Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biomaterials ; 30(36): 6940-6, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19781768

ABSTRACT

In the present study, we compared the effects of nanocrystalline fullerene suspension (nanoC(60)) on tumour cell growth in vitro and in vivo. NanoC(60) suspension was prepared by solvent exchange using tetrahydrofuran to dissolve C(60). In vitro, nanoC(60) caused oxidative stress, mitochondrial depolarization and caspase activation, leading to apoptotic and necrotic death in mouse B16 melanoma cells. Biodistribution studies demonstrated that intraperitoneally injected radiolabeled (125I) nanoC(60) readily accumulated in the tumour tissue of mice subcutaneously inoculated with B16 cells. However, intraperitoneal administration of nanoC(60) over the course of two weeks starting from melanoma cell implantation not only failed to reduce, but significantly augmented tumour growth. The tumour-promoting effect of nanoC(60) was accompanied by a significant increase in splenocyte production of the immunoregulatory free radical nitric oxide (NO), as well as by a reduction in splenocyte proliferative responses to T- and B-cell mitogens ConcanavalinA and bacterial lipopolysaccharide, respectively. A negative correlation between NO production and splenocyte proliferation indicated a possible role of NO in reducing the proliferation of splenocytes from nanoC(60)-injected mice. These data demonstrate that nanoC(60), in contrast to its potent anticancer activity in vitro, can potentiate tumour growth in vivo, possibly by causing NO-dependent suppression of anticancer immune response.


Subject(s)
Antineoplastic Agents , Cell Line, Tumor , Fullerenes , Immunosuppression Therapy , Nanoparticles/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Caspases/metabolism , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Enzyme Activation , Fullerenes/chemistry , Fullerenes/pharmacology , Materials Testing , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Spleen/cytology , Spleen/metabolism
2.
Eur J Pharmacol ; 591(1-3): 106-13, 2008 Sep 04.
Article in English | MEDLINE | ID: mdl-18606162

ABSTRACT

We investigated the influence of adenosine on inducible nitric oxide (NO) synthase (iNOS)-dependent NO synthesis and viability of cytokine-treated C6 rat glioma cells. Adenosine significantly inhibited interferon-gamma (IFN-gamma)+interleukin-1beta (IL-1beta)-induced synthesis of iNOS mRNA/protein and subsequent production of NO in C6 cells. The uptake of adenosine into glioma cells was not required for the suppression of iNOS induction, as confirmed by the inability of the adenosine transport blocker nitrobenzylthyoinosine to block the observed effect. Adenosine also blocked the IFN-gamma+IL-1beta-triggered expression of mRNA for the proinflammatory cytokine TNF-alpha, while it significantly enhanced the accumulation of cyclooxygenase-2 (COX-2) mRNA in glioma cells. However, blockade of TNF-alpha action and COX-2 activity with anti-TNF-alpha antibodies and indomethacin, respectively, revealed that modulation of TNF-alpha and COX-2 was not involved in adenosine-mediated iNOS suppression. Adenosine significantly inhibited cytokine-induced activation of mitogen-activated protein kinase (MAPK) family members p38 MAPK, p42/44 MAPK and c-Jun N-terminal kinase (JNK) in C6 cells. The levels of transcription factors IRF-1 and c-Fos, as well as the phosphorylation of c-Jun were also reduced in adenosine-treated C6 cells, while the activation of NF-kappaB was enhanced via increased phosphorylation of its inhibitory unit IkappaB. Importantly, adenosine-mediated suppression of NO release rescued glioma cells from NO-dependent cytokine cytotoxicity. These data suggest a possible role for adenosine-mediated inhibition of glial NO synthesis in regulation of the inflammatory CNS damage and brain cancer progression.


Subject(s)
Adenosine/pharmacology , Glioma/metabolism , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide/biosynthesis , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Gene Expression Regulation/drug effects , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
3.
Nitric Oxide ; 19(2): 177-83, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18460348

ABSTRACT

In this study we evaluated the effects of the new NO donating compound (S,R)-3-phenyl-4,5-dihydro-5-isoxazole acetic acid-nitric oxide (GIT-27NO) on the A375 human melanoma cell line. Treatment with the drug led to concentration-dependent reduction of mitochondrial respiration and number of viable cells in cultures. Decreased cell viability correlated with release and internalization of NO and was neutralized by the extracellular scavenger hemoglobin. GIT-27NO neither influenced cell division nor induced accidental or autophagic cell death. Early signs of apoptosis were observed upon coculture with the drug, and resulting in marked accumulation of hypodiploid cells, suggesting that the induction of apoptosis is one primary mode of action of the compound in A375 cells. GIT-27NO significantly inhibited the expression of the transcription repressor and apoptotic resistant factor YY1 and, in parallel, augmented the presence of total p53. The capacity of GIT-27NO to induce p53-mediated apoptosis along with inhibition of YY1 repressor in A375 melanoma cells indicates that GIT-27NO possesses an important anti-cancer pharmacological profile. The findings suggest the potential therapeutic use of GIT-27NO in the clinical setting.


Subject(s)
Apoptosis/drug effects , Melanoma/drug therapy , Nitric Oxide Donors/pharmacology , Tumor Suppressor Protein p53/drug effects , Acetates , Antineoplastic Agents , Cell Line, Tumor , Cell Survival/drug effects , Humans , Melanoma/pathology , Nitric Oxide Donors/therapeutic use , Oxazoles , YY1 Transcription Factor/antagonists & inhibitors
4.
Bioorg Med Chem ; 16(10): 5683-94, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18406151

ABSTRACT

The present study identifies xanthones gentiakochianin and gentiacaulein as the active principles responsible for the in vitro antiglioma action of ether and methanolic extracts of the plant Gentiana kochiana. Gentiakochianin and gentiacaulein induced cell cycle arrest in G(2)/M and G(0)/G(1) phases, respectively, in both C6 rat glioma and U251 human glioma cell lines. The more efficient antiproliferative action of gentiakochianin was associated with its ability to induce microtubule stabilization in a cell-free assay. Both the xanthones reduced mitochondrial membrane potential and increased the production of reactive oxygen species in glioma cells, but only the effects of gentiakochianin were pronounced enough to cause caspase activation and subsequent apoptotic cell death. The assessment of structure-activity relationship in a series of structurally related xanthones from G. kochiana and Gentianella austriaca revealed dihydroxylation at positions 7, 8 of the xanthonic nucleus as the key structural feature responsible for the ability of gentiakochianin to induce microtubule-associated G(2)/M cell block and apoptotic cell death in glioma cells.


Subject(s)
Antineoplastic Agents/pharmacology , Gentiana/chemistry , Glioma/drug therapy , Plant Extracts/pharmacology , Xanthones/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Cell Cycle/drug effects , Cell Death/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , G1 Phase/drug effects , G2 Phase/drug effects , Glioma/pathology , Humans , Mitochondrial Membranes/drug effects , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Rats , Reactive Oxygen Species/antagonists & inhibitors , Resting Phase, Cell Cycle/drug effects , Structure-Activity Relationship , Xanthones/chemistry , Xanthones/isolation & purification
5.
Mol Cancer Ther ; 7(3): 510-20, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18347138

ABSTRACT

Preclinical studies have shown that nitric oxide (NO)-donating nonsteroidal anti-inflammatory drugs possess anticancer activities. Here, we report in vitro and in vivo studies showing the antitumor effect of the NO-donating isoxazole derivative (S,R)-3-phenyl-4,5-dihydro-5-isoxazole acetic acid (GIT-27NO). GIT-27NO, but not the NO-deprived parental compound VGX-1027, significantly affected viability of both rodent (L929, B16, and C6) and human (U251, BT20, HeLa, and LS174) tumor cell lines. GIT-27NO triggered either apoptotic cell death (e.g., L929 cells) or autophagic cell death (C6 and B16 cells). Moreover, GIT-27NO hampered the viability of cisplatin-resistant B16 cells. NO scavenger hemoglobin completely prevented GIT-27NO-induced death, indicating that NO release mediated the tumoricidal effect of the compound. Increase in intracellular NO upon on the treatment was associated with intensified production of reactive oxygen species, whereas their neutralization by antioxidant N-acetylcysteine resulted in partial recovery of cell viability. The antitumor activity of the drug was mediated by the selective activation of mitogen-activated protein kinases in a cell-specific manner and was neutralized by their specific inhibitors. In vivo treatment with GIT-27NO significantly reduced the B16 melanoma growth in syngeneic C57BL/6 mice. The therapeutic effect occurred at dose (0.5 mg/mouse) up to 160 times lower than those needed to induce acute lethality (80 mg/mouse). In addition, a dose of GIT-27NO five times higher than that found effective in the melanoma model was well tolerated by the mice when administered for 4 consecutive weeks. These data warrant additional studies to evaluate the possible translation of these findings to the clinical setting.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Nitric Oxide Donors/pharmacology , Oxazoles/pharmacology , Animals , Antineoplastic Agents/toxicity , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Nitric Oxide Donors/toxicity
6.
Eur J Pharmacol ; 583(1): 1-10, 2008 Mar 31.
Article in English | MEDLINE | ID: mdl-18262519

ABSTRACT

To explore combined antiglioma effect of nitric oxide (NO) and hyperthermia, the rat C6 and human U251 glioma cells were exposed to NO-releasing agents sodium nitroprusside(SNP), S-nitrosoglutathione or PAPA-NONOate, followed by hyperthermia (1 h, 43 degrees C). While each treatment alone showed only moderate efficiency, a synergistic cytotoxicity of NO donors and hyperthermia was clearly demonstrated by crystal violet and MTT cytotoxicity assays. The flow cytometric analysis with the appropriate reporter fluorochromes confirmed that hyperthermia and SNP cooperated in inducing oxidative stress, mitochondrial depolarization, caspase activation and DNA fragmentation, leading to both necrotic and caspase-dependent apoptotic cell death. The acridine orange staining of intracellular acidic compartments revealed that SNP completely blocked hyperthermia-induced autophagy, while the inhibition of autophagy by 3-methyl adenine mimicked SNP-triggered oxidative stress, caspase activation and cell death in hyperthermia-exposed cells. Therefore, the synergistic cytotoxicity of SNP and hyperthermia could result from NO-mediated suppression of protective autophagic response in glioma cells.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Hyperthermia, Induced , Nitric Oxide/therapeutic use , Vasodilator Agents/therapeutic use , Animals , Autophagy/drug effects , Autophagy/physiology , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Combined Modality Therapy , DNA Fragmentation/drug effects , Enzyme Activation , Flow Cytometry , Humans , Hydrazines/therapeutic use , Mitochondria/drug effects , Mitochondria/physiology , Nitric Oxide Donors/therapeutic use , Nitrites/metabolism , Nitroprusside/therapeutic use , Oxidative Stress/drug effects , Rats , S-Nitrosoglutathione/therapeutic use
7.
Pharm Res ; 25(6): 1365-76, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17999162

ABSTRACT

PURPOSE: The fullerene (C60/C70 mixture-C60/70) nanocrystalline suspension prepared by solvent exchange method using tetrahydrofyran (THF/nC60/70) and polyhydroxylated C60/70 [C60/70(OH)n] were compared for their ability to modulate cytotoxicity of the proinflammatory cytokine tumor necrosis factor (TNF). MATERIALS AND METHODS: TNF-induced cytotoxicity was assessed in L929 fibrosarcoma cells by crystal violet assay. The type of cell death (apoptosis/necrosis), production of reactive oxygen species, mitochondrial depolarization and caspase activation were determined by flow cytometry using the appropriate reporter dyes. RESULTS: THF/nC60/70 augmented, while C60/70(OH)n reduced the cytotoxicity of TNF. The numbers of cells undergoing apoptosis/necrosis, as well as of those displaying the activation of apoptosis-inducing enzymes of caspase family, were respectively increased or reduced by THF/nC60/70 or C60/70(OH)n. The antioxidant N-acetylcysteine and mitochondrial permeability transition inhibitor cyclosporin A each partly blocked the cytotoxic action of TNF, indicating the involvement of oxidative stress and mitochondrial dysfunction in the TNF cytotoxicity. Accordingly, THF/nC60/70 or C60/70(OH)n potentiated or suppressed, respectively, TNF-triggered oxidative stress and mitochondrial depolarization. CONCLUSION: The ability of different fullerene preparations to modulate TNF-induced oxidative stress and subsequent cell death suggests their potential value in the TNF-based cancer therapy or prevention of TNF-dependent tissue damage.


Subject(s)
Apoptosis/drug effects , Fullerenes/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Caspases/metabolism , Cell Line, Tumor , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Necrosis , Reactive Oxygen Species/metabolism
8.
J Cell Physiol ; 215(3): 665-75, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18064633

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine of the innate immune system that plays a major role in the induction of immunoinflammatory responses. To examine the role of endogenous MIF in the pathogenesis of type 1 diabetes (TID) we evaluated the effects of administration of neutralizing anti-MIF antibodies to NOD mice with accelerated forms of diabetes induced by injection of cyclophosphamide or by transfer of diabetogenic spleen cells. Both accelerated forms of diabetes were markedly reduced by anti-MIF antibody. Furthermore, MIF-deficient (MIF(-/-)) mice were less susceptible to the induction of immunoinflammatory diabetes, insulitis and apoptosis within the endocrine pancreas by multiple low doses of streptozotocin (MLD-STZ) than genetically matched wild type (WT) mice. MIF deficiency resulted in lower proliferation and lymphocyte adhesion, as well as reduced production from the spleens and peritoneal cells of a variety of inflammatory mediators typically associated with development of the disease including IL-12, IL-23, TNF-alpha, and IL-1beta. Furthermore, MIF deletion affected the production of IL-18, TNF-alpha, IL-1beta, and iNOS in the islets of Langerhans. These data, along with the higher expression of IL-4 and TGF-beta observed in the periphery and in the pancreas of MLD-STZ-challenged MIF(-/-) mice as compared to WT controls suggest that MIF deficiency has induced an immune deviation towards protective type 2/3 response. These results suggest that MIF participates in T1D by controlling the functional activity of monocytes/macrophages and T cells and modulating their secretory capacity of pro- and anti-inflammatory molecules.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Adoptive Transfer , Animals , Antibodies, Monoclonal/pharmacology , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 1/chemically induced , Disease Progression , Female , Gene Expression Regulation/drug effects , Intramolecular Oxidoreductases/deficiency , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Islets of Langerhans/pathology , Leukocytes, Mononuclear/drug effects , Macrophage Migration-Inhibitory Factors/deficiency , Mice , Mice, Inbred NOD , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spleen/drug effects , Spleen/pathology , Streptozocin , Tyrosine/analogs & derivatives , Tyrosine/metabolism
9.
Biomaterials ; 28(36): 5437-48, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17884160

ABSTRACT

Because of the ability to induce cell death in certain conditions, the fullerenes (C(60)) are potential anticancer and toxic agents. The colloidal suspension of crystalline C(60) (nano-C(60), nC(60)) is extremely toxic, but the mechanisms of its cytotoxicity are not completely understood. By combining experimental analysis and mathematical modelling, we investigate the requirements for the reactive oxygen species (ROS)-mediated cytotoxicity of different nC(60) suspensions, prepared by solvent exchange method in tetrahydrofuran (THF/nC(60)) and ethanol (EtOH/nC(60)), or by extended mixing in water (aqu/nC(60)). With regard to their capacity to generate ROS and cause mitochondrial depolarization followed by necrotic cell death, the nC(60) suspensions are ranked in the following order: THF/nC(60)>EtOH/nC(60)>aqu/nC(60). Mathematical modelling of singlet oxygen ((1)O(2)) generation indicates that the (1)O(2)-quenching power (THF/nC(60)

Subject(s)
Fullerenes/toxicity , Reactive Oxygen Species/metabolism , Animals , Cell Line , Cell Survival/drug effects , Colloids , Humans , Mice , Solvents
10.
Eur J Pharmacol ; 568(1-3): 89-98, 2007 Jul 30.
Article in English | MEDLINE | ID: mdl-17560995

ABSTRACT

Using the rat glioma cell line C6 and the human glioma cell line U251, we demonstrate the multiple mechanisms underlying the in vitro anticancer effects of the C(60) fullerene water suspension (nano-C(60) or nC(60)) produced by solvent exchange method. Nano-C(60) in a dose-dependent manner reduced the tumor cell numbers after 24 h of incubation. The observed antiglioma action of nC(60) at high concentration (1 microg/ml) was due to a reactive oxygen species-mediated necrotic cell damage that was partly dependent on oxidative stress-induced activation of extracellular signal-regulated kinase (ERK). On the other hand, low-dose nC(60) (0.25 microg/ml) did not induce either necrotic or apoptotic cell death, but caused oxidative stress/ERK-independent cell cycle block in G(2)/M phase and subsequent inhibition of tumor cell proliferation. Treatment with either high-dose or low-dose nC(60) caused the appearance of acidified intracytoplasmic vesicles indicative of autophagy, but only the antiglioma effect of low-dose nC(60) was significantly attenuated by inhibiting autophagy with bafilomycin A1. Importantly, primary rat astrocytes were less sensitive than their transformed counterparts to a cytostatic action of low-dose nC(60). These data provide grounds for further development of nC(60) as an anticancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Fullerenes/pharmacology , Glioma/drug therapy , Nanoparticles , Animals , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Glial Fibrillary Acidic Protein/metabolism , Glioma/metabolism , Humans , Lipid Peroxidation/drug effects , Mitogen-Activated Protein Kinases/metabolism , Rats , Reactive Oxygen Species/metabolism
11.
Eur J Pharmacol ; 568(1-3): 248-59, 2007 Jul 30.
Article in English | MEDLINE | ID: mdl-17531219

ABSTRACT

We demonstrate the capacity of an herbal anthraquinone aloe emodin to reduce the cytotoxicity of the proinflammatory cytokine tumor necrosis factor (TNF) towards L929 mouse fibrosarcoma and U251 human glioma cell lines. Aloe emodin inhibited both TNF-induced cell necrosis and apoptosis, but it did not reduce cell death induced by UV radiation or hydrogen peroxide. Aloe emodin inhibited both basal and TNF-triggered activation of extracellular signal-regulated kinase (ERK), and a selective blockade of ERK activation mimicked the cytoprotective action of the drug. On the other hand, aloe emodin did not affect TNF-induced activation of p38 mitogen-activated protein kinase or generation of reactive oxygen species. The combination of aloe emodin and TNF caused an intracellular appearance of acidified autophagic vesicles, and the inhibition of autophagy with bafilomycin or 3-methyladenine efficiently blocked the cytoprotective action of aloe emodin. These data indicate that aloe emodin could prevent TNF-triggered cell death through mechanisms involving induction of autophagy and blockade of ERK activation.


Subject(s)
Autophagy/drug effects , Emodin/pharmacology , Tumor Necrosis Factor Inhibitors , Tumor Necrosis Factors/pharmacology , Animals , Anthraquinones , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Necrosis/drug therapy , Reactive Oxygen Species/metabolism , Recombinant Proteins/pharmacology
12.
J Pharmacol Exp Ther ; 320(3): 1038-49, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17148780

ABSTRACT

(S,R)-3-Phenyl-4,5-dihydro-5-isoxasole acetic acid (VGX-1027) is an isoxazole compound that exhibits various immunomodulatory properties. The capacity of VGX-1027 to prevent interleukin (IL)-1beta plus interferon-gamma-induced pancreatic islet death in vitro prompted us to evaluate its effects on the development of autoimmune diabetes in preclinical models of human type 1 diabetes mellitus (T1D). Administration of VGX-1027 to NOD mice with spontaneous or accelerated forms of diabetes induced either by injection of cyclophosphamide or by transfer of spleen cells from acutely diabetic syngeneic donors markedly reduced the cumulative incidence of diabetes and insulitis. In addition, VGX-1027 given either i.p. or p.o. to CBA/H mice made diabetic with multiple low doses of streptozotocin successfully counteracted the development of destructive insulitis and hyperglycemia. The animals receiving VGX-1027 exhibited reduced production of the proinflammatory mediators tumor necrosis factor-alpha, IL-1beta, macrophage migration inhibitory factor, and inducible nitric-oxide synthase-mediated nitric oxide generation in both pancreatic islets and peripheral compartments. These results indicate that VGX-1027 probably exerts its antidiabetogenic effects by limiting cytokine-mediated immunoinflammatory events, leading to inflammation and destruction of pancreatic islets. VGX-1027 seems worthy of being considered as a candidate drug in the development of new therapeutic strategies for the prevention and early treatment of T1D.


Subject(s)
Acetates/therapeutic use , Cytokines/immunology , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Immunologic Factors/therapeutic use , Inflammation Mediators/immunology , Oxazoles/therapeutic use , Acetates/chemistry , Acetates/pharmacokinetics , Animals , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Cyclophosphamide/pharmacology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/prevention & control , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Immunologic Factors/chemistry , Immunologic Factors/pharmacokinetics , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred CBA , Mice, Inbred NOD , Molecular Structure , Nitric Oxide/biosynthesis , Oxazoles/chemistry , Oxazoles/pharmacokinetics , Reverse Transcriptase Polymerase Chain Reaction , Streptozocin
13.
Biomaterials ; 27(29): 5049-58, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16784774

ABSTRACT

We investigated the effect of gamma-irradiation on the cytotoxicity of pure C60 solubilized in water by using tetrahydrofuran (THF/n-C60 or THF/n-C60). In contrast to THF/n-C60, its gamma-irradiated counterpart failed to generate oxygen radicals and cause extracellular signal-regulated kinase (ERK)-dependent necrotic cell death in various types of mammalian cells. Moreover, gamma-irradiated THF/n-C60 protected cells from the oxidative stress induced by native THF/n-C60 or hydrogen peroxide. The observed biological effects were associated with gamma-irradiation-mediated decomposition of THF and subsequent derivatization of the n-C60 surface. These results for the first time demonstrate gamma-irradiation-mediated changes in the physico-chemical properties of THF-prepared nanocrystalline C60, resulting in a complete loss of its cytotoxic effect and its conversion to a cytoprotective agent.


Subject(s)
Fullerenes/toxicity , Gamma Rays , Nanostructures , Animals , Cell Line, Tumor , Fullerenes/radiation effects , Furans/radiation effects , Furans/toxicity , Mice
14.
Toxicol Sci ; 91(1): 173-83, 2006 May.
Article in English | MEDLINE | ID: mdl-16476688

ABSTRACT

The mechanisms underlying the cytotoxic action of pure fullerene suspension (nano-C60) and water-soluble polyhydroxylated fullerene [C60(OH)n] were investigated. Crystal violet assay for cell viability demonstrated that nano-C60 was at least three orders of magnitude more toxic than C60(OH)n to mouse L929 fibrosarcoma, rat C6 glioma, and U251 human glioma cell lines. Flow cytometry analysis of cells stained with propidium iodide (PI), PI/annexin V-fluorescein isothiocyanate, or the redox-sensitive dye dihydrorhodamine revealed that nano-C60 caused rapid (observable after few hours), reactive oxygen species (ROS)-associated necrosis characterized by cell membrane damage without DNA fragmentation. In contrast, C60(OH)n caused delayed, ROS-independent cell death with characteristics of apoptosis, including DNA fragmentation and loss of cell membrane asymmetry in the absence of increased permeability. Accordingly, the antioxidant N-acetylcysteine protected the cell lines from nano-C60 toxicity, but not C60(OH)n toxicity, while the pan-caspase inhibitor z-VAD-fmk blocked C60(OH)n-induced apoptosis, but not nano-C60-mediated necrosis. Finally, C60(OH)n antagonized, while nano-C60 synergized with, the cytotoxic action of oxidative stress-inducing agents hydrogen peroxide and peroxynitrite donor 3-morpholinosydnonimine. Therefore, unlike polyhydroxylated C60 that exerts mainly antioxidant/cytoprotective and only mild ROS-independent pro-apoptotic activity, pure crystalline C60 seems to be endowed with strong pro-oxidant capacity responsible for the rapid necrotic cell death.


Subject(s)
Fullerenes/pharmacology , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor , Flow Cytometry , Humans , Hydroxylation , Mice , Rats , Reactive Oxygen Species
15.
Free Radic Biol Med ; 40(2): 226-35, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16413405

ABSTRACT

The influence of environmental pH on the production of tumoricidal free radical nitric oxide (NO) was investigated in mouse fibrosarcoma L929 and rat glioma C6 cell lines. A combination of IFN-gamma and IL-1 induced a significant NO release and subsequent reduction of cell viability in tumor cell lines. Acidification of cell culture medium reduced tumor cell NO production in a pH-dependent manner. While the inhibitory effect of acidosis on NO production in C6 cells was associated with a further decrease in cell viability, it completely rescued L929 cells from NO-dependent apoptotic and necrotic death. Acidic pH diminished IFN-gamma+ IL-1-induced expression of inducible NO synthase (iNOS) mRNA and protein, and abolished the activation of iNOS transcription factor IRF-1 in L929 cells. Moreover, extracellular acidosis significantly impaired cytokine-induced phosphorylation of MAP kinase p44/42 (ERK1/2) and subsequent expression of transcription factor c-Fos in L929 cells. Finally, mild acidosis (pH 6.8) augmented, while severe acidosis (pH 6.0) reduced, IFN-gamma-induced iNOS activation/NO release and NO-dependent anticancer activity of rat and mouse macrophages. Taken together, our findings indicate that modulation of macrophage and tumor cell iNOS by an acidic microenvironment might influence the progression of NO-sensitive solid tumors.


Subject(s)
Acidosis/metabolism , Fibrosarcoma/metabolism , Glioma/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/biosynthesis , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Fibrosarcoma/drug therapy , Free Radicals/metabolism , Glioma/drug therapy , Hydrogen-Ion Concentration , Interferon Regulatory Factor-1/antagonists & inhibitors , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Macrophages/drug effects , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/genetics , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/drug effects , RNA, Messenger/genetics , Rats , Tumor Cells, Cultured
16.
Eur J Pharmacol ; 517(1-2): 28-34, 2005 Jul 04.
Article in English | MEDLINE | ID: mdl-15970285

ABSTRACT

There has been a continuing effort for the discovery of novel platinum(IV)-based antitumor compounds with better therapeutic performances than cisplatin. In the present work, the anticancer action of recently synthesized Pt(IV)-based complex [Pt(HPxSC)Cl(3)] was investigated using rat and human astrocytoma cell lines C6 and U251. [Pt(HPxSC)Cl(3)] markedly reduced the number of cultured astrocytoma cells (IC(50), 80 microM), as determined by crystal violet assay. The Pt(IV) complex induced apoptotic death of tumor cells, as flow cytometry analysis of the propidium iodide-stained cellular DNA revealed approx. 30% of hypodiploid cells in [Pt(HPxSC)Cl(3)]-treated astrocytoma cell cultures. On the other hand, [Pt(HPxSC)Cl(3)] at 200 microM did not affect the viability of rat primary astrocytes, unlike the established anticancer drug cisplatin, which displayed high toxicity toward both astrocytoma cells (IC(50), 15 microM) and primary astrocytes (IC(50), 20 microM). Moreover, [Pt(HPxSC)Cl(3)] at 100 microM did not interfere with the ability of rat peritoneal macrophages to produce important antitumor molecules nitric oxide and tumor necrosis factor-alpha. Finally, we assessed the ability of [Pt(HPxSC)Cl(3)] to restrain growth of some bacterial and yeast strains, but it showed rather limited antimicrobial activity.


Subject(s)
Antineoplastic Agents/pharmacology , Organoplatinum Compounds/pharmacology , Platinum Compounds/pharmacology , Animals , Astrocytoma/drug therapy , Astrocytoma/metabolism , Astrocytoma/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , DNA, Neoplasm/metabolism , Dose-Response Relationship, Drug , Humans , Interferon-gamma/pharmacology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Microbial Sensitivity Tests , Nitric Oxide/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism
17.
Neuropharmacology ; 48(5): 720-31, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15814106

ABSTRACT

The role of iron in 6-hydroxydopamine (6-OHDA) toxicity towards astrocytes was investigated in vitro using rat primary astrocytes, rat astrocytoma cell line C6, and human astrocytoma cell line U251. The assessment of mitochondrial respiration or lactate dehydrogenase release has shown a dose-dependent decrease in the viability of astrocytes treated with 6-OHDA, which coincided with DNA fragmentation and the changes in cellular morphology. This was a consequence of the oxidative stress mediated by 6-OHDA autoxidation products hydrogen peroxide, superoxide anion, and hydroxyl radical. Both FeSO(4) and FeCl(3) markedly alleviated detrimental effects of 6-OHDA treatment, while MgSO(4) was without effect. The protective action of iron was neutralized by a membrane-permeable iron chelator o-phenanthroline, which also augmented astrocyte killing in the absence of exogenous iron. The mechanisms responsible for iron-mediated protection of astrocytes did not involve interference with either 6-OHDA autoxidation, hydrogen peroxide toxicity, or 6-OHDA-induced activation of extracellular signal-regulated kinase. Finally, the addition of iron potentiated and its chelation blocked 6-OHDA toxicity towards neuronal PC12 cells, suggesting the opposite roles for this transition metal in regulating the survival of astrocytes and dopaminergic neurons.


Subject(s)
Astrocytes/drug effects , Hydroxydopamines/toxicity , Iron/pharmacology , Animals , Animals, Newborn , Cell Adhesion Molecules/metabolism , Cell Survival/drug effects , Cells, Cultured , DNA Fragmentation/drug effects , DNA Fragmentation/physiology , Dose-Response Relationship, Drug , Drug Interactions , Ferric Compounds/pharmacology , Flow Cytometry/methods , Iron Chelating Agents/pharmacology , Junctional Adhesion Molecules , L-Lactate Dehydrogenase/metabolism , PC12 Cells/drug effects , PC12 Cells/metabolism , Phenanthrolines/pharmacology , Rats , Rats, Wistar , Staining and Labeling/methods , Tetrazolium Salts , Thiazoles , Time Factors
18.
Glia ; 47(2): 168-79, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15185395

ABSTRACT

Although astrocytes presumably participate in maintaining the immune privilege of the central nervous system (CNS), the mechanisms behind their immunoregulatory properties are still largely undefined. In this study, we describe the development of regulatory T cells upon contact with astrocytes. Rat T cells pre-incubated with astrocytes completely lost the ability to proliferate in response to mitogenic stimuli. The cells were blocked in G0/G1 phase of the cell cycle, expressed less IL-2R, and produced significantly lower amounts of interferon-gamma (IFN-gamma), but not interleukin-2 (IL-2), IL-10, or tumor necrosis factor (TNF). These anergic cells completely prevented mitogen-induced growth of normal T lymphocytes, as well as CNS antigen-driven proliferation of autoreactive T cells. The suppressive activity resided in both CD4+ and CD8+ T-cell compartments. Heat-sensitive soluble T-cell factors, not including transforming growth factor-beta (TGF-beta) or IL-10, were solely responsible for the observed suppression, as well as for the transfer of suppressive activity to normal T cells. The administration of astrocyte-induced regulatory T cells markedly alleviated CNS inflammation and clinical symptoms of CNS autoimmunity in rats with experimental allergic encephalomyelitis. Finally, the cells with suppressive properties were readily generated from human lymphocytes after contact with astrocytes. Taken together, these data indicate that astrocyte-induced regulatory T cells might represent an important mechanism for self-limitation of excessive inflammation in the brain.


Subject(s)
Astrocytes/immunology , Autoimmune Diseases of the Nervous System/immunology , Cell Communication/immunology , Immune Tolerance/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Animals, Newborn , Autoimmune Diseases of the Nervous System/physiopathology , Cell Cycle/immunology , Cell Division/immunology , Cells, Cultured , Cytokines/biosynthesis , Disease Models, Animal , Encephalitis/immunology , Encephalitis/physiopathology , Encephalitis/therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Humans , Mitogens/pharmacology , Rats , T-Lymphocytes/drug effects , T-Lymphocytes/transplantation
19.
Eur J Pharmacol ; 485(1-3): 81-8, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-14757126

ABSTRACT

Given the important role of gaseous free radical nitric oxide (NO) in tumor cell biology, we investigated the ability of the anti-cancer drugs 5-Aza-2'-deoxycytidine (ADC) and paclitaxel to modulate NO production in mouse L929 fibrosarcoma cells. Both drugs reduced IFN-gamma-stimulated NO release in cultures of L929 and primary fibroblasts, but not in mouse peritoneal macrophages. The inhibitory effect was due to the reduced expression of inducible NO synthase (iNOS), the enzyme responsible for cytokine-induced intracellular NO synthesis, as both agents markedly suppressed the interferon-gamma (IFN-gamma)-triggered increase in iNOS concentration in L929 cells. In addition, ADC and paclitaxel prevented the IFN-gamma-triggered activation of p44/p42 mitogen-activated protein (MAP) kinase in L929 fibroblasts, suggesting a possible mechanism for the observed inhibition of iNOS expression. These results might have important implications for the therapeutic effect of ADC and paclitaxel, since their inhibitory action on NO release partly neutralized the NO-dependent toxicity of IFN-gamma on L929 fibrosarcoma cells.


Subject(s)
Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Enzyme Inhibitors/pharmacology , Fibrosarcoma/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Paclitaxel/pharmacology , Animals , Azacitidine/therapeutic use , Cell Line, Tumor , Decitabine , Dose-Response Relationship, Drug , Enzyme Inhibitors/therapeutic use , Fibrosarcoma/drug therapy , Mice , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Paclitaxel/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...