6 PUFA concentrations (omental, r(2) = .71; P < .001; Retroperitoneal, r(2) = .20; P < .002), and inversely to n-3 PUFA concentrations (omental, r(2) = .59; P < .001; Retroperitoneal, r(2) = .19; P < .005) independent of diet. The relationship between total n-6 PUFA and SREBP1c mRNA expression persisted when the effects of n-3 PUFA were controlled for. Altering red blood cell concentrations of n-3 PUFA is thus associated with altered expression of lipogenic genes in a depot-specific manner and this effect is modulated by prevailing n-6 PUFA concentrations.

11.
Heart ; 95(10): 799-806, 2009 May.
Article in English | MEDLINE | ID: mdl-19074922

ABSTRACT

OBJECTIVE: To identify predictors of early TIMI 3 flow patency of the infarct-related artery after prehospital thrombolysis in patients with ST-segment elevation myocardial infarction (STEMI) using data from a "real-world" population, and to develop a nomogram for triaging patients to emergency angiography. DESIGN: Multicentre, observational, prospective, cohort study. SETTING: 79 Hospitals in France with a prehospital mobile intensive care unit and a coronary care unit with 24 h access to coronary angiography. PATIENTS: 997 Patients with STEMI. INTERVENTIONS: All patients received prehospital thrombolysis within 6 h of symptom onset and angiography was performed within 6 h of thrombolysis. MAIN OUTCOME MEASURES: Coronary patency (TIMI flow). RESULTS: The median age of the population was 59 years and the sample comprised 18% women. After multivariable logistic regression analysis, predictors of TIMI 3 flow in the infarct-related artery were current/previous smoking (odds ratio (OR) = 1.60, 95% confidence interval 1.15 to 2.22), < or =5 leads with ST-segment elevation before thrombolysis (OR = 1.59, 1.12 to 2.25), Killip class I (OR = 1.96, 1.05 to 3.67), chest pain relief (OR = 1.62, 1.17 to 2.25) and ST-segment resolution > or =70% (OR = 1.76, 1.29 to 2.38). A nomogram was developed to assess the probability of TIMI 3 flow, according to smoking status, number of leads with ST elevation before thrombolysis, Killip class, chest pain relief and ST-segment resolution. CONCLUSIONS: This study provides quantitative data for predicting success of prehospital thrombolysis. The nomogram is a simple tool for predicting likelihood of coronary patency, based on clinical and electrocardiographic data. It may help to identify patients who require emergency angiography and rescue percutaneous coronary intervention.


Subject(s)
Coronary Angiography/methods , Emergency Medical Services , Myocardial Infarction/drug therapy , Thrombolytic Therapy/methods , Vascular Patency/physiology , Aged , Epidemiologic Methods , Female , Humans , Male , Middle Aged , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Practice Guidelines as Topic , Treatment Outcome
12.
Arch Mal Coeur Vaiss ; 99(9): 823-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17067102

ABSTRACT

The effectiveness of thrombolytics has been clearly demonstrated in more than half the cases in the large cohorts of patients selected for trials during the acute phase of myocardial infarction. At individual level, thrombolysis will clinically either succeed or fail so, for the medical team managing the patient, choice of treatment may be likened to a gamble which in the best of cases (most often) leads to an uncomplicated success and, in the worst of cases, failure worsened by a severe complication. OPTIMAL is a multidisciplinary and multicentre, prospective cohort study associating mobile medical teams and interventional cardiology units to test the hypothesis that the outcome of prehospital thrombolysis does not depend on chance alone but also varies according to demographic, etiological, clinical and logistic factors involved in the occurrence and management of myocardial infarction. The primary objective of this French study, conducted over one year on more than 800 subjects, is to identify the predictors of the results of prehospital thrombolysis from a very early angiographic evaluation. The results for this cohort may be useful for setting up appropriate management strategies for acute myocardial infarction, from the prehospital phase (thrombolysis or not) up to in-hospital orientation of the patients (angiography room or Intensive Care Unit) and to determine the most judicious time for coronary angiography. OPTIMAL is to date the largest prospective serie of prehospital thrombolysis evaluated by an early angiographic control.


Subject(s)
Emergency Medical Services/organization & administration , Myocardial Infarction/drug therapy , Research Design , Thrombolytic Therapy , Coronary Angiography , Data Collection/methods , Electrocardiography , France , Humans , Myocardial Infarction/diagnostic imaging , Patient Selection , Prospective Studies , Registries
13.
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
14.
Cell Mol Life Sci ; 62(11): 1275-82, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15905960

ABSTRACT

The present study describes the ability of an anthraquinone derivative aloe emodin (AE) to reduce the cytotoxic activity of the platinum(II)-based anticancer agent cisplatin toward murine L929 fibrosarcoma and C6 glioma cell lines. The protective effect of AE was demonstrated by MTT and crystal violet assays for cell viability, and involved supression of cisplatin-induced apoptosis and necrosis, as assessed by lactate dehydrogenase release and flow cytometric analysis of DNA fragmentation or phosphatidylserine exposure. Cell-based ELISA and Western blot analysis revealed that AE abolished cisplatin-triggered activation of extracellular signal-regulated kinase (ERK) in tumor cells, while activation of c-Jun N-terminal kinase was not significantly altered. A selective blockade of ERK activation with PD98059 mimicked the protective effect of AE treatment in both tumor cell lines. Moreover, AE failed to protect tumor cells against the ERK-independent toxicity of the Pt(IV)-based complex tetrachloro(O,O-dibutyl-ethylenediamine-N,N'-di-3-propanoate)platinum(IV). Taken together, these data indicate that herbal anthraquinone AE can downregulate the anticancer activity of cisplatin by blocking the activation of ERK in tumor cells.


Subject(s)
Antineoplastic Agents/antagonists & inhibitors , Cisplatin/antagonists & inhibitors , Emodin/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibrosarcoma/drug therapy , Glioma/drug therapy , Animals , Anthraquinones , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Drug Antagonism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/physiology , Fibrosarcoma/enzymology , Fibrosarcoma/pathology , Flavonoids/pharmacology , Glioma/enzymology , Glioma/pathology , Mice , Platinum/chemistry , Rats , Structure-Activity Relationship
15.
Cell Mol Life Sci ; 61(14): 1805-15, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15241556

ABSTRACT

Aloe-emodin (AE) is a plant-derived hydroxyanthraquinone with potential anticancer activity. We investigated the ability of AE to modulate survival of mouse L929 fibrosarcoma and rat C6 astrocytoma cells through interference with the activation of inducible nitric oxide (NO) synthase (NOS) and subsequent production of tumoricidal free radical NO. Somewhat surprisingly, AE in a dose-dependent manner rescued interferon-gamma + interleukin-1-stimulated L929 cells from NO-dependent killing by reducing their autotoxic NO release. The observed protective effect was less pronounced in C6 cells, due to their higher sensitivity to a direct toxic action of the drug. AE-mediated inhibition of tumor cell NO release coincided with a reduction in cytokine-induced accumulation of transcription and translation products of genes encoding inducible NOS and its transcription factor IRF-1, while activation of NF-kappaB remained unaltered. These data indicate that the influence of AE on tumor growth might be more complex that previously recognized, the net effect being determined by the balance between the two opposing actions of the drug: its capacity to directly kill tumor cells, but also to protect them from NO-mediated toxicity.


Subject(s)
Apoptosis/drug effects , Cytokines/drug effects , Emodin/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/metabolism , Animals , Anthraquinones , Astrocytes/drug effects , DNA-Binding Proteins/drug effects , Down-Regulation , Fibroblasts/drug effects , Interferon Regulatory Factor-1 , Mice , NF-kappa B/drug effects , Nitric Oxide Synthase Type II , Phosphoproteins/drug effects , Rats , Tumor Cells, Cultured
16.
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
17.
Cell Mol Life Sci ; 61(10): 1167-75, 2004 May.
Article in English | MEDLINE | ID: mdl-15141302

ABSTRACT

Taxol is a microtubule-stabilizing agent that has recently been shown effective in the treatment of experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. As astrocytes could modulate central nervous system (CNS) autoimmunity through inducible nitric oxide synthase (iNOS)-mediated production of immunoregulatory free radical nitric oxide (NO), we investigated the effect of taxol on NO synthesis in rat astrocytes. Taxol, either alone or in combination with interferon-gamma, induced NO generation in primary astrocytes and astrocytoma C6 cells in a dose- and time-dependent manner. Accordingly, the drug markedly up-regulated the expression of both iNOS mRNA and protein in astrocytes. The observed effect of taxol was mediated through induction of iNOS transcription factors NF-kappaB and IRF-1, and required the activation of p38 MAP kinase and JNK. Finally, NO release by taxol-stimulated astrocytes was blocked with the microtubule-depolymerizing agent colchicine, suggesting the involvement of a microtubule-stabilizing activity of taxol in the observed effect.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Astrocytes/enzymology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Nitric Oxide Synthase/metabolism , Paclitaxel/pharmacology , Animals , Brain/metabolism , Cell Line, Tumor , Cells, Cultured , Colchicine/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Interferon-gamma/pharmacology , Models, Biological , Nitric Oxide Synthase Type II , Nitrites/metabolism , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic , Up-Regulation , p38 Mitogen-Activated Protein Kinases
19.
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
20.
J Neuroimmunol ; 119(2): 183-91, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585620

ABSTRACT

The effect of interleukin-17 (IL-17) on production of nitric oxide (NO) in rodent astrocytes was investigated. While IL-17 by itself did not induce NO production, it caused a dose-dependent enhancement of IFN-gamma-triggered NO synthesis in both mouse and rat primary astrocytes. In contrast, IL-17 was unable to stimulate NO synthesis in either murine or rat macrophages. IFN-gamma-triggered expression of mRNA for iNOS, but not for its transcription factor interferon regulatory factor-1 (IRF-1), was markedly elevated in IL-17-treated astrocytes. The induction of iNOS mRNA by IL-17 in IFN-gamma-pretreated astrocytes was abolished by antagonists of nuclear factor-kappaB (NF-kappaB) activation--a proteasome inhibitor MG132 and an antioxidant agent PDTC, as well as with specific p38 MAP kinase inhibitor SB203580. While IL-17 stimulated both IL-1beta and IL-6 production in astrocytes, only IL-1 was partly responsible for IL-17-induced NO release. Finally, IL-17 synergized with exogenous IL-1beta and TNF-alpha for astrocyte NO production. Having in mind a well-known neurotoxic action of NO, these results suggest a possible role for IL-17 in the inflammatory diseases of the CNS.


Subject(s)
Astrocytes/enzymology , Astrocytes/immunology , Interleukin-17/pharmacology , Nitric Oxide Synthase/metabolism , Animals , Astrocytes/cytology , Astrocytoma , Drug Synergism , Enzyme Activation/drug effects , Enzyme Activation/immunology , Gene Expression Regulation, Enzymologic/immunology , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Interleukin-1/immunology , Interleukin-1/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred CBA , NF-kappa B/immunology , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase Type II , RNA, Messenger/analysis , Rats , Rats, Inbred Strains , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...