Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters











Publication year range
1.
Cell Mol Biol (Noisy-le-grand) ; 62(11): 108-114, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27755961

ABSTRACT

Marrubium vulgare is a European medicinal plant with numerous beneficial effects on human health. The aim of the study was to isolate the plant ethanolic extract (MVE) and to investigate its anti-melanoma and anti-glioma effects. MVE was prepared by the modified pharmacopoeial percolation method and characterized by UHPLC-LTQ OrbiTrap MS. MVE dose-dependently reduced viability of melanoma (B16) and glioma (U251) cells, but not peripheral blood mononuclear cells. It arrested cell cycle in S+G2/M phase, which was associated with the activation of MAP kinase p38 and up-regulation of antiproliferative genes p53, p21 and p27. MVE induced oxidative stress, while antioxidants abrogated its antitumor effect. Furthermore, MVE induced mitochondrial depolarization, activation of caspase-9 and -3, Parp cleavage, phosphatidylserine exposure and DNA fragmentation. The mitochondrial apoptotic pathway was associated with the up-regulation of proapoptotic genes Pten, Bak1, Apaf1, and Puma and down-regulation of antiapoptotic genes survivin and Xiap. MVE also stimulated the expression of autophagy-related genes Atg5, Atg7, Atg12, Beclin-1, Gabarab and Sqstm1, as well as LC3-I conversion to the autophagosome associated LC3-II, while autophagy inhibitors exacerbated its cytotoxicity. Finally, the most abundant phenolic components of MVE, ferulic, p-hydroxybenzoic, caffeic and chlorogenic acids, did not exert a profound effect on viability of tumor cells, suggesting that other components individually or in concert are the mediators of the extracts' cytotoxicity. By demonstrating the ability of MVE to inhibit proliferation, induce apoptosis and cytoprotective autophagy, our results suggest that MVE, alone or combined with autophagy inhibitors, could be a good candidate for anti-melanoma and anti-glioma therapy.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cell Proliferation/drug effects , Marrubium/chemistry , Plant Extracts/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Ethanol/chemistry , Humans , Marrubium/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Phenol/chemistry , Phenol/toxicity , Plant Components, Aerial/chemistry , Plant Components, Aerial/metabolism , Plant Extracts/chemistry , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects
2.
J Biomed Mater Res A ; 102(6): 1707-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23775848

ABSTRACT

The SiO2 thin films (STFs) were deposited on the surfaces of stainless steel tapes and their activity was particularly investigated from the aspect of the number density of hydroxyl groups on their surfaces. The calculation procedure of density of active OH groups includes determination of average length of silica chains that constitute silica sol particles with almost uniform size, on the base of thermogravimetric analysis. The size of SiO2 particles is analyzed by transmission electron microscopy and dynamic light scattering method. Fibroblast (L929) cell densities on the surfaces of these films were investigated using phase contrast microcopy. It was shown that there is a relationship between OH group densities and density of attached cells. Besides, the cytotoxicity effect was studied and compared for various thermally treated STFs.


Subject(s)
Coated Materials, Biocompatible/metabolism , Fibroblasts/cytology , Silicon Dioxide/metabolism , Animals , Cell Line , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/toxicity , Fibroblasts/metabolism , Mice , Silicon Dioxide/chemistry , Silicon Dioxide/toxicity , Stainless Steel/chemistry , Surface Properties
3.
Ultrason Sonochem ; 20(1): 535-45, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23006998

ABSTRACT

Mesoporous silica materials have already proved to be non-toxic and biocompatible, and also to have large pore volume and very high specific surface area suitable for loading of small molecules. Having this in mind and the fact that silicon dioxide (SiO(2)) powders can be so designed to obtain particle structures organized at multi levels, SiO(2) was chosen as a potential carrier for metronidazole, an antibiotic drug. SiO(2) powder was synthesized in two stages: first silica sol was prepared by hydrothermal synthesis and second the sol was converted into powder by dry spraying with simultaneous incorporation of the antibiotic into its structure. Scanning and transmission electron microscopy study revealed very complex structure and sub-structure of SiO(2) particles. Cell viability tests were used for estimation of cytotoxicity of so synthesized SiO(2). The drug release data showed that the system can provide drug release for a long time. Also, the device behavior is fully predictable, according to our theoretical model of multilevel structure design, and gives many opportunities for model investigations of drug release and its kinetics. The pore sizes and their distribution were observed as a limiting factor of drug release kinetics. Therefore, as the pore sizes are given as a set of discrete values, the kinetics of drug release might also be given as a set of corresponding discrete values.


Subject(s)
Drug Carriers/chemistry , Silicon Dioxide/chemistry , Ultrasonics/methods , Kinetics , Particle Size , Porosity
4.
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
5.
Nanotechnology ; 20(44): 445602, 2009 Nov 04.
Article in English | MEDLINE | ID: mdl-19801777

ABSTRACT

In this work we describe a novel method for highly efficient functionalization of single wall carbon nanotubes (SWCNTs) by DNA wrapping. Exposure of SWCNTs to gamma-irradiation (50 kGy) has lowered by one order of magnitude the amount of single stranded deoxyribonucleic acid (ssDNA) required for SWCNT modification. The resulting hybrids of gamma-irradiated SWCNTs and ssDNA were characterized by optical absorbance spectroscopy, Raman spectroscopy and Fourier transform infrared spectroscopy. Atomic force microscopy was used to investigate the morphology of hybrids. While gamma-irradiation in three different media has significantly improved the process of SWCNT dispersion, irradiation in ammonia was the most efficient. The gamma-irradiated SWCNTs functionalized with ssDNA were stabilized by electrostatic forces. This preliminary study suggests that gamma-irradiation can significantly improve the functionalization of SWCNTs with DNA.


Subject(s)
DNA, Single-Stranded/chemistry , Gamma Rays , Nanotechnology/methods , Nanotubes, Carbon/chemistry , Nanotubes, Carbon/radiation effects , Air , Ammonia/chemistry , Animals , Microscopy, Atomic Force , Models, Molecular , Osmolar Concentration , Salmon , Sodium Chloride/chemistry , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared , Spectrum Analysis, Raman , Static Electricity , Water/chemistry
6.
J Cell Mol Med ; 13(9B): 3644-54, 2009 Sep.
Article in English | MEDLINE | ID: mdl-20196784

ABSTRACT

The role of autophagy in cisplatin anticancer action was investigated using human U251 glioma, rat C6 glioma and mouse L929 fibrosarcoma cell lines. A dose- and time-dependent induction of autophagy was observed in tumour cells following cisplatin treatment, as demonstrated by up-regulation of autophagy-inducing protein beclin-1 and subsequent appearance of acridine orange-stained acidic autophagic vesicles. The presence of autophagosomes in cisplatin-treated cells was also confirmed by electron microscopy. Inhibition of autophagy with lysosomal inhibitors bafilomycin A1 and chloroquine, or a PI3 kinase inhibitor wortmannin, markedly augmented cisplatin-triggered oxidative stress and caspase activation, leading to an increase in DNA fragmentation and apoptotic cell death. The mechanisms underlying the protective effect of autophagy apparently involved the interference with cisplatin-induced modulation of Bcl-2 family proteins, as inhibition of autophagy potentiated cisplatin-mediated up-regulation of proapoptotic Bax and down-regulation of anti-apoptotic Bcl-2. Autophagy induction in cisplatin-treated cells was preceded by activation of adenosine monophosphate-activated protein kinase (AMPK) and concomitant down-regulation of mammalian target of rapamycin (mTOR)-mediated phosphorylation of p70S6 kinase. The ability of cisplatin to trigger autophagy was reduced by small interfering RNA (siRNA)-mediated AMPK silencing, while transfection with mTOR siRNA was sufficient to trigger autophagy in tumour cells. Finally, siRNA-mediated AMPK down-regulation and AMPK inhibitor compound C increased cisplatin-induced tumour cell death, while mTOR siRNA and AMPK activator metformin protected tumour cells from cisplatin. Taken together, these data suggest that cisplatin-triggered activation of AMPK and subsequent suppression of mTOR activity can induce an autophagic response that protects tumour cells from cisplatin-mediated apoptotic death.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis , Cisplatin/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Autophagy , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Silencing , Humans , Mice , Microscopy, Electron, Transmission/methods , Rats , TOR Serine-Threonine Kinases/metabolism
7.
Int J Obes (Lond) ; 33(1): 151-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18982006

ABSTRACT

OBJECTIVE: To compare the concentrations of cytokines belonging to Th17 axis (interleukin (IL)-17 and IL-23) and Th1 axis (IL-12 and interferon (IFN)-gamma) in obese and lean women, and to investigate their relationships with the proinflammatory adipokine leptin, proinflammatory cytokine macrophage migration inhibitory factor (MIF) and anthropometric and metabolic parameters of obesity. DESIGN: Cross-sectional study. SUBJECTS: Twenty-six obese women (age 20-52 years, body mass index (BMI): 30-48 kg/m(2)) and 20 healthy lean women (age 23-46 years, BMI: 18-25 kg/m(2)). MEASUREMENTS: Plasma levels of cytokines and leptin, BMI, waist circumference (WC) and insulin resistance index HOMA (homeostatic model assessment). RESULTS: Blood concentrations of IL-17, IL-23, MIF and leptin, but not IL-12 or IFN-gamma, were higher in obese compared with lean women (P=0.002, 0.046, 0.006 and 0.002, respectively). There was a positive correlation between IL-17 and IL-23 (r(s)=0.530), which was at the border of statistical significance (P=0.065). Neither IL-17 nor IL-23 correlated with leptin or MIF, and there was no association between IL-17 and IL-23 levels with BMI, WC or HOMA index. CONCLUSION: Interleukin-23/IL-17 axis is stimulated in obese women independently of the increase in abdominal fat, insulin resistance, leptin and MIF levels.


Subject(s)
Interleukin-17/blood , Interleukin-23/blood , Obesity/blood , Adult , Body Mass Index , Chi-Square Distribution , Cross-Sectional Studies , Female , Humans , Insulin Resistance , Interferon-gamma/blood , Interleukin-12/blood , Intramolecular Oxidoreductases/blood , Leptin/blood , Macrophage Migration-Inhibitory Factors/blood , Middle Aged , Obesity/immunology , Waist Circumference , Young Adult
8.
Skin Pharmacol Physiol ; 21(4): 227-34, 2008.
Article in English | MEDLINE | ID: mdl-18509257

ABSTRACT

BACKGROUND: Vitamin A and D analogues play an important role in epidermal homeostasis and are used in the treatment of various skin diseases. The failure of retinoid and vitamin D treatments is sometimes difficult to explain. METHODS: We analyzed the effect of all-trans retinoic acid (all-trans RA), 13-cis retinoic acid (13-cis RA), ergocalciferol and cholecalciferol in keratinocyte cultures established from adult donors, on the cell proliferation by means of [(3)H]thymidine incorporation and apoptosis after fluorescein diacetate/trypan blue staining. RESULTS: All tested agents exerted a dose-dependent inhibition of keratinocyte proliferation in the concentration range of 1.25-5 microM. Based on IC(50) values, the antiproliferative efficiency was as follows: cholecalciferol > ergocalciferol = all-trans RA > 13-cis RA. The observed effect of cholecalciferol and ergocalciferol, but not retinoids, involved the induction of apoptotic cell death. Combining vitamins A and D did not further increase the proliferation block and even displayed an antagonistic effect. CONCLUSION: The susceptibility of keratinocytes to the antiproliferative action of vitamins A and D was markedly different in cell cultures derived from different donors, indicating a possible predictive value of the in vitro testing for the efficiency of the clinical response to these agents.


Subject(s)
Cell Proliferation/drug effects , Keratinocytes/drug effects , Vitamin A/analogs & derivatives , Vitamin A/pharmacology , Vitamin D/analogs & derivatives , Vitamin D/pharmacology , Apoptosis/drug effects , Cells, Cultured , Cholecalciferol/pharmacology , Dose-Response Relationship, Drug , Ergocalciferols/pharmacology , Humans , Isotretinoin/pharmacology , Keratinocytes/metabolism , Tretinoin/pharmacology
9.
Cell Mol Life Sci ; 64(10): 1290-302, 2007 May.
Article in English | MEDLINE | ID: mdl-17447005

ABSTRACT

The present study reports for the first time a dual antiglioma effect of the well-known antidiabetic drug metformin. In low-density cultures of the C6 rat glioma cell line, metformin blocked the cell cycle progression in G(0)/G(1) phase without inducing significant cell death. In confluent C6 cultures, on the other hand, metformin caused massive induction of caspase-dependent apoptosis associated with c-Jun N-terminal kinase (JNK) activation, mitochondrial depolarization and oxidative stress. Metformin-triggered apoptosis was completely prevented by agents that block mitochondrial permeability transition (cyclosporin A) and oxygen radical production (N-acetylcisteine), while the inhibitors of JNK activation (SP600125) or glycolysis (sodium fluoride, iodoacetate) provided partial protection. The antiglioma effect of metformin was reduced by compound C, an inhibitor of AMP-activated protein kinase (AMPK), and was mimicked by the AMPK agonist AICAR. Similar effects were observed in the human glioma cell line U251, while rat primary astrocytes were completely resistant to the antiproliferative and proapoptotic action of metformin.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Metformin/pharmacology , AMP-Activated Protein Kinases , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspase Inhibitors , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Flow Cytometry , Glioma/drug therapy , Glioma/metabolism , Glioma/pathology , Glycolysis/drug effects , Humans , Hydrogen Peroxide/pharmacology , Hypoglycemic Agents/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Time Factors
10.
Cell Biol Toxicol ; 22(5): 371-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16897441

ABSTRACT

Chloramphenicol (CAP) is a broad-spectrum antibacterial drug that is widely used for topical application in ophthalmology and dermatology. In the present study we investigated the influence of CAP on human keratinocyte proliferation and apoptosis in vitro. CAP significantly inhibited proliferation and induced apoptosis of cultivated human keratinocytes, as revealed by incorporation of radioactive thymidine and flow cytometry analysis of intracellular esterase activity in fluorescein diacetate-stained cells, respectively. CAP-induced keratinocyte apoptosis was associated with activation of caspases and increased production of reactive oxygen species. The pro-apoptotic action of CAP was antagonized by the antioxidant agent N-acetylcysteine, the protein synthesis inhibitor cycloheximide, and PD98059, a selective inhibitor of extracellular signal-regulated kinase (ERK) activation. Taken together, these data indicate that CAP inhibits keratinocyte proliferation through induction of oxidative stress and ERK-mediated caspase-dependent apoptosis.


Subject(s)
Apoptosis/drug effects , Chloramphenicol/pharmacology , Keratinocytes/cytology , Keratinocytes/drug effects , Caspases/metabolism , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , Reactive Oxygen Species/metabolism
11.
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
12.
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
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.
Mini Rev Med Chem ; 4(7): 741-6, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15379641

ABSTRACT

The focus of this review is the influence of an immunosuppressive xenobiotic drug mycophenolic acid on the induction of nitric oxide production in various cell types. The potential therapeutic significance of the cell-specific fine-tuning of nitric oxide release by mycophenolic acid, as well as the mechanisms behind the drug action are discussed.


Subject(s)
Autoimmune Diseases/drug therapy , Enzyme Inhibitors/pharmacology , Immunosuppressive Agents/pharmacology , Mycophenolic Acid/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/biosynthesis , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , Cells, Cultured , Enzyme Inhibitors/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Mycophenolic Acid/therapeutic use , Nitric Oxide Synthase Type II
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
18.
Brain Res ; 998(1): 83-90, 2004 Feb 13.
Article in English | MEDLINE | ID: mdl-14725971

ABSTRACT

The influence of a nucleoside analog 5-aza-2'-deoxycytidine (5-AzadC) on inducible nitric oxide synthase (iNOS)-dependent nitric oxide (NO) production in various rat cell types was investigated. In C6 astrocytoma cell line and primary astrocytes, 5-AzadC enhanced proinflammatory cytokine (IFN-gamma, TNF-alpha, IL-1)-triggered NO synthesis in a time- and dose-dependent manner. In contrast, 5-AzadC did not potentiate NO production in IFN-gamma-stimulated macrophages, fibroblasts, or endothelial cells. Blockade of transcription or translation in C6 cells abolished the observed effect, suggesting the iNOS gene expression, rather than its catalytic activity, as a target for the drug action. Accordingly, 5-AzadC upregulated IFN-gamma-induced expression of iNOS mRNA in C6 astrocytes. The effect of 5-AzadC on astrocyte NO release was blocked by the inhibitor of p44/42 mitogen activated protein kinase-dependent signaling. Finally, the observed stimulatory effect of 5-AzadC on iNOS expression was apparently independent of DNA demethylation, as DNA digestion with methylation-sensitive restriction enzyme HpaII showed that 5-AzadC failed to demethylate cellular DNA in conditions used for iNOS induction.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Astrocytes/drug effects , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , DNA Modification Methylases/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Animals , Animals, Newborn , Astrocytoma/pathology , Blotting, Northern , Cells, Cultured , DNA Methylation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Decitabine , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Induction/drug effects , Gene Expression/drug effects , Interferon Regulatory Factor-1 , Interferons/pharmacology , Interleukin-1/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
19.
Curr Drug Targets Inflamm Allergy ; 2(1): 63-79, 2003 Mar.
Article in English | MEDLINE | ID: mdl-14561177

ABSTRACT

Among the numerous genes controlled by cyclic adenosine monophosphate (cAMP)/protein kinase A signalling machinery is the gene encoding the inducible nitric oxide synthase (iNOS), an enzyme catalyzing the synthesis of a highly reactive free radical nitric oxide (NO). While being a major microbicidal and tumoricidal molecule, iNOS-derived NO has also been implicated in tissue destruction, as well as in regulation of inflammatory/immune cell function in various disorders associated with excessive inflammation. A feasible way for cAMP-dependent therapeutic control of inflammation, including iNOS-mediated NO synthesis, could involve the administration of drugs that block the enzymatic activity of cAMP-degrading phosphodiesterases (PDE). Indeed, cAMP-elevating PDE inhibitors can influence iNOS activation in different cell types in vitro, and their potent anti-inflammatory effects in experimental disease models and clinical studies were frequently accompanied with profound modulation of NO production. A set of conflicting data has been generated over the years, ranging from strong suppression to marked enhancement of NO release by cAMP-increasing PDE inhibitors, depending on cell-type, iNOS stimuli, and/or the agents used. The present review summarizes the data on iNOS modulation by cAMP-elevating PDE inhibitors and possible mechanisms behind it, speculating on its contribution to the therapeutic effects of these drugs.


Subject(s)
Cyclic AMP/biosynthesis , Nitric Oxide Synthase/biosynthesis , Phosphodiesterase Inhibitors/pharmacology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , Clinical Trials as Topic , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Enzyme Activation , Humans , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type II , Shock, Septic/drug therapy , Shock, Septic/metabolism
20.
Clin Exp Immunol ; 134(1): 70-7, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12974757

ABSTRACT

To explore the role of the 10-kDa Mycobacterium tuberculosis-specific secreted antigen (MTSA-10 or CFP-10) in modulation of macrophage function, J774 macrophages were transfected stably with DNA encoding MTSA-10. Compared to normal or mock-transfected controls, MTSA-10-expressing macrophages had markedly lower levels of co-stimulatory molecule B7.1 on their surface, while the expression of B7.2 and ICAM-1 was not affected. MTSA-transfected cells also produced significantly less microbicidal free radical nitric oxide (NO) upon stimulation with interferon (IFN)-gamma, lipopolysaccharide or M. tuberculosis cell lysate. Western blot analysis revealed the absence of tyrosine-phosphorylated protein slightly larger than 112 kDa in MTSA-transfected macrophages. Moreover, the treatment of control J774 cells with protein tyrosine kinase inhibitor genistein completely mimicked the effects of transfection with MTSA-10, selectively down-regulating NO and B7.1, but not B7.2 or ICAM-1 expression. The observed MTSA-10-mediated block of B7.1 expression and NO release might contribute to the suppression of antimycobacterial response in tuberculosis.


Subject(s)
Antigens, Bacterial/metabolism , B7-1 Antigen/metabolism , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Nitric Oxide/metabolism , Animals , Blotting, Western , Cell Line , Flow Cytometry , Macrophages/metabolism , Mice , Phosphorylation , Transfection/methods , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL