Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Molecules ; 22(9)2017 Sep 08.
Article En | MEDLINE | ID: mdl-28885586

Natural products are considered as an important source for the discovery of new drugs to treat aging-related degenerative diseases and liver injury. The present study profiled the chemical constituents of a methanol extract from Senna singueana bark using HPLC-PDA-ESI-MS/MS and 36 secondary metabolites were identified. Proanthocyanidins dominated the extract. Monomers, dimers, trimers of (epi)catechin, (epi)gallocatechin, (epi)guibourtinidol, (ent)cassiaflavan, and (epi)afzelechin represented the major constituents. The extract demonstrated notable antioxidant activities in vitro: In DPPH (EC50 of 20.8 µg/mL), FRAP (18.16 mM FeSO4/mg extract) assays, and total phenolic content amounted 474 mg gallic acid equivalent (GAE)/g extract determined with the Folin-Ciocalteu method. Also, in an in vivo model, the extract increased the survival rate of Caenorhabditis elegans worms pretreated with the pro-oxidant juglone from 43 to 64%, decreased intracellular ROS inside the wild-type nematodes by 47.90%, and induced nuclear translocation of the transcription factor DAF-16 in the transgenic strain TJ356. Additionally, the extract showed a remarkable hepatoprotective activity against d-galactosamine (d-GalN) induced hepatic injury in rats. It significantly reduced elevated AST (aspartate aminotransferase), and total bilirubin. Moreover, the extract induced a strong cytoplasmic Bcl-2 expression indicating suppression of apoptosis. In conclusion, the bark extract of S. sengueana represents an interesting candidate for further research in antioxidants and liver protection.


Antioxidants/pharmacology , Caenorhabditis elegans/drug effects , Chemical and Drug Induced Liver Injury/prevention & control , Protective Agents/pharmacology , Senna Plant/chemistry , Animals , Antioxidants/chemistry , Antioxidants/isolation & purification , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/genetics , Bilirubin/blood , Biphenyl Compounds/antagonists & inhibitors , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Catechols/chemistry , Catechols/isolation & purification , Catechols/pharmacology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Galactosamine/toxicity , Male , Methanol/chemistry , Naphthoquinones/antagonists & inhibitors , Naphthoquinones/pharmacology , Oxidants/antagonists & inhibitors , Oxidants/pharmacology , Phenols/chemistry , Phenols/isolation & purification , Phenols/pharmacology , Picrates/antagonists & inhibitors , Plant Bark/chemistry , Plant Extracts/chemistry , Proanthocyanidins/chemistry , Proanthocyanidins/isolation & purification , Proanthocyanidins/pharmacology , Protective Agents/chemistry , Protective Agents/isolation & purification , Proto-Oncogene Proteins c-bcl-2/agonists , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Solvents/chemistry
2.
Sci Rep ; 7(1): 4814, 2017 07 06.
Article En | MEDLINE | ID: mdl-28684787

Electrophiles can activate redox signal transduction pathways, through actions of effector molecules (e.g., kinases and transcription factors) and sensor proteins with low pKa thiols that are covalently modified. In this study, we investigated whether 1,4-naphthoquinone (1,4-NQ) could affect the phosphatase and tensin homolog (PTEN)-Akt signaling pathway and persulfides/polysulfides could modulate this adaptive response. Simultaneous exposure of primary mouse hepatocytes to Na2S4 and 1,4-NQ markedly decreased 1,4-NQ-mediated cell death and S-arylation of cellular proteins. Modification of cellular PTEN during exposure to 1,4-NQ was also blocked in the presence of Na2S4. 1,4-NQ, at up to 10 µM, increased phosphorylation of Akt and cAMP response element binding protein (CREB). However, at higher concentrations, 1,4-NQ inhibited phosphorylation of both proteins. These bell-shaped dose curves for Akt and CREB activation were right-shifted in cells treated with both 1,4-NQ and Na2S4. Incubation of 1,4-NQ with Na2S4 resulted in formation of 1,4-NQ-S-1,4-NQ-OH. Unlike 1,4-NQ, authentic 1,4-NQ-S-1,4-NQ-OH adduct had no cytotoxicity, covalent binding capability nor ability to activate PTEN-Akt signaling in cells. Our results suggested that polysulfides, such as Na2S4, can increase the threshold of 1,4-NQ for activating PTEN-Akt signaling and cytotoxicity by capturing this electrophile to form its sulfur adducts.


Cyclic AMP Response Element-Binding Protein/genetics , Naphthoquinones/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Protective Agents/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Sodium Compounds/pharmacology , Sulfides/pharmacology , Animals , Cell Death/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Gene Expression Regulation , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Mice , Mice, Inbred C57BL , Naphthoquinones/toxicity , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Primary Cell Culture , Protective Agents/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sodium Compounds/chemistry , Sulfides/chemistry
3.
Microbiol Immunol ; 61(6): 232-238, 2017 Jun.
Article En | MEDLINE | ID: mdl-28463406

JC polyomavirus (JCPyV) is the causative agent of the demyelinating disease of the central nervous system known as progressive multifocal leukoencephalopathy (PML), which occurs in immunocompromised patients. Moreover, patients treated with natalizumab for multiple sclerosis or Crohn disease can develop PML, which is then termed natalizumab-related PML. Because few drugs are currently available for treating PML, many antiviral agents are being investigated. It has been demonstrated that the topoisomerase I inhibitors topotecan and ß-lapachone have inhibitory effects on JCPyV replication in IMR-32 cells. However, both of these drugs have marginal inhibitory effects on virus propagation in JC1 cells according to RT-PCR analysis. In the present study, the inhibitory effect of another topoisomerase I inhibitor, 7-ethy-10-[4-(1-piperidino)-1-piperidino] carbonyloxy camptothecin (CPT11), was assessed by investigating viral replication, propagation, and viral protein 1 (VP1) production in cultured cells. JCPyV replication was assayed using real-time PCR combined with Dpn I treatment in IMR-32 cells transfected with JCPyV DNA. It was found that JCPyV replicates less in IMR-32 cells treated with CPT11 than in untreated cells. Moreover, CPT11 treatment of JCI cells persistently infected with JCPyV led to a dose-dependent reduction in JCPyV DNA and VP1 production. Additionally, the inhibitory effect of CPT11 was found to be stronger than those of topotecan and ß-lapachone. These findings suggest that CPT11 may be a potential anti-JCPyV agent that could be used to treat PML.


Antiviral Agents/antagonists & inhibitors , Camptothecin/antagonists & inhibitors , JC Virus/drug effects , Virus Replication/drug effects , Camptothecin/administration & dosage , Camptothecin/toxicity , Cell Line/drug effects , Cell Line/virology , Cell Proliferation/drug effects , DNA Replication/drug effects , DNA, Viral/genetics , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/drug therapy , Naphthoquinones/antagonists & inhibitors , Real-Time Polymerase Chain Reaction/methods , Topoisomerase I Inhibitors/pharmacology , Topotecan/antagonists & inhibitors , Viral Proteins/drug effects
4.
Free Radic Biol Med ; 99: 352-363, 2016 10.
Article En | MEDLINE | ID: mdl-27567538

Reactive oxygen species (ROS) signaling is critical in T helper (Th) cell differentiation; however its role in differentiated Th cell functions is unclear. In this study, we investigated the role of oxidative stress on the effector functions of in vitro differentiated mouse Th17 and Th1 cells or CD4+ T cells from patients with Rheumatoid Arthritis using pro-oxidants plumbagin (PB) and hydrogen peroxide. We found that in mouse Th cells, non-toxic concentration of pro-oxidants inhibited reactivation induced expression of IL-17A in Th17 and IFN-γ in Th1 cells by reducing the expression of their respective TFs, RORγt and T-bet. Interestingly, in both the subsets, PB increased the expression of IL-4 by enhancing reactivation induced ERK1/2 phosphorylation. We further investigated the cytokine modulatory effect of PB on CD4+ T cells isolated from PBMCs of patients with Rheumatoid Arthritis, a well-known Th17 and or Th1 mediated disease. In human CD4+ T cells from Rheumatoid Arthritis patients, PB reduced the frequencies of IL-17A+ (Th17), IFN-γ+ (Th1) and IL-17A+/IFN-γ+ (Th17/1) cells and also inhibited the production of pro-inflammatory cytokines TNF-α and IL-6. N-Acetyl Cysteine (NAC) an antioxidant completely reversed PB mediated cytokine modulatory effects in both mouse and human cells indicating a direct role for ROS. Together our data suggest that oxidative microenvironment can alter cytokine response of terminally differentiated cells and thus altering intracellular ROS could be a potential way to target Th17 and Th1 cells in autoimmune disorders.


Arthritis, Rheumatoid/immunology , Cytokines/immunology , Gene Expression Regulation/immunology , Oxidative Stress/immunology , Th1 Cells/drug effects , Th17 Cells/drug effects , Acetylcysteine/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Case-Control Studies , Cell Differentiation , Cytokines/genetics , Female , Humans , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Inbred BALB C , Middle Aged , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Naphthoquinones/antagonists & inhibitors , Naphthoquinones/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Oxidants/antagonists & inhibitors , Oxidants/pharmacology , Phosphorylation/drug effects , Primary Cell Culture , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
5.
Drug Discov Ther ; 10(2): 93-102, 2016.
Article En | MEDLINE | ID: mdl-27074996

Detrimental effects of ionizing radiation (IR) are observed at the doses above 1 Gy. Treatment modalities are available up to doses of 6 Gy including bonemarrow transplantation and administration of antibiotics. However, exposure to IR doses above 8 Gy results in gastro-intestinal (GI) syndrome characterised by denudated villi, apoptosis of crypt cells and elevated inflammatory responses. Multiple strategies have been employed to investigate novel agents to protect against IR induced injury. Since cellular redox homeostasis plays a pivotal role in deciding the cell fate, present study was undertaken to explore the potential of 1,4-naphthoquinone (NQ), a pro-oxidant, to ameliorate IR induced GI syndrome. NQ protected INT 407 cells against IR induced cell death of intestinal epithelial cells in vitro. NQ induced perturbation in cellular redox status and induced the activation of nuclear factor-erythroid 2-related factor 2 (Nrf2) pathway. Thiol antioxidant and inhibitors of Nrf2 pathway abrogated the radioprotection offered by NQ. Further, knocking down Nrf2 rescind the NQ mediated protection against IR induced cell death. In conclusion, NQ protects against IR radiation induced GI syndrome in vitro by perturbing cellular redox and activating Nrf2 pathway. This is the first report highlighting the potential of a pro-oxidant to ameliorate IR induced GI injury.


Gastrointestinal Diseases/drug therapy , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Naphthoquinones/therapeutic use , Oxidants/therapeutic use , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/therapeutic use , Animals , Antioxidants/pharmacology , Caspase 3/biosynthesis , Caspase 3/genetics , Cell Line , Cell Survival/drug effects , Colony-Forming Units Assay , Gastrointestinal Diseases/etiology , Gene Knockdown Techniques , Humans , Membrane Potential, Mitochondrial/drug effects , Naphthoquinones/antagonists & inhibitors , Naphthoquinones/pharmacology , Oxidation-Reduction , RNA, Small Interfering/pharmacology , Radiation-Protective Agents/pharmacology , Reactive Oxygen Species/metabolism
6.
Nanotechnology ; 27(8): 085103, 2016 Feb 26.
Article En | MEDLINE | ID: mdl-26808118

Chemical-based pesticides are widely used in agriculture to protect crops from insect infestation and diseases. However, the excessive use of highly toxic pesticides causes several human health (neurological, tumor, cancer) and environmental problems. Therefore nanoparticle-based green pesticides have become of special importance in recent years. The antifungal activities of pure and Ag doped (solid and hollow) TiO2 nanoparticles are studied against two potent phytopathogens, Fusarium solani (which causes Fusarium wilt disease in potato, tomato, etc) and Venturia inaequalis (which causes apple scab disease) and it is found that hollow nanoparticles are more effective than the other two. The antifungal activities of the nanoparticles were further enhanced against these two phytopathogens under visible light exposure. The fungicidal effect of the nanoparticles depends on different parameters, such as particle concentration and the intensity of visible light. The minimum inhibitory dose of the nanoparticles for V. inaequalis and F. solani are 0.75 and 0.43 mg/plate. The presence of Ag as a dopant helps in the formation of stable Ag-S and disulfide bonds (R-S-S-R) in cellular protein, which leads to cell damage. During photocatalysis generated (•)OH radicals loosen the cell wall structure and this finally leads to cell death. The mechanisms of the fungicidal effect of nanoparticles against these two phytopathogens are supported by biuret and triphenyl tetrazolium chloride analyses and field emission electron microscopy. Apart from the fungicidal effect, at a very low dose (0.015 mg/plate) the nanoparticles are successful in arresting production of toxic napthoquinone pigment for F. solani which is related to the fungal pathogenecity. The nanoparticles are found to be effective in protecting potatoes affected by F. solani or other fungi from spoiling.


Fungicides, Industrial/pharmacology , Fusarium/drug effects , Nanoparticles/chemistry , Plant Diseases/prevention & control , Saccharomycetales/drug effects , Silver/pharmacology , Titanium/pharmacology , Cell Wall/drug effects , Disulfides/chemistry , Fungicides, Industrial/chemical synthesis , Fusarium/pathogenicity , Fusarium/physiology , Fusarium/radiation effects , Green Chemistry Technology , Hydroxyl Radical/antagonists & inhibitors , Hydroxyl Radical/metabolism , Light , Solanum lycopersicum/drug effects , Solanum lycopersicum/microbiology , Malus/drug effects , Malus/microbiology , Microbial Sensitivity Tests , Mycotoxins/antagonists & inhibitors , Mycotoxins/biosynthesis , Nanoparticles/ultrastructure , Naphthoquinones/antagonists & inhibitors , Naphthoquinones/metabolism , Photochemical Processes , Plant Diseases/microbiology , Saccharomycetales/pathogenicity , Saccharomycetales/physiology , Saccharomycetales/radiation effects , Silver/chemistry , Solanum tuberosum/drug effects , Solanum tuberosum/microbiology , Titanium/chemistry
7.
Toxicol Appl Pharmacol ; 281(3): 285-93, 2014 Dec 15.
Article En | MEDLINE | ID: mdl-25448047

Beta-lapachone (beta-Lp) derived from the Lapacho tree is a potentially novel anticancer agent currently under clinical trials. Previous studies suggested that redox activation of beta-Lp catalyzed by NAD(P)H: quinone oxidoreductase 1 (NQO1) accounted for its killing of cancer cells. However, the exact mechanisms of this effect remain largely unknown. Using chemiluminescence and electron paramagnetic resonance (EPR) spin-trapping techniques, this study for the first time demonstrated the real-time formation of ROS in the redox activation of beta-lapachone from cancer cells mediated by mitochondria and NQO1 in melanoma B16-F10 and hepatocellular carcinoma HepG2 cancer cells. ES936, a highly selective NQO1 inhibitor, and rotenone, a selective inhibitor of mitochondrial electron transport chain (METC) complex I were found to significantly block beta-Lp meditated redox activation in B16-F10 cells. In HepG2 cells ES936 inhibited beta-Lp-mediated oxygen radical formation by ~80% while rotenone exerted no significant effect. These results revealed the differential contribution of METC and NQO1 to beta-lapachone-induced ROS formation and cancer cell killing. In melanoma B16-F10 cells that do not express high NQO1 activity, both NOQ1 and METC play a critical role in beta-Lp redox activation. In contrast, in hepatocellular carcinoma HepG2 cells expressing extremely high NQO1 activity, redox activation of beta-Lp is primarily mediated by NQO1 (METC plays a minor role). These findings will contribute to our understanding of how cancer cells are selectively killed by beta-lapachone and increase our ability to devise strategies to enhance the anticancer efficacy of this potentially novel drug while minimizing its possible adverse effects on normal cells.


Antineoplastic Agents, Phytogenic/metabolism , Electron Transport Complex I/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/metabolism , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Prodrugs/metabolism , Activation, Metabolic/drug effects , Animals , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Humans , Indolequinones/pharmacology , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Naphthoquinones/antagonists & inhibitors , Naphthoquinones/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/metabolism , Oxidation-Reduction/drug effects , Prodrugs/chemistry , Prodrugs/pharmacology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Rotenone/pharmacology
8.
Toxicol In Vitro ; 27(7): 2094-104, 2013 Oct.
Article En | MEDLINE | ID: mdl-23933437

Novel ß-lapachone analogs 2-phenyl-3,4-dihydro-2H-benzo[h]chromene-5,6-dione (NQ1), 2-p-tolyl-3,4-dihydro-2H-benzo[h]chromene-5,6-dione (NQ3) and 2-methyl-2-phenyl-3,4-dihydro-2H-benzo[h]chromene-5,6-dione (NQ7), which have trypanocidal activity, were assayed for cytotoxic effects on murine EL-4 T lymphoma cells. The NQs inhibited the proliferation of EL-4 cells at concentrations above 1µM. Nuclear staining of the EL-4 cells revealed chromatin condensation and a nuclear morphology compatible with the induction of apoptosis. Flow cytometry assays with annexin V-FITC and propidium iodide confirmed the cell death by apoptosis. Using electron paramagnetic resonance (EPR), a semiquinone radical was detected in EL-4 cells treated with NQs. In addition, a decrease in the GSH level in parallel with reactive oxygen species (ROS) production was observed. Preincubation with n-acetyl-l-cysteine (NAC) was able to reverse the inhibitory effects of the NQs on cell proliferation, indicating that ROS generation is involved in NQ-induced apoptosis. In addition, the NQs induced a decrease in the mitochondrial membrane potential and increased the proteolytic activation of caspases 9 and 3 and the cleavage of Poly (ADP-Ribose) Polymerase (PARP). In conclusion, these results indicate that redox cycling is induced by the NQs in the EL-4 cell line, with the generation of ROS and other free radicals that could inhibit cellular proliferation as a result of the induction of the intrinsic apoptosis pathway.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Lymphoma, T-Cell/drug therapy , Naphthoquinones/pharmacology , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects , Acetylcysteine/pharmacology , Animals , Antineoplastic Agents/antagonists & inhibitors , Benzopyrans/antagonists & inhibitors , Benzopyrans/pharmacology , Benzoquinones/metabolism , Cell Line, Tumor , Cell Nucleus Shape/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromatin Assembly and Disassembly/drug effects , Free Radical Scavengers/pharmacology , Glutathione/antagonists & inhibitors , Glutathione/metabolism , Kinetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Naphthoquinones/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Trypanocidal Agents/antagonists & inhibitors , Trypanocidal Agents/pharmacology
9.
Pharm Biol ; 51(8): 948-54, 2013 Aug.
Article En | MEDLINE | ID: mdl-23607906

CONTEXT: Mansonone G and mansorin A are major bioactive constituents from Mansonia gagei Drumm (Sterculiaceae) wood, and their mild anti-estrogenic activity was reported previously by the authors. OBJECTIVE: In order to increase the potency of their anti-estrogenic effect and to clarify their binding way to estrogen receptor on a molecular level, several derivatives of both compounds will be prepared and a docking study of the original compounds and their derivatives on estrogen receptor alpha (ERα) was carried out. MATERIALS AND METHODS: The original compounds were isolated from the heartwood of M. gagei. Nine alkyl derivatives were prepared by acetylation, methylation, or adding a basic side chain to the free hydroxyl group of both compounds. The estrogenic/anti-estrogenic activities of the derivatives compared to the original compounds were carried out using ERα competitive binding screen and yeast two-hybrid assay expressing ERα and ERß using concentrations ranging from 10 to 100 µM. RESULTS: Acetyl mansonone G showed a 10-fold increase in its binding ability to ERα compared to mansonone G with an IC50 630 µM. Similarly, methyl mansonone G and acetyl mansonone G showed 50% and 35% inhibition of 17ß-estradiol-induced ß-galactosidase activity at 10 µM in the yeast expressing ERα, and 42% and 30%, respectively, at 10 µM in the yeast expressing ERß. Virtual docking of acetyl mansonone G to ERα showed that it binds, with its acetyl oxygen, in a similar way to the 17ß-OH of estradiol. DISCUSSION AND CONCLUSION: The phenolic hydroxyl group in mansonones and mansorins was not essential for binding to estrogen receptors. In addition, acetyl mansonone G could represent a promising starting material for the synthesis of anti-estrogenic agents.


Coumarins/pharmacology , Estrogen Receptor alpha/drug effects , Malvaceae/chemistry , Naphthoquinones/pharmacology , Coumarins/antagonists & inhibitors , Coumarins/isolation & purification , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estrogen Antagonists/administration & dosage , Estrogen Antagonists/isolation & purification , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Inhibitory Concentration 50 , Molecular Docking Simulation , Naphthoquinones/antagonists & inhibitors , Naphthoquinones/isolation & purification , Two-Hybrid System Techniques , beta-Galactosidase/drug effects , beta-Galactosidase/metabolism
10.
Cell Biochem Funct ; 28(8): 695-705, 2010 Dec 02.
Article En | MEDLINE | ID: mdl-21104938

Furano-1,2-naphthoquinone (FNQ), prepared from 2-hydroxy-1,4-naphthoquinone and chloroacetaldehyde in an efficient one-pot reaction, exhibits an anti-carcinogenic effect. FNQ exerted anti-proliferative activity with the G(2)/M cell cycle arrest and apoptosis in A549 cells. FNQ-induced G(2)/M arrest was correlated with a marked decrease in the expression levels of cyclin A and cyclin B, and their activating partner cyclin-dependent kinases (Cdk) 1 and 2 with concomitant induction of p53, p21, and p27. FNQ-induced apoptosis was accompanied with Bax up-regulation and the down-regulation of Bcl-2, X-linked inhibitor of apoptosis (XIAP), and survivin, resulting in cytochrome c release and sequential activation of caspase-9 and caspase-3. Western blot analysis revealed that FNQ suppressed EGFR phosphorylation and JAK2, STAT3, and STAT5 activation, but increased in activation of p38 MAPK and c-Jun NH2-terminal kinase (JNK) stress signal. The combined treatment of FNQ with AG1478 (a specific EGFR inhibitor) significantly enhanced the G(2)/M arrest and apoptosis, and also led to up-regulation in Bax, p53, p21, p27, release of mitochondrial cytochrome c, and down-regulation of Bcl-2, XIAP, survivin, cyclin A, cyclin B, Cdk1, and Cdk2 in A549 cells. These findings suggest that FNQ-mediated cytotoxicity of A549 cell related with the G(2)/M cell cycle arrest and apoptosis via inactivation of EGFR-mediated signaling pathway.


Apoptosis/drug effects , Cell Division/drug effects , ErbB Receptors/physiology , Furans/pharmacology , G2 Phase/drug effects , Naphthoquinones/pharmacology , Cell Proliferation , Enzyme Activation/drug effects , Furans/antagonists & inhibitors , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Janus Kinase 2/metabolism , Lung Neoplasms , Mitochondria/drug effects , Mitochondria/physiology , Mitogen-Activated Protein Kinases/metabolism , Naphthoquinones/antagonists & inhibitors , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/biosynthesis , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured
11.
Eur J Pharmacol ; 638(1-3): 21-8, 2010 Jul 25.
Article En | MEDLINE | ID: mdl-20420821

Plumbagin, derived from the plant Plumbago zeylanica, has been shown to chronically activate ERK1/2 and inhibit Akt activity in cancer cells. However, the acute effects of plumbagin on ERK1/2 and Akt activities remain unknown. In this study, we examined the effects of plumbagin on ERK1/2 and Akt activities in 3T3-L1 cells. Exposure of 3T3-L1 cells to plumbagin generated superoxide and activated both ERK1/2 and Akt. The plumbagin-stimulated ERK1/2 and Akt activities were sensitive to an antioxidant NAC, superoxide dismutase mimetic MnTBAP, superoxide scavenger Tiron and NAD(P)H oxidase inhibitor DPI. Plumbagin-stimulated ERK1/2 activity was attenuated by the MEK1/2 inhibitor PD98059 and Ras inhibitor manumycin A, whereas plumbagin-stimulated Akt activity was blocked by the PI3K inhibitor LY294002. Both plumbagin-stimulated ERK1/2 and Akt activities were attenuated by PP2, a Src inhibitor. Interestingly, inhibition of phosphatidylinositol 3-kinase (PI3-kinase), but not Akt, activity leaded to attenuation of plumbagin-stimulated ERK1/2 activity. These results suggest that plumbagin activates NAD(P)H oxidase, Src, and PI3K, and that the activated PI3K or PDK1 subsequently stimulate Akt and Ras-Raf-MEK1/2-ERK1/2 in 3T3-L1 cells.


Mitogen-Activated Protein Kinase 1/biosynthesis , Naphthoquinones/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/biosynthesis , Superoxides/metabolism , src-Family Kinases/antagonists & inhibitors , 3T3-L1 Cells , Animals , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Chromones/pharmacology , Drug Interactions , Enzyme Activation , Flavonoids/pharmacology , Free Radical Scavengers/pharmacology , Mice , Morpholines/pharmacology , Naphthoquinones/antagonists & inhibitors , Polyenes/pharmacology , Polyunsaturated Alkamides/pharmacology , Pyrimidines/pharmacology
12.
J Physiol Pharmacol ; 60 Suppl 4: 111-6, 2009 Oct.
Article En | MEDLINE | ID: mdl-20083859

Resveratrol, an important antioxidant found in grapes and wine, is likely to contribute to red wine's potential to prevent human cardiovascular disease. In addition to its known (direct) antioxidant effect, we have found that resveratrol also regulates the gene expression of pro-oxidative and anti-oxidative enzymes in human endothelial cells. NADPH oxidases (Nox) are the predominant producers of superoxide in the vasculature, whereas superoxide dismutase (SOD) and glutathione peroxidase 1 (GPx1) are the major enzymes responsible for the inactivation of superoxide and hydrogen peroxide, respectively. Incubation of human umbilical vein endothelial cells (HUVEC) and HUVEC-derived EA.hy 926 cells with resveratrol resulted in a concentration- and time-dependent downregulation of Nox4, the most abundant NADPH oxidase catalytic subunit (quantitative real-time RT-PCR). The same resveratrol regimen upregulated the mRNA expression of SOD1 and GPx1. The addition the protein levels of SOD1 and GPx1 were enhanced by resveratrol in a concentration-dependent manner (Western blot analyses). Pretreatment of EA.hy 926 cells with resveratrol completely abolished DMNQ-induced oxidative stress. Thus, the expressional suppression of pro-oxidative genes (such as NADPH oxidase) and induction of anti-oxidative enzymes (such as SOD1 and GPx1) might be an important component of the vascular protective effect of resveratrol.


Antioxidants/pharmacology , Endothelium, Vascular/physiology , Glutathione Peroxidase/biosynthesis , NADPH Oxidases/biosynthesis , Oxidative Stress/drug effects , Stilbenes/pharmacology , Superoxide Dismutase/biosynthesis , Blotting, Western , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelium, Vascular/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glutathione Peroxidase/genetics , Humans , NADPH Oxidase 4 , NADPH Oxidases/genetics , Naphthoquinones/antagonists & inhibitors , Naphthoquinones/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reactive Oxygen Species , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Glutathione Peroxidase GPX1
13.
Int J Radiat Oncol Biol Phys ; 61(1): 212-9, 2005 Jan 01.
Article En | MEDLINE | ID: mdl-15629614

PURPOSE: To reveal the interaction between beta-lapachone (beta-lap) and ionizing radiation (IR) in causing clonogenic death in cancer cells and to elucidate the potential usefulness of beta-lap treatment in combination with radiotherapy of cancer. METHODS AND MATERIALS: FSaII tumor cells of C3H mice were used. The cytotoxicity of beta-lap alone or in combination with IR in vitro was determined using clonogenic survival assay method. The IR-induced changes in the expression and the enzymatic activity of NAD(P)H:quinone oxidoreductase (NQO1), a mediator of beta-lap cytotoxicity, were elucidated and the relationship between the NQO1 level and the sensitivity of cells to beta-lap was investigated. The combined effect of IR and beta-lap to suppress tumor growth was studied using FSaII tumors grown subcutaneously in the thigh of C3H mice. RESULTS: beta-Lap caused clonogenic death of FSaII tumor cells in vitro in a dose- and time-dependent manner. When cells were treated first with beta-lap and then exposed to IR in vitro, the resultant cell death was only additive. On the contrary, exposing cells to IR at 2.5 Gy first and then treating the cells with beta-lap killed the cells in a synergistic manner. Importantly, the 2.5 Gy cells were sensitive to beta-lap as long as 10 h after irradiation, which was long after the sublethal radiation damage was repaired. Irradiation of FSaII cells in vitro with 2.5 Gy significantly increased the expression and enzymatic activity of NQO1. The growth delay of FSaII tumors caused by an intraperitoneal injection of beta-lap in combination with 20 Gy irradiation of tumor was significantly greater than that caused by beta-lap or 20 Gy irradiation alone. CONCLUSION: The sensitivity of cells to beta-lap is dependent on NQO1 activity. IR caused a long-lasting increase in NQO1 activity in cancer cells, thereby sensitizing cells to beta-lap and treatment of experimental mouse tumors with IR and beta-lap suppressed tumor growth in a synergistic manner. The combination of beta-lap and radiotherapy is a potentially effective regimen for the treatment of human cancer.


NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/pharmacology , Neoplasm Proteins/metabolism , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Dicumarol/pharmacology , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , Mice , Mice, Inbred C3H , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/radiation effects , Naphthoquinones/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/radiation effects , Radiation, Ionizing
14.
FEBS Lett ; 555(2): 217-22, 2003 Dec 04.
Article En | MEDLINE | ID: mdl-14644418

Vitamin K-related analogs induce growth inhibition via a cell cycle arrest through cdc25A phosphatase inhibition in various cancer cell lines. We report that 2,3-dichloro-5,8-dihydroxy-1,4-naphthoquinone (DDN), a naphthoquinone analog, induces mitochondria-dependent apoptosis in human promyelocytic leukemia HL-60 cells. DDN induced cytochrome c release, Bax translocation, cleavage of Bid and Bad, and activation of caspase-3, -8, -9 upon the induction of apoptosis. Cleavage of Bid, the caspase-8 substrate, was inhibited by the broad caspase inhibitor z-Val-Ala-Asp(OMe)-fluoromethylketone (zVAD-fmk), whereas cytochrome c release was not affected, suggesting that activation of caspase-8 and subsequent Bid cleavage occur downstream of cytochrome c release. DDN inhibited the activation of Akt detected by decreasing level of phosphorylation. Overexpression of constitutively active Akt protected cells from DDN-induced apoptosis, while dominant negative Akt moderately enhanced cell death. Furthermore, Akt prevented release of cytochrome c and cleavage of Bad in DDN-treated HL-60 cells. Taken together, DDN-induced apoptosis is associated with mitochondrial signaling which involves cytochrome c release via a mechanism inhibited by Akt.


Apoptosis/physiology , Enzyme Inhibitors/pharmacology , Mitochondria/metabolism , Naphthoquinones/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , cdc25 Phosphatases/antagonists & inhibitors , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/metabolism , Caspase Inhibitors , Caspases/metabolism , Cytochromes c/metabolism , Enzyme Activation/drug effects , HL-60 Cells , Humans , Immunoblotting , Naphthoquinones/antagonists & inhibitors , Protein Serine-Threonine Kinases/pharmacology , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , bcl-2-Associated X Protein , bcl-Associated Death Protein
15.
Comp Biochem Physiol C Toxicol Pharmacol ; 135C(3): 337-43, 2003 Jul.
Article En | MEDLINE | ID: mdl-12927908

The aim of this study was to evaluate effects of modified glucomannans (Mycosorb) on egg yolk and liver of the day-old quail after aurofusarin inclusion in the maternal diet. Fifty-four 45-day-old Japanese quail were divided into three groups and were fed ad libitum a corn-soya diet balanced in all nutrients. The diet of the experimental quail was supplemented with aurofusarin at the level of 26.4 mg/kg feed in the form of Fusarium graminearum culture enriched with aurofusarin or with aurofusarin plus Mycosorb at 1 g/kg feed. Eggs obtained after 8 weeks of feeding were analysed and incubated in standard conditions of 37.5 degrees C/55% RH. Samples of quail liver were collected from day-old hatchlings. Main polyunsaturated fatty acids (PUFAs) of the egg yolk were analysed by gas chromatography, and tocopherols and tocotrienols were analysed by HPLC-based methods. Inclusion of aurofusarin in the maternal diet was associated with decreased proportions of docosahexaenoic acid and increased proportions of linoleic acid in major lipid fractions of the egg yolk as well as with decreased concentrations of alpha- and gamma-tocopherols, alpha- and gamma-tocotrienols in egg yolk and liver of a day-old quail. Inclusion of modified glucomannans (Mycosorb) into the quail diet simultaneously with aurofusarin showed a significant protective effect against changes in PUFA and antioxidant composition in the egg yolk and liver of quail. It is suggested that a combination of mycotoxin adsorbents and natural antioxidants could be the next step in counteracting mycotoxins in animal feed.


Coturnix/embryology , Mannans/pharmacology , Naphthoquinones/toxicity , Ovum/drug effects , Animals , Coturnix/metabolism , Egg Yolk/drug effects , Egg Yolk/metabolism , Fatty Acids, Nonesterified/metabolism , Female , Liver/drug effects , Liver/metabolism , Male , Naphthoquinones/antagonists & inhibitors , Ovum/metabolism , Vitamin E/metabolism
16.
Cancer Res ; 63(15): 4527-32, 2003 Aug 01.
Article En | MEDLINE | ID: mdl-12907627

Expression of mdr-1 is complex and highly regulated. Several lines of evidence indirectly suggest that transcription factor c-Jun may negatively regulate human mdr-1 gene expression. We recently found that salvicine, a novel topoisomerase II inhibitor, is cytotoxic for multidrug resistance (MDR) tumor cells and down-regulates mdr-1 expression in MDR K562/A02 cells. Salvicine also stimulates a significant increase in the level of c-jun mRNA in HL60 cells. This study investigated the relationship between c-Jun activation and down-regulation of mdr-1 expression by salvicine in K562/A02 cells. Reverse-transcription PCR and Western blotting analyses revealed that salvicine suppressed mdr-1 expression in MDR cells and promoted c-jun expression in both MDR and parental K562 cells. Moreover, levels of c-jun expression were enhanced by salvicine before reduction of mdr-1 expression in K562/A02 cells. Furthermore, c-jun antisense oligodeoxynucleotides prevented salvicine-stimulated enhancement of c-Jun protein and reduction of mdr-1 gene expression, but did not affect the increase in c-jun mRNA levels. Salvicine promoted phosphorylation of c-Jun-N-terminal kinase and c-Jun protein in MDR K562/A02 and parental K562 cells. Electrophoretic mobility shift assay analysis showed that salvicine enhanced DNA binding activity of transcription factor activator protein 1. Additionally, c-jun antisense oligodeoxynucleotides also inhibited salvicine-induced apoptosis and cytotoxicity in MDR and parental K562 cells. A possible pathway emerges from these results: salvicine stimulates c-Jun-N-terminal kinase phosphorylation and activation, resulting in c-Jun phosphorylation and activation. Activated c-Jun promotes expression of c-jun itself, represses mdr-1 transcription, and triggers pro-apoptotic signals, resulting in low mdr-1 expression and cell death. The present results demonstrate that transcription factor c-Jun plays a principal role in down-regulation of mdr-1 expression and induction of apoptosis in salvicine-treated human MDR K562/A02 cells, providing new insights into the complicated mechanisms regulating mdr-1 expression. The findings also suggest that c-Jun might be a potential drug target for circumventing tumor MDR.


Gene Expression Regulation, Leukemic/physiology , Genes, MDR/genetics , Proto-Oncogene Proteins c-jun/genetics , Apoptosis/drug effects , DNA, Neoplasm/metabolism , Down-Regulation/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , JNK Mitogen-Activated Protein Kinases , K562 Cells , Mitogen-Activated Protein Kinases/metabolism , Naphthoquinones/antagonists & inhibitors , Naphthoquinones/pharmacology , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcription Factor AP-1/metabolism , Transcriptional Activation/drug effects
17.
Toxicol Appl Pharmacol ; 182(2): 116-25, 2002 Jul 15.
Article En | MEDLINE | ID: mdl-12140175

beta-Lapachone, a novel anticancer drug, induces various human carcinoma cells to undergo apoptotic cell death. However, we report here that, in human osteocarcinoma (U2-OS) cells, beta-lapachone induces necrosis rather than apoptosis. beta-Lapachone-induced necrotic cell death in U2-OS cells was characterized by propidium iodide uptake, cytochrome c release, a decreased mitochondrial membrane potential, and ATP depletion. The mitochondrial potential transition (MPT), including the reduction of the mitochondrial transmembrane potential and the release of mitochondrial cytochrome c, occurred in beta-lapachone-treated cells; cotreatment of these cells with cyclosporin A, an inhibitor of MPT pore, failed to prevent necrotic cell death. This indicates that the MPT transition does not play a crucial role in this process. Furthermore, beta-lapachone-induced necrosis was independent of oxidative stress and caspase activation. However, excessive poly(ADP-ribose) polymerase (PARP) activation and subsequent depletion of intracellular NAD(+) and ATP were seen in beta-lapachone-treated U2-OS cells. Cotreatment with a PARP inhibitor, 3-aminobenzamide, decreased beta-lapachone-induced PARP activation and provided significant protection from necrosis by preventing depletion of intracellular NAD(+) and ATP. Taken together, our results suggest that PARP plays an important role in the signaling pathway for beta-lapachone-induced necrosis in U2-OS cells.


Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Naphthoquinones/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors , Adenosine Triphosphate/metabolism , Antineoplastic Agents, Phytogenic/toxicity , Apoptosis/drug effects , Blotting, Western , Bone Neoplasms/enzymology , Cell Cycle/drug effects , Cytochrome c Group/metabolism , DNA Damage/drug effects , DNA Fragmentation , Enzyme Activation/drug effects , Flow Cytometry , Genes, p53/genetics , Humans , In Situ Nick-End Labeling , Membrane Potentials/drug effects , NAD/metabolism , Naphthoquinones/toxicity , Osteosarcoma/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Cells, Cultured
18.
Free Radic Biol Med ; 19(5): 565-74, 1995 Nov.
Article En | MEDLINE | ID: mdl-8529915

Cultured primary hepatocytes pretreated (protected) with the iron chelator deferoxamine or the antioxidant N,N'-diphenyl-p-phenylenediamine (DPPD) were resistant to the toxicity of 5 microM naphthazarin (5,8-dihydroxy-1,4-naphthoquinone) during a 180-min exposure. Hepatocytes exposed to naphthazarin without any protection were abruptly depleted of intracellular reduced glutathione, and the level of cytosolic Ca2+ was rapidly increased. This was followed by lipid peroxidation, measured as accumulation of malondialdehyde (MDA) and 4-hydroxyalkenals (4-HNA) intra- and extracellularly; decrease in ATP levels; destabilization of lysosomes; and finally cell death. The stability of the lysosomal membranes was evaluated by determining retention of the lysosomotropic weak base acridine orange (AO). Naphthazarin exposure caused leakage of protons from the acidic compartment, as indicated by relocalization of AO to the cytosol. Protection of the cell cultures with deferoxamine or DPPD prevented destabilization of lysosomes and cell killing. It also reduced the loss of ATP but did not prevent the depletion of glutathione or the increase in Ca2+. In cells subjected to naphthazarin exposure, DPPD protection also completely inhibited lipid peroxidation, whereas deferoxamine pretreatment only slightly reduced the intracellular accumulation of MDA and 4-HNA but completely prevented cell rupture and the leakage of these lipid peroxidation products to the medium that took place in large amounts from unprotected cells exposed to naphthazarin. Deferoxamine is taken up by endocytosis and is thus transported to the acidic vacuolar apparatus, whereas the lipophilic DPPD is rapidly distributed throughout the cells. Inhibiting endocytosis during deferoxamine pretreatment, by incubating at +4 degrees C or by preexposure to a mixture of the endocytosis-inhibitors cytochalasin B and monensin, abolished the protective effect of deferoxamine. The findings suggest that naphthazarin-induced cell killing is not caused directly by either thiol oxidation or an increase in cytosolic free Ca2+, but rather is preceded by lysosomal destabilization, which may be prevented either by inhibition of cellular peroxidation in general or by prevention of iron-catalyzed oxidative reactions, and involves peroxidation of cellular membranes, energy depletion, and leakage of lysosomal content. DPPD would protect against cell killing by preventing lipid peroxidation of cellular membranes in general, whereas deferoxamine seems to allow a limited general cellular peroxidation but specifically prevents peroxidation and fragmentation of lysosomal membranes by chelating intralysosomal iron and, consequently, leakage of destructive lysosomal contents with ensuing cell rupture and death. Thus, a certain degree of cellular peroxidation does not appear to be lethal as long as lysosomal membranes are protected, placing lysosomes into a category of cellular loci minora resistentia.


Antineoplastic Agents/toxicity , Liver/pathology , Lysosomes/pathology , Naphthoquinones/toxicity , Oxidative Stress , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/antagonists & inhibitors , Antioxidants/pharmacology , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Cytochalasin B/pharmacology , Cytosol/metabolism , Deferoxamine/pharmacology , Endocytosis/drug effects , Glutathione/metabolism , Iron Chelating Agents/pharmacology , Kinetics , Liver/drug effects , Liver/physiology , Lysosomes/drug effects , Lysosomes/physiology , Male , Monensin/pharmacology , Naphthoquinones/antagonists & inhibitors , Phenylenediamines/pharmacology , Rats , Rats, Wistar , Time Factors
19.
Biochem Pharmacol ; 46(4): 589-95, 1993 Aug 17.
Article En | MEDLINE | ID: mdl-8363631

Exposure of rat hepatocytes to hydrazine, carbon tetrachloride or 1,4-naphthoquinone results in cytotoxicity determined as uptake of Trypan blue and leakage of lactate dehydrogenase (LDH). After exposure of hepatocytes to hydrazine and 1,4-naphthoquinone, ATP was also measured and was found to be depleted. Addition of the beta-amino acid taurine to the hepatocyte incubation buffer partially protects the cells against the cytotoxicity of these three different cytotoxic compounds, as indicated by Trypan blue uptake and LDH leakage. Taurine also reduces the depletion of ATP caused by 1,4-naphthoquinone but not hydrazine. It is suggested that taurine may have a cytoprotective effect in vitro and may be a useful tool for the investigation of mechanisms of cytotoxicity.


Carbon Tetrachloride/antagonists & inhibitors , Hydrazines/antagonists & inhibitors , Liver/drug effects , Naphthoquinones/antagonists & inhibitors , Taurine/pharmacology , Animals , Carbon Tetrachloride/toxicity , Cell Survival/drug effects , Cells, Cultured , Hydrazines/toxicity , L-Lactate Dehydrogenase/metabolism , Liver/cytology , Liver/enzymology , Male , Naphthoquinones/toxicity , Rats , Rats, Sprague-Dawley , Trypan Blue/metabolism
20.
Biochem Pharmacol ; 44(2): 215-21, 1992 Jul 22.
Article En | MEDLINE | ID: mdl-1642637

In rat hepatocytes exposed to the quinones menadione and 2,3-dimethoxy-1,4-naphthoquinone (2,3-diOMe-1,4-NQ) a decrease in NAD+ is observed. DNA damage and activation of poly(ADP-ribose)polymerase are often associated with a decrease in NAD+. Using rat hepatocytes and human myeloid leukaemic cells (K562), we examined the extent of DNA damage induced by these quinones at non-toxic concentrations, i.e. at concentrations at which the cells completely exclude the dye trypan blue. Both quinones caused significant DNA damage at very low concentrations (5-100 microM). With 2,3-diOME-1,4-NQ (15 microM) or menadione (15 microM) single strand breaks (SSB) were observed at very early time points (less than 5 min), reaching a maximum between 20 and 30 min. Most SSB were repaired within 45 min of the removal of the quinones. Whilst extensive repair was observed within 4 hr of the removal of 2,3-diOMe-1,4-NQ (15 microM), only partial repair was observed following exposure to menadione (15 microM). SSB induced by 2,3-diOMe-1,4-NQ (15 microM) were completely inhibited by the iron chelator 1,10-phenanthroline (25 microM), whereas in cells exposed to menadione (15 microM) they were only partially inhibited. Finally, although the membrane integrity of K562 cells was unaffected by exposure to high concentrations of both quinones (less than or equal to 400 microM), cytostasis was observed at much lower concentrations (50 microM). Our results demonstrate that at very low concentrations these quinones induce extensive DNA damage possibly caused by hydroxyl radicals. The DNA damage was accompanied by an early cytostasis but no loss of membrane integrity.


DNA, Single-Stranded/drug effects , Liver/drug effects , Naphthoquinones/pharmacology , Tumor Cells, Cultured/drug effects , Vitamin K/physiology , Animals , Cell Division/drug effects , DNA Damage , Dose-Response Relationship, Drug , Humans , Liver/metabolism , NAD/metabolism , Naphthoquinones/antagonists & inhibitors , Phenanthrolines/pharmacology , Rats , Trypan Blue , Tumor Cells, Cultured/metabolism , Vitamin K/antagonists & inhibitors
...